Regenerative Potential (RP) of MSCEVs: Comparison
Please note this is a comparison between Version 2 by Catherine Yang and Version 1 by Rishabh Malviya.

Mesenchymal stem cell extracellular vesicles (MSCEVs) obtained from MSCs can have numerous therapeutic applications via regeneration of various body tissues. MSCEV action can be potentiated by modifying the mesenchymal stem cells culturing methodology and bioengineering extracellular vesicles (EVs). 

  • extracellular vesicles
  • regenerative potential
  • therapeutics

1. MSCEVs and Cardiac Tissue Regeneration

Endogenous repair of damaged myocardium is usually slow and depends on the inadequate division of pre-existing cardiomyocytes and recruitment and differentiation of local cardiac stem cells [29,30][1][2]. Exogenous MSCEVs address the inadequate response to myocardial injury. Preliminary investigation recognized the ability of human embryonic-derived MSCEVs (EMSCEVs) to reduce the size of infarct in a myocardial ischemia/reperfusion injury (MI) mouse model [31][3] by activating the PI3K/Akt signaling pathway, which increases myocardial viability and inhibits adverse remodeling [32][4]. Based on this, human Akt-transfected umbilical cord-derived MSCEVs (UCMSCEVs) were developed, which exhibited enhanced endothelial cell proliferation, migration, and blood vessel formation in vivo and tube-like structures in vitro, compared to nonmodified human UCMSCEVs [33][5].
A study revealed that human amniotic fluid-derived mesenchymal stem cells (AFMSCs) and murine-induced pluripotent mesenchymal stem cells (iPMSCs) triggered cardiac regeneration via paracrine modulation of endogenous mechanisms and improved cardiac repair in an MI murine model. Administration of iPMSCEVs was shown to be safer compared to iPMSCs [34,35][6][7]. Another MI rat model study revealed that combination treatment of rat bone marrow mesenchymal stem cells (BMMSCs) and derived BMMSCEVs improved cardiac functioning, reduced infarct size, and increased neo-vascularization compared to individual treatment with BMMSCs or BMMSCEVs [36][8].
Human BMMSCs preconditioned with hypoxia improved in vitro cell biological activity [37][9]. The report suggests that the efficacy of BMMSCs from Cynomolgous monkeys improves when implanted for treatment of MI in monkeys [38][10]. Hypoxia enhanced the therapeutic effectiveness of secretory EVs, such that hypoxia conditioned human BMMSCEVs with 1% O2 for 3 days exhibit more cardiac regeneration in the rat MI model compared to BMMSCEVs in normoxic conditions. The report suggests that the underlining mechanism is increased angiogenesis at the infract site [39][11]. Hypoxia reconditioned with murine and rat BMMSCEVs (at 1% O2 for 72 h or 0.5% O2 for 24 h) inhibited apoptosis of cardiomyocytes by enriching miR-125b-5p-EV and miR-210-EV. In this case, the underline mechanism is the suppression of proapoptotic p53 and BCL2-antagonist/killer 1 (BAK1) genes and higher recruitment of cardiac progenitor cells in the infarcted heart [40,41][12][13]. MSCEVs, when encapsulated as a hydrogel, offer targeted and controlled delivery to cardiac defects. For example, a sustained release profile and higher cardiac regeneration are provided by human UCMSCEVs loaded in peptide hydrogel. The EV/hydrogel complex improves myocardial functioning via reduction of apoptosis, inflammation, and fibrosis and enhancement of angiogenesis in the infarcted boundary zone of rat hearts [42][14].

2. MSCEVs and Nervous Tissue Regeneration

A small injury or disease in the nervous system may lead to serious or lethal effects. The complex physiological system and limited healing capacity present the biggest challenge to nerve repair in regenerative medicine [43][15]. Various studies reported the repair of injured peripheral nerves (PNs) by different MSCEVs. For example, rat bone marrow MSCEVs (BMMSCEVs) regenerated injured sciatic PNs using a rat model. BMMSCEVs exhibited higher growth-associated protein (GAP43) expression, improved histomorphometric repair, and enhanced the sciatic functioning index [44][16]. Similarly, human umbilical cord MSCEVs (UCMSCEVs) were reported to regenerate PNs at the sites of sciatic nerve defects (SNDs) in rats. Accumulation of UCMSCEVs at the site of injured PNs stimulates axons and generates Schwann cells to fence off individual axons, reduce muscle atrophy (denervated), and modulate inflammation by upregulating anti-inflammatory cytokines (e.g., interleukin (IL)-10) and downregulating pro-inflammatory cytokines (e.g., IL1β and IL6) [45][17]. Rat adipose-derived MSCEVs (AMSCEVs) are also known to promote PN regeneration and neurite growth in sciatic PN defects via Schwann cell modulation [46][18]. The AMSCEV perinuclear location and accumulation in vesicular-like structures in Schwann cells stimulate and proliferate damaged cells, thereby indicating an endocytosis facilitated internalization pathway [47][19]. PN regeneration by gingiva MSCEVs (GMSCEVs) indicates proliferation and migration of Schwann cells through activation of the c-Jun N-terminal kinase (JNK) pathway and upregulation of Notch1, c-Jun, glial fibrillary acidic protein (GFAP), and Sox2 genes for de-differentiation or repair of the Schwann cell phenotype [48][20].
MSCEVs also have therapeutic potential to repair the central nervous system (CNS). Reports suggest that BMMSCEVs cause neurite development (via miR-133b transfer into neural cells) in a middle cerebral artery (MCA) stroke rat model [49][21]. BMMSCEV administration raised the axon density and synaptophysin-positive area (along the ischemic zone of striatum and cortex) and increased the expression of von Willebrand factor (endothelial cell marker) and doublecortin (neuroblast marker) in MCA rats [50[22][23],51], which suggest their potential for neurite remodeling, angiogenesis, and neurogenesis in stroke treatment [52][24].
BMMSCEVs, when evaluated for their potential in traumatic brain injury (TBI) using a rat model, showed improvement in the recovery of brain functioning by enhancing the counts of new immature and mature neurons in the dentate gyrus and new endothelial cells in the lesion boundary and dentate gyrus [53][25]. MSCEVs have also shown potential in the recovery of spinal cord injury (SCI) characterized by disrupted microvascular stability and high blood–spinal cord barrier (BSCB) permeation (due to pericyte migration) [54,55][26][27]. Mouse BMMSCEV treatment inhibited pericyte migration, thus improving BSCB structural integrity and motor functioning in an SCI rat model [56][28].
Another mechanism for SCI recovery by BMMSCEVs suggests the inhibition of neuronal apoptosis via activation of the Wnt/β-catenin signaling pathway [57][29]. Modification of rat BMMSCEVs with miR-133b activated the ERK1/2 and STAT3 pathways in the SCI model, which enhanced axon regeneration, neuron preservation, and locomotor functioning, compared to BMMSCEVs that were not modified with miR-133b [58][30]. Human placental MSCEVs (PMSCEVs) exhibited regeneration of myelin through differentiation of oligodendrocyte precursor cells (endogenous) into myelinating oligodendrocytes in vitro and enhanced myelination in the spinal cord of treated mice (multiple sclerosis model), followed by improved motor functioning [59][31]. Human BMMSCEVs stimulated by interferon-gamma (IFN-γ) exhibited a reduction in demyelination and neuroinflammation, thereby improving motor skills, in a multiple sclerosis experimental autoimmune encephalomyelitis (EAE) mouse model [60][32]. Also, in an Alzheimer disease mouse model, MSCEVs enhanced neurogenesis and cognitive function recovery [61][33].
Evidence suggests that neonatal hypoxic ischemic (HI) encephalopathy is one of the major reasons for newborn disability and death. MSCEVs exert a regenerative effect on HI encephalopathy; for example, a study reported the neuroprotective effect of MSCEVs in a Rice–Vannucci mouse model of severe HI-induced neonatal brain insult [62][34]. Another study found protective effects of MSCEVs in a preclinical model of preterm HI brain injury. The study involved in utero intravenous administration of MSCEVs into ovine fetuses with induced global HI. The study revealed that MSCEV administration improved the functioning of the brain via a reduction of seizure duration and count and preservation of the reflex sensitivity of baroreceptors [63][35].

3. MSCEVs and Bone Regeneration

MSCEVs provide excellent benefits in skeletal regeneration. To date, various MSCEVs have been tested for their potential in regeneration of injured bone. Human iPMSCEVs and BMMSCEVs enhanced in vitro osteogenic differentiation of BMMSCs on the one hand, and in vivo angiogenesis and bone formation in a rat model with a critical-sized calvarial defect, on the other hand [64,65][36][37]. The report suggests that dimethyl-oxaloyl glycin human BMMSCEVs stimulate angiogenesis via the Akt/mTOR pathway [66][38]. The efficacy of ADMSCEVs for bone regeneration can be enhanced via pre-conditioning of ADMSCs with the cytokine tumor necrosis factor alpha (TNF-α). This is based on the proliferation and osteogenic differentiation of osteoblastic cells in an in vitro experiment [67][39]. The study revealed that human BMMSCEV administration in a CD9 mouse model with femoral shaft fracture (with impaired bone healing ability) improved fracture healing [68][40].
In dental regeneration, administration of human dental pulp MSCEVs (DPMSCEVs) causes in vitro odontogenic differentiation. This is based on DPMSCEV endocytosis, which stimulated the P38 MAPK pathway and regenerated dental pulp-like tissue in a tooth root slice model [69][41]. MSCEVs are known to stimulate the migration and proliferation of periodontal ligament cells via CD73-facilitated adenosine receptor activation of pro-survival Akt and ERK signaling and periodontal regeneration in a periodontal defect rat model [70][42]. For improved performance of scaffold and bone healing, MSCEVs are combined with tissue-engineered constructs. For example, human ADMSCEVs are immobilized with biotin-doped polypyrrole titanium and poly (lactic-co-glycolic acid) scaffolds. In vitro investigations of BMMSCEV-based and unmodified scaffolds revealed that BMMSCEV scaffolds led to high osteoinduction of BMMSCs and osteoblasts, whereas in vivo studies with a murine model of bone defect showed improved collagen and bone tissue formation [71][43]. Loading of human BMMSCEVs into the tricalcium phosphate scaffold enhanced bone healing of calvarial defects via activation of the PI3K/Akt signaling pathway [72][44]. The report suggests that rat BMMSCEVs encapsulated in decalcified bone matrix scaffold stimulates bone regeneration by promoting vascularization in grafts [73][45].

4. MSCEVs and Liver Tissue Regeneration

Human embryonic MSCEVs are known to promote hepatic regeneration in a carbon tetrachloride (CCl4)-induced liver injury mouse model by increasing the proliferation of hepatocytes and reducing their apoptosis [74][46]. A report suggests that human iPMSCEVs cause regeneration of hepatic cells in a hepatic ischemia–reperfusion injury rat model via inhibition of hepatocyte apoptosis, suppression of inflammatory response, and attenuation of the oxidative stress response [75][47]. Human iPMSCEVs are also known to induce in vitro and in vivo proliferation of hepatocytes (in a dose-dependent manner), which is related to the activation of the sphingosine kinase and sphingosine-1-phosphate signaling pathway (which promotes cell proliferation) [76,77][48][49]. The ability of UCMSCEVs to ameliorate neutrophil infiltration and inhibit oxidative stress in hepatic tissue supports their property of protecting against hepatic apoptosis [78][50]. The benefits of human embryonic MSCEVs can be further enhanced by encapsulating them into PEG hydrogel, intended to sustain systemic delivery against hepatic failure. MSCEV accumulation in the liver for a prolonged period of time in a chronic hepatic fibrosis rat model had superior antiapoptotic, antifibrotic, and regenerative properties compared to conventional MSCEV administration [79][51].

5. MSCEVs and Kidney Regeneration

Many studies have reported the regenerative potential of MSCEVs in chronic kidney damage (CKD) and acute kidney injury (AKI). An earlier study reported that human BMMSCEVs stimulated cell proliferation, hastened the recovery of injured tubular cells, supported the functional recovery of glycerol-induced AKI, and prevented apoptosis [80][52]. MSCEVs exhibit their therapeutic actions using several mechanisms. One mechanism includes transportation of genetic material (mRNAs and miRNAs) to renal cells (injured), which exerts antiapoptotic, proangiogenetic, anti-inflammatory, and antifibrotic effects on AKI [81][53]. The second mechanism involves the parallel transfer of human IGF-1 receptor mRNA (present in MSCEVs) into tubular cells [82][54]. Administration of mouse and human BMMSCEVs in rat and mouse AKI models caused protection from AKI and enhanced renal functioning by stimulating tubular epithelial cell proliferation and inhibiting apoptosis [83][55]. The study showed that human BMMSCEVs ameliorated kidney morphology and functioning in a cisplatin-induced AKI mouse model. Based on this, an in vitro study of cisplatin-treated human tubular epithelial cells showed that BMMSCEVs upregulated antiapoptotic genes (β-cell lymphoma 2, β-cell lymphoma, extra-large and baculoviral IAP repeat-containing 8) and downregulates the genes that participate in the execution phase of cell apoptosis (caspase-1,8 and α-lymphotoxin) [84][56].
For controlled and targeted release at the site of AKI (after ischemia–reperfusion) in a mouse model, loading mouse BMMSCEVs onto self-assembling peptide nanofiber hydrogel provides a substantial increase in efficacy (improved renal function) [85][57]. Human UCMSCEVs are reported to induce in vitro and in vivo kidney repair in a cisplatin-induced AKI rat model by de-differentiation of tubular cells, promotion of cell proliferation, and reduction in cell apoptosis and oxidative stress [86][58]. Renal regeneration is also exhibited by human Wharton’s jelly MSCEVs when administered in an AKI rat model. These MSCEVs improvise renal functioning by augmenting tubular cell proliferation and reducing apoptosis and inflammation through mitochondrial fission [87][59]. Human glomerular and liver MSCEVs are also reported to stimulate recovery after AKI [88][60]. A study reported the MSCEV effect in diabetes-associated chronic kidney damage (CKD) [89][61].
Human urinary MSCEVs (UMSCEVs) are known to prevent the progression of CKD through the promotion of vascular regeneration, inhibition of podocyte apoptosis, and cell survival in a streptozotocin-induced diabetic nephropathy rat model [90][62]. The administration of UMSCEVs in diabetic mice showed improved renal morphology and anti-apoptotic performance of tubular epithelial cells [91][63]. A report suggested that human BMMSCEVs and liver MSCEVs prevent fibrosis and its progression in a diabetic nephropathy mouse model mediated by miRNA (profibrotic gene downregulator) [92][64]. Similar action was reported with the administration of human liver MSCEVs in an aristolochic acid-induced CKD model [93][65]. The Murine BMMSCEVs also exhibit protective action after renal injury in vitro and in vivo [94][66]. The administration of human BMMSCEVs has the potential to repair damaged mitochondria renal proximal tubule apical/basolateral membranes and improve renal functioning in a medaka model of cadmium exposure that resembles CKD on long-term exposure to heavy metals [95][67]. A clinical trial of 40 patients with stage III and IV CKD (n = 20 administered MSCEVs, n = 20 administered placebo) reported that UCMSCEVs provided safety and ameliorated CKD progression [96][68].

6. MSCEVs and Muscle Regeneration

MSCEVs also show potential in skeletal muscle regeneration. For example, human BMMSCEVs can potentially augment myogenesis and angiogenesis in vitro (mediated by miR-494) and enhance muscle regeneration [97][69]. Amniotic fluid MSCEVs comprise several proteins and miRNAs that can regulate inflammation and angiogenesis, boosting skeletal muscle regeneration [98][70]. A bioinformatics study (miRNA profiling and proteomics) that evaluated the regenerative potential of human AMSCEVs in muscle injury revealed that repair was mediated by factors that were distributed in both MSCEVs and the soluble fraction of secretomes [99][71].
Reports suggest that human AMSC treatment protects from muscle injury associated with torn rotator cuff. MSCEV treatment in a rat model inhibited atrophy, fatty infiltration, inflammation, and vascularization of muscles in torn rotator cuffs, and enhanced the myofiber regeneration and biomechanical properties [100][72]. MSCEVs derived from human urine are also known to promote the repair of pubococcygeus muscle injury in a stress urinary incontinence rat model. This is done via stimulation of extracellular regulated protein (ERP) kinase phosphorylation and activation, proliferation, and differentiation of muscle satellite cells [101][73]. In addition, human ASCEVs were also reported to prevent muscle damage in a mouse model of critical hindlimb ischemia via neuregulin 1 protein (NRG1)-mediated signals, which play an important role in angiogenesis, muscle protection, and prevention of inflammation [102][74].

7. MSCEVs and Cartilage Regeneration

Injured articular cartilage has restricted endogenous regeneration ability. Poor healing of cartilage may result in osteoarthritis (OA) [103][75]. A study highlighted the therapeutic action of MSCEVs on their cellular origin during OA treatment. The study compared amniotic fluid MSCs (AFMSCs) and AFMSCEVs. Animals with defects treated using AFMSCEVs exhibited higher pain tolerance and histological scores compared to AFMSCs [104][76]. Human BMMSCEVs stimulate in vitro regeneration of cartilage by triggering type-2 collagen and proteoglycans of chondrocyte production, which assists in cartilage repair [105][77]. OA is related to cartilage degradation via Wnt5A (noncanonical Wnt protein), which activates matrix metalloproteinase (MMP) and reduces cartilage ECM formation [106][78]. Another investigation suggests that miR92a-3p enriched human BMMSCEVs suppress degradation of cartilage and stimulate cartilage repair in an OA mouse model in vitro as well as in vivo, attributed to miR-92a-3p targeting Wnt5A [107][79]. Also, pre-conditioning of rat MSCs with transforming growth factor beta (TGFβ) increases the quantity of miR-135b in derived EVs, which causes stimulation of chondrocyte proliferation in vitro via specificity protein 1 (Sp1) regulation and cartilage tissue repair in an OA rat model [108][80].
A study suggests that administration of human embryonic MSCEVs in an osteochondral defect rat and mouse model caused osteochondral regeneration through well-arranged mechanisms, such as augmentation of chondrocyte proliferation, attenuation of apoptosis, and regulation of immunoreactivity at the injury site, along with formation of balance and degradation of cartilage ECM, and restoration of matrix homeostasis [109,110][81][82]. Three-dimensional culture of UCMSCs in a hollow-fiber bioreactor resulted in high yield and exceptional therapeutic potential of UCMSCEVs in a cartilage defect rabbit model compared to MSCEVs from a conventional 2D culture [111][83]. To retain MSCEVs at the cartilage injury site, human iPSCEVs can be incorporated in situ with hydrogel glue. Such cellular tissue patches can assimilate with the native cartilage matrix and enhance cell deposition at cartilage defect sites, thereby resulting in cartilage repair [112][84]. Three-dimensional printing is an advanced fabrication technique for tissue engineering that enables the development of complex forms with high precision [113][85]. BMMSCEVs are fabricated with cartilage ECM/gelatin methacrylate hydrogel as a bio-ink to design bio-scaffolds. The 3D-printed device enables target delivery of BMMSCEVs to prevent mitochondrial dysfunction in degenerative chondrocytes in vitro and to assist in cartilage regeneration in an osteochondral defect rabbit model in vivo [114][86].

8. MSCEVs and Wound Healing

The wound healing process involves complex molecular and cellular events, such as angiogenesis, cellular migration, ECM deposition, proliferation, and tissue remodeling [115][87]. Wounds with impaired healing fail to progress via normal healing stages (inflammation, homeostasis, proliferation, and remodeling), which leads to excessive scar formation [116][88]. MSCEVs exhibit beneficial effects in several chronic types of wounds. A study demonstrated that BMMSCEVs enhanced fibroblast proliferation and migration in healthy people and chronic wound patients (in a dose-dependent manner) ex vivo, and mediated tube formation by endothelial cells via activation of Akt, ERK, and STAT3 wound healing pathways [117][89]. An in vitro study suggested that human iPSCEVs had potential in cutaneous wound healing, by increasing collagen synthesis and angiogenesis [118][90]. AMSCEVs also showed the potential to increase collagen and elastin synthesis in photodamaged human dermal fibroblasts in vitro [119][91]. In vivo administration of AMSCEVs using a mouse skin incision model showed increased wound healing by modification of the phenotype character of fibroblasts. Collagen 1 and 3 secretion from fibroblasts increases during an early stage of wound healing, whereas collagen synthesis diminishes to reduce scar formation during the later stages [120][92]. A report suggested that AMSCEVs trigger keratinocyte and fibroblast migration and proliferation in an excisional wound splinting rat model via activation of the Akt pathway [121][93]. A comparative study of BMMSCEVs and rabbit ADMSCEVs in a rat cutaneous wound model showed significant healing [122][94]. Human UCMSCEVs are known to promote healing of second-degree burn wounds in vivo by activating the Wnt/β-catenin signaling pathway, increasing dermal fibroblast proliferation and angiogenesis, and reducing skin cell apoptosis [123,124][95][96]. Wound healing and suppressed scar formation are facilitated by inhibition of myofibroblast differentiation at the site of the skin defect upon treatment with human UCMSCEVs. Such effect is attributed to the activity of specific microRNAs (miR-21, 23a, 125b, and 145) [125][97].

9. MSCEVs and Other Tissue Regeneration

Apart from their regenerative potential in the mentioned organs and tissues, MSCEVs have regenerative potential against injuries or diseases of several other organs, such as blood vessels, esophagus, lung, and bowel. For example, human placenta MSCEVs attenuated in vitro lung cell injury (by lipopolysaccharide stimulation) [126][98]; swine BMMSCEVs improved in vivo lung functioning in a pig model of influenza virus-induced acute lung injury [127][99]; human BMMSCEVs alleviated pulmonary vascular permeability and lung injury (induced by hemorrhagic shock and trauma) in a mouse model (via activation of proteins and pathways linked to cytoskeletal rearrangement of vascular permeability) [128][100]; and human placenta MSCEVs inhibited calcification of synthetic vascular grafts by immunomodulation and improved vascular performance and functionality in a hyperlipidemia rat model [129][101].
In the same way, AMSCEVs limit abnormal proliferation and migration of vascular smooth muscle cells, and neointimal hyperplasia in vein graft bypass surgery [130][102]; Mouse BMMSCEVs are known to improve ulcerative colitis symptoms in a dextran sodium sulfate-induced mouse model [131][103]; and human UCMSCEVs ameliorate severe ischemic injury in a hindlimb ischemia mouse model [132][104]. AMSCEVs (inserted into thermo response hydrogels) are also known to augment healing and ensure target delivery at the site of disease in cases of tracheoesophageal diseases like fistula [133][105]. Similarly, numerous MSCEVs are applied in tissue engineering of various tissues and organs (Figure 1).
Figure 1.
Applications of mesenchymal stem cells derived extracellular vesicles (MSCEVs) in tissue engineering.

References

  1. Beltrami, A.P.; Urbanek, K.; Kajstura, J.; Yan, S.-M.; Finato, N.; Bussani, R.; Nadal-Ginard, B.; Silvestri, F.; Leri, A.; Beltrami, C.A. Evidence That Human Cardiac Myocytes Divide after Myocardial Infarction. New Engl. J. Med. 2001, 344, 1750–1757.
  2. Senyo, S.E.; Steinhauser, M.L.; Pizzimenti, C.L.; Yang, V.K.; Cai, L.; Wang, M.; Wu, T.-D.; Guerquin-Kern, J.-L.; Lechene, C.P.; Lee, R.T. Mammalian Heart Renewal by Pre-Existing Cardiomyocytes. Nature 2013, 493, 433–436.
  3. Lai, R.C.; Arslan, F.; Lee, M.M.; Sze, N.S.K.; Choo, A.; Chen, T.S.; Salto-Tellez, M.; Timmers, L.; Lee, C.N.; El Oakley, R.M. Exosome Secreted by MSC Reduces Myocardial Ischemia/Reperfusion Injury. Stem Cell Res. 2010, 4, 214–222.
  4. Arslan, F.; Lai, R.C.; Smeets, M.B.; Akeroyd, L.; Choo, A.; Aguor, E.N.; Timmers, L.; van Rijen, H.V.; Doevendans, P.A.; Pasterkamp, G. Mesenchymal Stem Cell-Derived Exosomes Increase ATP Levels, Decrease Oxidative Stress and Activate PI3K/Akt Pathway to Enhance Myocardial Viability and Prevent Adverse Remodeling after Myocardial Ischemia/Reperfusion Injury. Stem Cell Res. 2013, 10, 301–312.
  5. Ma, J.; Zhao, Y.; Sun, L.; Sun, X.; Zhao, X.; Sun, X.; Qian, H.; Xu, W.; Zhu, W. Exosomes Derived from AKt-Modified Human Umbilical Cord Mesenchymal Stem Cells Improve Cardiac Regeneration and Promote Angiogenesis via Activating Platelet-Derived Growth Factor D. Stem Cells Transl. Med. 2017, 6, 51–59.
  6. Adamiak, M.; Cheng, G.; Bobis-Wozowicz, S.; Zhao, L.; Kedracka-Krok, S.; Samanta, A.; Karnas, E.; Xuan, Y.-T.; Skupien-Rabian, B.; Chen, X. Induced Pluripotent Stem Cell (IPSC)–Derived Extracellular Vesicles Are Safer and More Effective for Cardiac Repair than IPSCs. Circ. Res. 2018, 122, 296–309.
  7. Balbi, C.; Lodder, K.; Costa, A.; Moimas, S.; Moccia, F.; van Herwaarden, T.; Rosti, V.; Campagnoli, F.; Palmeri, A.; De Biasio, P. Reactivating Endogenous Mechanisms of Cardiac Regeneration via Paracrine Boosting Using the Human Amniotic Fluid Stem Cell Secretome. Int. J. Cardiol. 2019, 287, 87–95.
  8. Huang, P.; Wang, L.; Li, Q.; Xu, J.; Xu, J.; Xiong, Y.; Chen, G.; Qian, H.; Jin, C.; Yu, Y. Combinatorial Treatment of Acute Myocardial Infarction Using Stem Cells and Their Derived Exosomes Resulted in Improved Heart Performance. Stem Cell Res. Ther. 2019, 10, 1–12.
  9. Hu, X.; Wu, R.; Shehadeh, L.A.; Zhou, Q.; Jiang, C.; Huang, X.; Zhang, L.; Gao, F.; Liu, X.; Yu, H. Severe Hypoxia Exerts Parallel and Cell-Specific Regulation of Gene Expression and Alternative Splicing in Human Mesenchymal Stem Cells. BMC Genom. 2014, 15, 303.
  10. Hu, X.; Xu, Y.; Zhong, Z.; Wu, Y.; Zhao, J.; Wang, Y.; Cheng, H.; Kong, M.; Zhang, F.; Chen, Q. A Large-Scale Investigation of Hypoxia-Preconditioned Allogeneic Mesenchymal Stem Cells for Myocardial Repair in Nonhuman Primates: Paracrine Activity without Remuscularization. Circ. Res. 2016, 118, 970–983.
  11. Bian, S.; Zhang, L.; Duan, L.; Wang, X.; Min, Y.; Yu, H. Extracellular Vesicles Derived from Human Bone Marrow Mesenchymal Stem Cells Promote Angiogenesis in a Rat Myocardial Infarction Model. J. Mol. Med. 2014, 92, 387–397.
  12. Zhu, L.-P.; Tian, T.; Wang, J.-Y.; He, J.-N.; Chen, T.; Pan, M.; Xu, L.; Zhang, H.; Qiu, X.-T.; Li, C.-C. 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.
  13. Zhu, J.; Lu, K.; Zhang, N.; Zhao, Y.; Ma, Q.; Shen, J.; Lin, Y.; Xiang, P.; Tang, Y.; Hu, X. 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. Artif. Cells Nanomed. Biotechnol. 2018, 46, 1659–1670.
  14. Han, C.; Zhou, J.; Liang, C.; Liu, B.; Pan, X.; Zhang, Y.; Wang, Y.; Yan, B.; Xie, W.; Liu, F. Human Umbilical Cord Mesenchymal Stem Cell Derived Exosomes Encapsulated in Functional Peptide Hydrogels Promote Cardiac Repair. Biomater. Sci. 2019, 7, 2920–2933.
  15. Boni, R.; Ali, A.; Shavandi, A.; Clarkson, A.N. Current and Novel Polymeric Biomaterials for Neural Tissue Engineering. J. Biomed. Sci. 2018, 25, 90.
  16. Ma, Y.; Ge, S.; Zhang, J.; Zhou, D.; Li, L.; Wang, X.; Su, J. Mesenchymal Stem Cell-Derived Extracellular Vesicles Promote Nerve Regeneration after Sciatic Nerve Crush Injury in Rats. Int. J. Clin. Exp. Pathol. 2017, 10, 10032.
  17. Ma, Y.; Dong, L.; Zhou, D.; Li, L.; Zhang, W.; Zhen, Y.; Wang, T.; Su, J.; Chen, D.; Mao, C. Extracellular Vesicles from Human Umbilical Cord Mesenchymal Stem Cells Improve Nerve Regeneration after Sciatic Nerve Transection in Rats. J. Cell. Mol. Med. 2019, 23, 2822–2835.
  18. Bucan, V.; Vaslaitis, D.; Peck, C.-T.; Strauß, S.; Vogt, P.M.; Radtke, C. Effect of Exosomes from Rat Adipose-Derived Mesenchymal Stem Cells on Neurite Outgrowth and Sciatic Nerve Regeneration after Crush Injury. Mol. Neurobiol. 2019, 56, 1812–1824.
  19. Haertinger, M.; Weiss, T.; Mann, A.; Tabi, A.; Brandel, V.; Radtke, C. Adipose Stem Cell-Derived Extracellular Vesicles Induce Proliferation of Schwann Cells via Internalization. Cells 2020, 9, 163.
  20. Mao, Q.; Nguyen, P.D.; Shanti, R.M.; Shi, S.; Shakoori, P.; Zhang, Q.; Le, A.D. Gingiva-Derived Mesenchymal Stem Cell-Extracellular Vesicles Activate Schwann Cell Repair Phenotype and Promote Nerve Regeneration. Tissue Eng. Part. A 2019, 25, 887–900.
  21. Xin, H.; Li, Y.; Buller, B.; Katakowski, M.; Zhang, Y.; Wang, X.; Shang, X.; Zhang, Z.G.; Chopp, M. Exosome-Mediated Transfer of MiR-133b from Multipotent Mesenchymal Stromal Cells to Neural Cells Contributes to Neurite Outgrowth. Stem Cells 2012, 30, 1556–1564.
  22. Zanetta, L.; Marcus, S.G.; Vasile, J.; Dobryansky, M.; Cohen, H.; Eng, K.; Shamamian, P.; Mignatti, P. Expression of Von Willebrand Factor, an Endothelial Cell Marker, is up-Regulated by Angiogenesis Factors: A Potential Method for Objective Assessment of Tumor Angiogenesis. Int. J. Cancer 2000, 85, 281–288.
  23. Couillard-Despres, S.; Winner, B.; Schaubeck, S.; Aigner, R.; Vroemen, M.; Weidner, N.; Bogdahn, U.; Winkler, J.; Kuhn, H.-G.; Aigner, L. Doublecortin Expression Levels in Adult Brain Reflect Neurogenesis. Eur. J. Neurosci. 2005, 21, 1–14.
  24. Xin, H.; Li, Y.; Cui, Y.; Yang, J.J.; Zhang, Z.G.; Chopp, M. Systemic Administration of Exosomes Released from Mesenchymal Stromal Cells Promote Functional Recovery and Neurovascular Plasticity after Stroke in Rats. J. Cereb. Blood Flow Metab. 2013, 33, 1711–1715.
  25. Zhang, Y.; Chopp, M.; Meng, Y.; Katakowski, M.; Xin, H.; Mahmood, A.; Xiong, Y. Effect of Exosomes Derived from Multipluripotent Mesenchymal Stromal Cells on Functional Recovery and Neurovascular Plasticity in Rats after Traumatic Brain Injury. J. Neurosurg. 2015, 122, 856–867.
  26. Cheng, J.; Korte, N.; Nortley, R.; Sethi, H.; Tang, Y.; Attwell, D. Targeting Pericytes for Therapeutic Approaches to Neurological Disorders. Acta Neuropathol. 2018, 136, 507–523.
  27. Figley, S.A.; Khosravi, R.; Legasto, J.M.; Tseng, Y.-F.; Fehlings, M.G. Characterization of Vascular Disruption and Blood–Spinal Cord Barrier Permeability Following Traumatic Spinal Cord Injury. J. Neurotrauma 2014, 31, 541–552.
  28. Lu, Y.; Zhou, Y.; Zhang, R.; Wen, L.; Wu, K.; Li, Y.; Yao, Y.; Duan, R.; Jia, Y. Bone Mesenchymal Stem Cell-Derived Extracellular Vesicles Promote Recovery Following Spinal Cord Injury via Improvement of the Integrity of the Blood-Spinal Cord Barrier. Front. Neurosci. 2019, 13, 209.
  29. Li, C.; Jiao, G.; Wu, W.; Wang, H.; Ren, S.; Zhang, L.; Zhou, H.; Liu, H.; Chen, Y. Exosomes from Bone Marrow Mesenchymal Stem Cells Inhibit Neuronal Apoptosis and Promote Motor Function Recovery via the Wnt/β-Catenin Signaling Pathway. Cell Transplant. 2019, 28, 1373–1383.
  30. Li, D.; Zhang, P.; Yao, X.; Li, H.; Shen, H.; Li, X.; Wu, J.; Lu, X. Exosomes Derived from MiR-133b-Modified Mesenchymal Stem Cells Promote Recovery after Spinal Cord Injury. Front. Neurosci. 2018, 12, 845.
  31. Clark, K.; Zhang, S.; Barthe, S.; Kumar, P.; Pivetti, C.; Kreutzberg, N.; Reed, C.; Wang, Y.; Paxton, Z.; Farmer, D. Placental Mesenchymal Stem Cell-Derived Extracellular Vesicles Promote Myelin Regeneration in an Animal Model of Multiple Sclerosis. Cells 2019, 8, 1497.
  32. Riazifar, M.; Mohammadi, M.R.; Pone, E.J.; Yeri, A.; Lasser, C.; Segaliny, A.I.; McIntyre, L.L.; Shelke, G.V.; Hutchins, E.; Hamamoto, A. Stem Cell-Derived Exosomes as Nanotherapeutics for Autoimmune and Neurodegenerative Disorders. ACS Nano 2019, 13, 6670–6688.
  33. Reza-Zaldivar, E.E.; Hernández-Sapiéns, M.A.; Gutiérrez-Mercado, Y.K.; Sandoval-Ávila, S.; Gomez-Pinedo, U.; Márquez-Aguirre, A.L.; Vázquez-Méndez, E.; Padilla-Camberos, E.; Canales-Aguirre, A.A. Mesenchymal Stem Cell-Derived Exosomes Promote Neurogenesis and Cognitive Function Recovery in a Mouse Model of Alzheimer’s Disease. Neural Regen. Res. 2019, 14, 1626.
  34. Sisa, C.; Kholia, S.; Naylor, J.; Herrera Sanchez, M.B.; Bruno, S.; Deregibus, M.C.; Camussi, G.; Inal, J.M.; Lange, S.; Hristova, M. Mesenchymal Stromal Cell Derived Extracellular Vesicles Reduce Hypoxia-Ischaemia Induced Perinatal Brain Injury. Front. Physiol. 2019, 10.
  35. Ophelders, D.R.; Wolfs, T.G.; Jellema, R.K.; Zwanenburg, A.; Andriessen, P.; Delhaas, T.; Ludwig, A.-K.; Radtke, S.; Peters, V.; Janssen, L. Mesenchymal Stromal Cell-Derived Extracellular Vesicles Protect the Fetal Brain after Hypoxia-Ischemia. Stem Cells Transl. Med. 2016, 5, 754–763.
  36. Tsiapalis, D.; O’Driscoll, L. Mesenchymal Stem Cell Derived Extracellular Vesicles for Tissue Engineering and Regenerative Medicine Applications. Cells 2020, 9, 991.
  37. Qin, Y.; Wang, L.; Gao, Z.; Chen, G.; Zhang, C. Bone Marrow Stromal/Stem Cell-Derived Extracellular Vesicles Regulate Osteoblast Activity and Differentiation in Vitro and Promote Bone Regeneration in Vivo. Sci. Rep. 2016, 6, 1–11.
  38. Liang, B.; Liang, J.-M.; Ding, J.-N.; Xu, J.; Xu, J.-G.; Chai, Y.-M. Dimethyloxaloylglycine-Stimulated Human Bone Marrow Mesenchymal Stem Cell-Derived Exosomes Enhance Bone Regeneration through Angiogenesis by Targeting the AKT/MTOR Pathway. Stem Cell Res. Ther. 2019, 10, 335.
  39. Lu, Z.; Chen, Y.; Dunstan, C.; Roohani-Esfahani, S.; Zreiqat, H. Priming Adipose Stem Cells with Tumor Necrosis Factor-Alpha Preconditioning Potentiates Their Exosome Efficacy for Bone Regeneration. Tissue Eng. Part. A 2017, 23, 1212–1220.
  40. Furuta, T.; Miyaki, S.; Ishitobi, H.; Ogura, T.; Kato, Y.; Kamei, N.; Miyado, K.; Higashi, Y.; Ochi, M. Mesenchymal Stem Cell-Derived Exosomes Promote Fracture Healing in a Mouse Model. Stem Cells Transl. Med. 2016, 5, 1620–1630.
  41. Huang, C.-C.; Narayanan, R.; Alapati, S.; Ravindran, S. Exosomes as Biomimetic Tools for Stem Cell Differentiation: Applications in Dental Pulp Tissue Regeneration. Biomaterials 2016, 111, 103–115.
  42. Naruphontjirakul, P.; Tsigkou, O.; Li, S.; Porter, A.E.; Jones, J.R. Human Mesenchymal Stem Cells Differentiate into an Osteogenic Lineage in Presence of Strontium Containing Bioactive Glass Nanoparticles. Acta Biomater. 2019, 90, 373–392.
  43. Li, W.; Liu, Y.; Zhang, P.; Tang, Y.; Zhou, M.; Jiang, W.; Zhang, X.; Wu, G.; Zhou, Y. Tissue-Engineered Bone Immobilized with Human Adipose Stem Cells-Derived Exosomes Promotes Bone Regeneration. ACS Appl. Mater. Interfaces 2018, 10, 5240–5254.
  44. Zhang, J.; Liu, X.; Li, H.; Chen, C.; Hu, B.; Niu, X.; Li, Q.; Zhao, B.; Xie, Z.; Wang, Y. Exosomes/Tricalcium Phosphate Combination Scaffolds Can Enhance Bone Regeneration by Activating the PI3K/Akt Signaling Pathway. Stem Cell Res. Ther. 2016, 7, 1–14.
  45. Xie, H.; Wang, Z.; Zhang, L.; Lei, Q.; Zhao, A.; Wang, H.; Li, Q.; Cao, Y.; Zhang, W.J.; Chen, Z. Extracellular Vesicle-Functionalized Decalcified Bone Matrix Scaffolds with Enhanced pro-Angiogenic and pro-Bone Regeneration Activities. Sci. Rep. 2017, 7, 45622.
  46. Tan, C.Y.; Lai, R.C.; Wong, W.; Dan, Y.Y.; Lim, S.-K.; Ho, H.K. Mesenchymal Stem Cell-Derived Exosomes Promote Hepatic Regeneration in Drug-Induced Liver Injury Models. Stem Cell Res. Ther. 2014, 5, 76.
  47. Nong, K.; Wang, W.; Niu, X.; Hu, B.; Ma, C.; Bai, Y.; Wu, B.; Wang, Y.; Ai, K. Hepatoprotective Effect of Exosomes from Human-Induced Pluripotent Stem Cell–Derived Mesenchymal Stromal Cells against Hepatic Ischemia-Reperfusion Injury in Rats. Cytotherapy 2016, 18, 1548–1559.
  48. Du, Y.; Li, D.; Han, C.; Wu, H.; Xu, L.; Zhang, M.; Zhang, J.; Chen, X. Exosomes from Human-Induced Pluripotent Stem Cell–Derived Mesenchymal Stromal Cells (HiPSC-MSCs) Protect Liver against Hepatic Ischemia/Reperfusion Injury via Activating Sphingosine Kinase and Sphingosine-1-Phosphate Signaling Pathway. Cell. Physiol. Biochem. 2017, 43, 611–625.
  49. Ng, M.L.; Yarla, N.S.; Menschikowski, M.; Sukocheva, O.A. Regulatory Role of Sphingosine Kinase and Sphingosine-1-Phosphate Receptor Signaling in Progenitor/Stem Cells. World J. Stem Cells 2018, 10, 119.
  50. Yao, J.; Zheng, J.; Cai, J.; Zeng, K.; Zhou, C.; Zhang, J.; Li, S.; Li, H.; Chen, L.; He, L. Extracellular Vesicles Derived from Human Umbilical Cord Mesenchymal Stem Cells Alleviate Rat Hepatic Ischemia-Reperfusion Injury by Suppressing Oxidative Stress and Neutrophil Inflammatory Response. FASEB J. 2019, 33, 1695–1710.
  51. Mardpour, S.; Ghanian, M.H.; Sadeghi-Abandansari, H.; Mardpour, S.; Nazari, A.; Shekari, F.; Baharvand, H. Hydrogel-Mediated Sustained Systemic Delivery of Mesenchymal Stem Cell-Derived Extracellular Vesicles Improves Hepatic Regeneration in Chronic Liver Failure. ACS Appl. Mater. Interfaces 2019, 11, 37421–37433.
  52. Bruno, S.; Grange, C.; Deregibus, M.C.; Calogero, R.A.; Saviozzi, S.; Collino, F.; Morando, L.; Falda, M.; Bussolati, B.; Tetta, C. Mesenchymal Stem Cell-Derived Microvesicles Protect against Acute Tubular Injury. J. Am. Soc. Nephrol. 2009, 20, 1053–1067.
  53. Zhao, L.; Hu, C.; Zhang, P.; Jiang, H.; Chen, J. Genetic Communication by Extracellular Vesicles Is an Important Mechanism Underlying Stem Cell-Based Therapy-Mediated Protection against Acute Kidney Injury. Stem Cell Res. Ther. 2019, 10, 1–9.
  54. Nawaz, M.; Fatima, F.; Vallabhaneni, K.C.; Penfornis, P.; Valadi, H.; Ekström, K.; Kholia, S.; Whitt, J.D.; Fernandes, J.D.; Pochampally, R.; et al. Extracellular Vesicles: Evolving Factors In Stem Cell Biology. Stem Cells Int. 2016.
  55. Shen, B.; Liu, J.; Zhang, F.; Wang, Y.; Qin, Y.; Zhou, Z.; Qiu, J.; Fan, Y. CCR2 Positive Exosome Released by Mesenchymal Stem Cells Suppresses Macrophage Functions and Alleviates Ischemia/Reperfusion-Induced Renal Injury. Stem Cells Int. 2016, 2016, 1240301.
  56. Bruno, S.; Grange, C.; Collino, F.; Deregibus, M.C.; Cantaluppi, V.; Biancone, L.; Tetta, C.; Camussi, G. Microvesicles Derived from Mesenchymal Stem Cells Enhance Survival in a Lethal Model of Acute Kidney Injury. PLoS ONE 2012, 7, e33115.
  57. Zhou, Y.; Liu, S.; Zhao, M.; Wang, C.; Li, L.; Yuan, Y.; Li, L.; Liao, G.; Bresette, W.; Zhang, J. Injectable Extracellular Vesicle-Released Self-Assembling Peptide Nanofiber Hydrogel as an Enhanced Cell-Free Therapy for Tissue Regeneration. J. Control. Release 2019, 316, 93–104.
  58. Ju, G.; Cheng, J.; Zhong, L.; Wu, S.; Zou, X.; Zhang, G.; Gu, D.; Miao, S.; Zhu, Y.; Sun, J. Microvesicles Derived from Human Umbilical Cord Mesenchymal Stem Cells Facilitate Tubular Epithelial Cell Dedifferentiation and Growth via Hepatocyte Growth Factor Induction. PLoS ONE 2015, 10, e0121534.
  59. Gu, D.; Zou, X.; Ju, G.; Zhang, G.; Bao, E.; Zhu, Y. Mesenchymal Stromal Cells Derived Extracellular Vesicles Ameliorate Acute Renal Ischemia Reperfusion Injury by Inhibition of Mitochondrial Fission through MiR-30. Stem Cells Int. 2016, 2016, 2093940.
  60. Ranghino, A.; Bruno, S.; Bussolati, B.; Moggio, A.; Dimuccio, V.; Tapparo, M.; Biancone, L.; Gontero, P.; Frea, B.; Camussi, G. The Effects of Glomerular and Tubular Renal Progenitors and Derived Extracellular Vesicles on Recovery from Acute Kidney Injury. Stem Cell Res. Ther. 2017, 8, 24.
  61. Piccoli, G.B.; Grassi, G.; Cabiddu, G.; Nazha, M.; Roggero, S.; Capizzi, I.; De Pascale, A.; Priola, A.M.; Di Vico, C.; Maxia, S. Chapter II. Diabetic Nephropathy. Rev. Diabet Stud. 2015, 12, 87–109.
  62. Jiang, Z.; Liu, Y.; Niu, X.; Yin, J.; Hu, B.; Guo, S.; Fan, Y.; Wang, Y.; Wang, N. Exosomes Secreted by Human Urine-Derived Stem Cells Could Prevent Kidney Complications from Type I Diabetes in Rats. Stem Cell Res. Ther. 2016, 7, 24.
  63. Nagaishi, K.; Mizue, Y.; Chikenji, T.; Otani, M.; Nakano, M.; Konari, N.; Fujimiya, M. Mesenchymal Stem Cell Therapy Ameliorates Diabetic Nephropathy via the Paracrine Effect of Renal Trophic Factors Including Exosomes. Sci. Rep. 2016, 6, 1–16.
  64. Grange, C.; Tritta, S.; Tapparo, M.; Cedrino, M.; Tetta, C.; Camussi, G.; Brizzi, M.F. Stem Cell-Derived Extracellular Vesicles Inhibit and Revert Fibrosis Progression in a Mouse Model of Diabetic Nephropathy. Sci. Rep. 2019, 9, 1–13.
  65. Kholia, S.; Herrera Sanchez, M.B.; Cedrino, M.; Papadimitriou, E.; Tapparo, M.; Deregibus, M.C.; Brizzi, M.F.; Tetta, C.; Camussi, G. Human Liver Stem Cell-Derived Extracellular Vesicles Prevent Aristolochic Acid-Induced Kidney Fibrosis. Front. Immunol. 2018, 9, 1639.
  66. He, J.; Wang, Y.; Lu, X.; Zhu, B.; Pei, X.; Wu, J.; Zhao, W. Micro-Vesicles Derived from Bone Marrow Stem Cells Protect the Kidney Both in Vivo and in Vitro by MicroRNA-Dependent Repairing. Nephrology 2015, 20, 591–600.
  67. Matsukura, T.; Inaba, C.; Weygant, E.A.; Kitamura, D.; Janknecht, R.; Matsumoto, H.; Hyink, D.P.; Kashiwada, S.; Obara, T. Extracellular Vesicles from Human Bone Marrow Mesenchymal Stem Cells Repair Organ Damage Caused by Cadmium Poisoning in a Medaka Model. Physiol. Rep. 2019, 7, e14172.
  68. Nassar, W.; El-Ansary, M.; Sabry, D.; Mostafa, M.A.; Fayad, T.; Kotb, E.; Temraz, M.; Saad, A.-N.; Essa, W.; Adel, H. Umbilical Cord Mesenchymal Stem Cells Derived Extracellular Vesicles Can Safely Ameliorate the Progression of Chronic Kidney Diseases. Biomater. Res. 2016, 20, 21.
  69. Nakamura, Y.; Miyaki, S.; Ishitobi, H.; Matsuyama, S.; Nakasa, T.; Kamei, N.; Akimoto, T.; Higashi, Y.; Ochi, M. Mesenchymal-Stem-Cell-Derived Exosomes Accelerate Skeletal Muscle Regeneration. Febs Lett. 2015, 589, 1257–1265.
  70. Mellows, B.; Mitchell, R.; Antonioli, M.; Kretz, O.; Chambers, D.; Zeuner, M.-T.; Denecke, B.; Musante, L.; Ramachandra, D.L.; Debacq-Chainiaux, F. Protein and Molecular Characterization of a Clinically Compliant Amniotic Fluid Stem Cell-Derived Extracellular Vesicle Fraction Capable of Accelerating Muscle Regeneration through Enhancement of Angiogenesis. Stem Cells Dev. 2017, 26, 1316–1333.
  71. Mitchell, R.; Mellows, B.; Sheard, J.; Antonioli, M.; Kretz, O.; Chambers, D.; Zeuner, M.-T.; Tomkins, J.E.; Denecke, B.; Musante, L. Secretome of Adipose-Derived Mesenchymal Stem Cells Promotes Skeletal Muscle Regeneration through Synergistic Action of Extracellular Vesicle Cargo and Soluble Proteins. Stem Cell Res. Ther. 2019, 10, 116.
  72. Wang, C.; Song, W.; Chen, B.; Liu, X.; He, Y. Exosomes Isolated from Adipose-Derived Stem Cells: A New Cell-Free Approach to Prevent the Muscle Degeneration Associated with Torn Rotator Cuffs. Am. J. Sports Med. 2019, 47, 3247–3255.
  73. Wu, R.; Huang, C.; Wu, Q.; Jia, X.; Liu, M.; Xue, Z.; Qiu, Y.; Niu, X.; Wang, Y. Exosomes Secreted by Urine-Derived Stem Cells Improve Stress Urinary Incontinence by Promoting Repair of Pubococcygeus Muscle Injury in Rats. Stem Cell Res. Ther. 2019, 10, 1–13.
  74. Figliolini, F.; Ranghino, A.; Grange, C.; Cedrino, M.; Tapparo, M.; Cavallari, C.; Rossi, A.; Togliatto, G.; Femminò, S.; Gugliuzza, M.V. Extracellular Vesicles from Adipose Stem Cells Prevent Muscle Damage and Inflammation in a Mouse Model of Hind Limb Ischemia: Role of Neuregulin-1. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 239–254.
  75. Loeser, R.F.; Goldring, S.R.; Scanzello, C.R.; Goldring, M.B. Osteoarthritis: A Disease of the Joint as an Organ. Arthritis Rheum. 2012, 64, 1697.
  76. Zavatti, M.; Beretti, F.; Casciaro, F.; Bertucci, E.; Maraldi, T. Comparison of the Therapeutic Effect of Amniotic Fluid Stem Cells and Their Exosomes on Monoiodoacetate-Induced Animal Model of Osteoarthritis. Biofactors 2020, 46, 106–117.
  77. Vonk, L.A.; van Dooremalen, S.F.; Liv, N.; Klumperman, J.; Coffer, P.J.; Saris, D.B.; Lorenowicz, M.J. Mesenchymal Stromal/Stem Cell-Derived Extracellular Vesicles Promote Human Cartilage Regeneration in Vitro. Theranostics 2018, 8, 906.
  78. Hosseini-Farahabadi, S.; Geetha-Loganathan, P.; Fu, K.; Nimmagadda, S.; Yang, H.J.; Richman, J.M. Dual Functions for WNT5A during Cartilage Development and in Disease. Matrix Biol. 2013, 32, 252–264.
  79. Mao, G.; Zhang, Z.; Hu, S.; Zhang, Z.; Chang, Z.; Huang, Z.; Liao, W.; Kang, Y. Exosomes Derived from MiR-92a-3p-Overexpressing Human Mesenchymal Stem Cells Enhance Chondrogenesis and Suppress Cartilage Degradation via Targeting WNT5A. Stem Cell Res. Ther. 2018, 9, 1–13.
  80. Wang, R.; Xu, B.; Xu, H. TGF-Β1 Promoted Chondrocyte Proliferation by Regulating Sp1 through MSC-Exosomes Derived MiR-135b. Cell Cycle 2018, 17, 2756–2765.
  81. Zhang, S.; Chuah, S.J.; Lai, R.C.; Hui, J.H.P.; Lim, S.K.; Toh, W.S. MSC Exosomes Mediate Cartilage Repair by Enhancing Proliferation, Attenuating Apoptosis and Modulating Immune Reactivity. Biomaterials 2018, 156, 16–27.
  82. Wang, Y.; Yu, D.; Liu, Z.; Zhou, F.; Dai, J.; Wu, B.; Zhou, J.; Heng, B.C.; Zou, X.H.; Ouyang, H. Exosomes from Embryonic Mesenchymal Stem Cells Alleviate Osteoarthritis through Balancing Synthesis and Degradation of Cartilage Extracellular Matrix. Stem Cell Res. Ther. 2017, 8, 1–13.
  83. Yan, L.; Wu, X. Exosomes Produced from 3D Cultures of Umbilical Cord Mesenchymal Stem Cells in a Hollow-Fiber Bioreactor Show Improved Osteochondral Regeneration Activity. Cell Biol. Toxicol. 2020, 36, 165–178.
  84. Liu, X.; Yang, Y.; Li, Y.; Niu, X.; Zhao, B.; Wang, Y.; Bao, C.; Xie, Z.; Lin, Q.; Zhu, L. Integration of Stem Cell-Derived Exosomes with in Situ Hydrogel Glue as a Promising Tissue Patch for Articular Cartilage Regeneration. Nanoscale 2017, 9, 4430–4438.
  85. Bishop, E.S.; Mostafa, S.; Pakvasa, M.; Luu, H.H.; Lee, M.J.; Wolf, J.M.; Ameer, G.A.; He, T.-C.; Reid, R.R. 3-D Bioprinting Technologies in Tissue Engineering and Regenerative Medicine: Current and Future Trends. Genes Dis. 2017, 4, 185–195.
  86. Chen, P.; Zheng, L.; Wang, Y.; Tao, M.; Xie, Z.; Xia, C.; Gu, C.; Chen, J.; Qiu, P.; Mei, S. Desktop-Stereolithography 3D Printing of a Radially Oriented Extracellular Matrix/Mesenchymal Stem Cell Exosome Bioink for Osteochondral Defect Regeneration. Theranostics 2019, 9, 2439.
  87. Velnar, T.; Bailey, T.; Smrkolj, V. The Wound Healing Process: An Overview of the Cellular and Molecular Mechanisms. J. Int. Med. Res. 2009, 37, 1528–1542.
  88. Zhao, R.; Liang, H.; Clarke, E.; Jackson, C.; Xue, M. Inflammation in Chronic Wounds. Int. J. Mol. Sci. 2016, 17, 2085.
  89. Shabbir, A.; Cox, A.; Rodriguez-Menocal, L.; Salgado, M.; Badiavas, E.V. Mesenchymal Stem Cell Exosomes Induce Proliferation and Migration of Normal and Chronic Wound Fibroblasts, and Enhance Angiogenesis in vitro. Stem Cells Dev. 2015, 24, 1635–1647.
  90. 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.
  91. Choi, J.S.; Cho, W.L.; Choi, Y.J.; Kim, J.D.; Park, H.-A.; Kim, S.Y.; Park, J.H.; Jo, D.-G.; Cho, Y.W. Functional Recovery in Photo-Damaged Human Dermal Fibroblasts by Human Adipose-Derived Stem Cell Extracellular Vesicles. J. Extracell. Vesicles 2019, 8, 1565885.
  92. Hu, L.; Wang, J.; Zhou, X.; Xiong, Z.; Zhao, J.; Yu, R.; Huang, F.; Zhang, H.; Chen, L. Exosomes Derived from Human Adipose Mensenchymal Stem Cells Accelerates Cutaneous Wound Healing via Optimizing the Characteristics of Fibroblasts. Sci. Rep. 2016, 6, 32993.
  93. Da Ferreira, A.F.; da Cunha, P.S.; Carregal, V.M.; de Silva, P.C.; de Miranda, M.C.; Kunrath-Lima, M.; de Melo, M.I.A.; Faraco, C.C.F.; Barbosa, J.L.; Frezard, F. Extracellular Vesicles from Adipose-Derived Mesenchymal Stem/Stromal Cells Accelerate Migration and Activate AKT Pathway in Human Keratinocytes and Fibroblasts Independently of MiR-205 Activity. Stem Cells Int. 2017, 2017, 9841035.
  94. Pelizzo, G.; Avanzini, M.A.; Icaro Cornaglia, A.; De Silvestri, A.; Mantelli, M.; Travaglino, P.; Croce, S.; Romano, P.; Avolio, L.; Iacob, G. Extracellular Vesicles Derived from Mesenchymal Cells: Perspective Treatment for Cutaneous Wound Healing in Pediatrics. Regen. Med. 2018, 13, 385–394.
  95. Casado-Díaz, A.; Quesada-Gómez, J.M.; Dorado, G. Extracellular vesicles derived from mesenchymal stem cells (MSC) in regenerative medicine: Applications in skin wound healing. Front. Bioeng. Biotechnol. 2020, 8, 146.
  96. Zhang, B.; Wu, X.; Zhang, X.; Sun, Y.; Yan, Y.; Shi, H.; Zhu, Y.; Wu, L.; Pan, Z.; Zhu, W. Human Umbilical Cord Mesenchymal Stem Cell Exosomes Enhance Angiogenesis through the Wnt4/β-Catenin Pathway. Stem Cells Transl. Med. 2015, 4, 513–522.
  97. Fang, S.; Xu, C.; Zhang, Y.; Xue, C.; Yang, C.; Bi, H.; Qian, X.; Wu, M.; Ji, K.; Zhao, Y. Umbilical Cord-Derived Mesenchymal Stem Cell-Derived Exosomal Micrornas Suppress Myofibroblast Differentiation by Inhibiting the Transforming Growth Factor-β/SMAD2 Pathway during Wound Healing. Stem Cells Transl. Med. 2016, 5, 1425–1439.
  98. Kim, S.Y.; Joglekar, M.V.; Hardikar, A.A.; Phan, T.H.; Khanal, D.; Tharkar, P.; Limantoro, C.; Johnson, J.; Kalionis, B.; Chrzanowski, W. Placenta Stem/Stromal Cell–Derived Extracellular Vesicles for Potential Use in Lung Repair. Proteomics 2019, 19, 1800166.
  99. Khatri, M.; Richardson, L.A.; Meulia, T. Mesenchymal Stem Cell-Derived Extracellular Vesicles Attenuate Influenza Virus-Induced Acute Lung Injury in a Pig Model. Stem Cell Res. Ther. 2018, 9, 1–13.
  100. Potter, D.R.; Miyazawa, B.Y.; Gibb, S.L.; Deng, X.; Togaratti, P.P.; Croze, R.H.; Srivastava, A.K.; Trivedi, A.; Matthay, M.; Holcomb, J.B. Mesenchymal Stem Cell-Derived Extracellular Vesicles Attenuate Pulmonary Vascular Permeability and Lung Injury Induced by Hemorrhagic Shock and Trauma. J. Trauma Acute Care Surg. 2018, 84, 245.
  101. Wei, Y.; Wu, Y.; Zhao, R.; Zhang, K.; Midgley, A.C.; Kong, D.; Li, Z.; Zhao, Q. MSC-Derived SEVs Enhance Patency and Inhibit Calcification of Synthetic Vascular Grafts by Immunomodulation in a Rat Model of Hyperlipidemia. Biomaterials 2019, 204, 13–24.
  102. Almeria, C.; Weiss, R.; Roy, M.; Tripisciano, C.; Kasper, C.; Weber, V.; Egger, D. Hypoxia Conditioned Mesenchymal Stem Cell-Derived Extracellular Vesicles Induce Increased Vascular Tube Formation in Vitro. Front. Bioeng. Biotechnol. 2019, 7, 292.
  103. Cao, L.; Xu, H.; Wang, G.; Liu, M.; Tian, D.; Yuan, Z. Extracellular Vesicles Derived from Bone Marrow Mesenchymal Stem Cells Attenuate Dextran Sodium Sulfate-Induced Ulcerative Colitis by Promoting M2 Macrophage Polarization. Int. Immunopharmacol. 2019, 72, 264–274.
  104. Zhu, Q.; Li, Q.; Niu, X.; Zhang, G.; Ling, X.; Zhang, J.; Wang, Y.; Deng, Z. Extracellular Vesicles Secreted by Human Urine-Derived Stem Cells Promote Ischemia Repair in a Mouse Model of Hind-Limb Ischemia. Cell. Physiol. Biochem. 2018, 47, 1181–1192.
  105. Silva, A.K.; Perretta, S.; Perrod, G.; Pidial, L.; Lindner, V.; Carn, F.; Lemieux, S.; Alloyeau, D.; Boucenna, I.; Menasché, P. Thermoresponsive Gel Embedded with Adipose Stem-Cell-Derived Extracellular Vesicles Promotes Esophageal Fistula Healing in a Thermo-Actuated Delivery Strategy. ACS Nano 2018, 12, 9800–9814.
More
Video Production Service