Skip to main content

Therapeutic angiogenesis using stem cell-derived extracellular vesicles: an emerging approach for treatment of ischemic diseases

Abstract

Ischemic diseases, which are caused by a reduction of blood supply that results in reduced oxygen transfer and nutrient uptake, are becoming the leading cause of disabilities and deaths. Therapeutic angiogenesis is key for the treatment of these diseases. Stem cells have been used in animal models and clinical trials to treat various ischemic diseases. Recently, the efficacy of stem cell therapy has increasingly been attributed to exocrine functions, particularly extracellular vesicles. Extracellular vesicles are thought to act as intercellular communication vehicles to transport informational molecules including proteins, mRNA, microRNAs, DNA fragments, and lipids. Studies have demonstrated that extracellular vesicles promote angiogenesis in cellular experiments and animal models. Herein, recent reports on the use of extracellular vesicles for therapeutic angiogenesis during ischemic diseases are presented and discussed. We believe that extracellular vesicles-based therapeutics will be an ideal treatment method for patients with ischemic diseases.

Background

With the development of society and improvement of living standards, ischemic diseases have become a leading cause of disabilities and deaths in humans. Ischemic diseases are characterized by a reduction of blood supply with limited oxygen transfer and nutrient uptake. Thus, angiogenesis and blood supply reconstruction are key for treatment of ischemic diseases. Current clinical treatments primarily involve medical therapy (thrombolytic drugs and vasodilator drugs [1]) and surgery [2]. However, it remains difficult to achieve the purpose of vascular remodeling using either drugs [3] or surgery [4]. Inspired by the fact that the body undergoes natural angiogenesis in response to an insufficient blood supply, scientists have learned to enhance the efficiency of angiogenesis as a treatment strategy. The concept of therapeutic angiogenesis involves introducing an agent to promote the growth of new blood vessels in ischemic tissue. Stem cell therapy is a technology that has shown great prospects for ischemic diseases [5, 6]. Indeed, stem cells have been used in animal models and clinical trials to treat various ischemic diseases. However, as transplantation of stem cells continues to be limited by ethical issues, tumorigenicity, and immune rejection, such therapies are not widely available in the clinic.

Recent studies have found that stem cell supernatants can promote repair of damaged tissue [7, 8]. Accordingly, researchers began to focus on the exocrine function of stem cells. Extracellular vesicles (EVs), which are secreted from cells, have been discovered for more than 30 years. In the past, scientists have thought of EVs as cellular dust. Today, EVs are thought to be carriers of intercellular biological information, as they may contain nucleic acids, lipids, and proteins, thereby playing an indispensable role in cell-to-cell communication [9]. Furthermore, the composition of EVs varies according to their origin, and the information they carry also varies [10]. Biological characteristics and functions of EVs suggest their potential application for cell-free regeneration strategies, which may avoid the disadvantages of current stem cell transplantation techniques.

In particular, recent studies have reported that EVs accelerate angiogenesis in cellular experiments and animal models [10,11,12]. Here, we first summarize the characteristics and properties of EVs (Table 1) and then discuss the emerging role of stem cell-derived EVs in ischemic diseases, such as chronic wound, ischemic cardiomyopathy, and ischemic stroke. We believe that EV-based therapeutics will be an ideal option for patients who suffer from ischemic diseases.

Table 1 The main characteristics of extracellular vesicles

Biological characteristics of EVs

EVs have three distinct types including exosomes, microvesicles (MVs), and apoptotic bodies (ApoBDs) [13], as classified by their biogenesis and origin. Here, we mainly summarize the generation, composition, and isolation of EVs.

Generation of EVs (Fig. 1)

Exosomes, defined as 50–150-nm-sized vesicles, were found and named in 1987 [14]. The process of exosome generation can be summarized into three parts. First, the cytomembrane recesses inward to form early endosomes. Second, these early endosomes further develop into multi-vesicular endosomes (MVEs) in which intraluminal vesicles (ILVs) are formed by intraluminal budding. Finally, MVEs fuse with cell membranes to release ILVs as exosomes into the extracellular space, where they can be taken up by donor cells [9]. Released exosomes can travel to distant tissues to affect the behavior and biological function of target cells [15], which bind to the surface of exosomes through specific ligands. There are two ways in which exosomes enter target cells [16], namely cellular endocytosis and membrane fusion, whereby they release their cargoes. Unlike exosomes, MVs are in the range of 100–1000 nm in diameter [17] and are usually larger than exosomes. MVs bud from plasma membrane directly and then are released extracellularly under the condition of various stresses including irradiation, injury, and hypoxia [18]. Many studies have shown that exosomes and MVs are generated from healthy cells, while ApoBDs are mainly produced by dying cells or apoptotic cells [19]. The role of ApoBDs in intercellular communication is currently unclear. Researchers consider the primary functions of ApoBDs are self-cleaning of aging cells and intercellular immune regulation [20,21,22].

Fig. 1
figure 1

Generation and release of extracellular vesicles (EVs). a Healthy cells produce exosomes and MVs. Exosomes occur through three steps: cytomembrane recess inward to form early endosomes, intraluminal vesicle forming in multi-vesicular endosomes (MVEs) by intraluminal budding, and MVE fusing with cell membranes to release ILVs as exosomes. MVs bud outward directly from the plasma membrane. b Apoptotic cells produce ApoBDs. ApoBDs bud outward directly from the apoptotic membrane. ApoBDs are thought to be connected with self-cleaning of aging cells and intercellular immune regulation

Composition of EVs

Proteins

Proteins in EVs are mainly derived from plasma membrane, cytosol, Golgi, and nucleus [23, 24]. As more EV proteins are identified, it has been apparent that EVs contain a common set of EV proteins and cell-type-specific components. The common proteins include cytoskeletal proteins, heat-shock proteins, metabolic enzymes, annexins, ribosomal proteins, tetraspanins, vesicle trafficking-related proteins, and major histocompatibility complex (MHC). The purity of EV preparation is often demonstrated by protein markers enriched in EVs. In fact, tetraspanins including CD9, CD63, CD81, and CD82; heat-shock proteins (e.g., HSP70 and HSP90); MHC classes (I and II); Tsg101; 14-3-3 proteins; and the endosomal sorting complex required for transport (ESCRT-3) binding protein Alix have been regarded as “specific” exosomes markers for years. However, these proteins can also be detectable in ApoBDs and MVs [24, 25]. In addition, the types of cell-type-specific proteins are dependent on their parental cells and conditions under which the EVs are secreted. These proteins include immune-modulating proteins, cell-surface antigens, proteases, angiogenic and molecules [26].

Lipids

EVs are rich in lipids such as cholesterol, phosphatidylserine, diglyceride, phospholipid, phosphatidylcholine, phosphatidylinositol, polyglycerol, and phosphatidylethanolamine. Specifically, exosome plasma membranes contain a lot of cholesterol, sphingomyelin, ceramide, lipid rafts, and phosphatidylserine. MV and ApoBD membranes have high concentration of phosphatidylserine [27]. The stability of EV membrane is attributed in part to the lipid content of their membranes [28]. As a result of their high lipid content, EVs have the capacity to pass through biological barriers, escape phagocytosis by the reticuloendothelial system, and protect informational molecules contained within EVs [29]. Interestingly, lipids contained in EVs are somewhat different from other lipids present in their source cells, which might be affected by the micro-environment around EVs. For example, tumor micro-environments may lead to an enrichment of certain tumor progressive or immunosuppressive lipids, such as prostaglandins [30].

Nucleic acids

Besides proteins and lipids, EVs also incorporate coding RNA (mRNAs), non-coding RNAs (nc-RNAs), and DNA fragments [31,32,33]. According to nucleotide length, nc-RNAs are divided into long nc-RNAs (lncRNAs, longer than 200 nucleotides) [34] and small nc-RNAs (sncRNAs, smaller than 200 nucleotides) [35]. LncRNAs encompass the largest proportion of the non-coding transcriptome, but their functions are so far not well defined except for their role in tumor genesis [34]. SncRNAs in EVs include microRNAs (miRNAs), mitochondrial RNAs, piwi-RNA (pi-RNAs), small nuclear RNA, small nucleolar RNA (snoRNA), transfer RNA, Y-RNA, vault RNA, and small interfering RNA (siRNA) [36,37,38]. Among these sncRNAs, miRNAs [39], Y-RNAs [37, 40], pi-RNA [41], snoRNA [42], and siRNA [43] have been shown to mediate the therapeutic effect of EVs. MiRNAs in particular are the well-known group of sncRNAs and have already been studied extensively. In addition, DNA fragments in EVs such as ApoBDs may be related to cell apoptosis [19].

Isolation of EVs

The most common method for EVs isolation is differential centrifugation, which can separate similarly sized vesicle particles. Johnstone et al. originally developed differential centrifugation for the separation of EVs in reticulocyte tissue culture fluid [14]. Later, Théry et al. optimized and improved this method [15]. The first step involves centrifugation at 300×g, 2000×g, and 10,000×g to remove cells, dead cells, and cell debris, respectively. The second step is ultracentrifugation (> 100,000×g) to obtain a crude EV-rich extract. The third step repeats ultracentrifugation twice to remove contaminating proteins, which allows clear EVs to be obtained. Today, this method is widely used for various biological samples and considered a “gold standard” for isolating EVs. Advantages of this method are simple operation and production of a large number of EVs. However, the whole process is time-consuming and repeated centrifugation operations may damage the EVs. Thus, further improvement of this method is necessary.

EVs promote angiogenesis in chronic wound healing

Chronic wounds, which have the characteristics of complex pathogenesis, prolonged disease, easy recurrence, prolonged treatment time, high cost, and high disability rate, refer to wounds that cannot attain anatomical and functional wound healing standards after regular treatment for 4 weeks or more [44]. Most recalcitrant wounds result from pressure ulcers [45], diabetic ulcers [46], venous ulcers [47], vascular insufficiency (e.g., arteriosclerosis [48] or critical limb ischemia [49]), and trauma such as burns [50] and frostbite [51]. Difficulties associated with chronic wound healing have primarily been ascribed to a lack of angiogenesis [52]. Furthermore, without neovascularization, acute wounds will become chronic wounds [53]. Recently, EVs derived from many sources of stem cells have been reported as one of the most promising treatments for chronic wounds by promoting angiogenesis (Table 2).

Table 2 Extracellular vesicles derived from stem cells promote angiogenesis in chronic wound healing

BM-MSC-EVs

Bone marrow-mesenchymal stem cells (BM-MSCs) are a type of adult stem cells derived from the mesoderm, which mainly exist in bone marrow stroma to support hematopoiesis. Transplantation of BM-MSCs seeded in a collagen scaffold resulted in increased wound healing and enhanced angiogenesis [66]. Furthermore, when BM-MSCs were seeded directly onto the wound site and injected into the wound edges, increased dermal vascularity was observed in the wound [67]. Some studies have suggested that the paracrine functions of BM-MSCs elicit angiogenesis in the wound by activating vascular endothelial cells [68,69,70]. EVs as an important paracrine factor of BM-MSCs (BM-MSC-EVs) have been examined as potential BM-MSC-based therapies. Experiments showed that BM-MSC-EVs were internalized by human umbilical vein endothelial cells (HUVECs) and promoted endothelial angiogenesis in vitro [54]. This finding is in line with many reports demonstrating the angiogenic potential of BM-MSC-conditioned medium. Further research demonstrated that BM-MSC-EVs activated important signaling cascades including AKT, STAT3, and ERK in recipient cells. These pathways were probably responsible for increased transcription of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and transforming growth factor beta (TGF-β), which can improve endothelial neovascularization. In another study, CD63+ exosomes were isolated by flow cytometry of magnetic beads coated with anti-CD63+ antibodies. For the first time, CD63+ exosomes containing Wnt3a exteriorly were found to stimulate tube length formation in vitro by activating the canonical Wnt signaling pathway [55].

ADSC-EVs

In addition to their self-renewal ability and multi-directional differentiation potential, adipose-derived stem cells (ADSCs) are an abundant resource that can rapidly expand in vitro. Indeed, ADSCs have been shown to maintain both a stable phenotype and multipotent differentiation ability after in vitro culture for 40 generations or cryopreservation [71]. In vivo experiments have revealed that ADSC-derived therapies can significantly improve mean capillary count in chronic wounds [72, 73]. Moreover, ADSC-induced acceleration of VEGF levels in diabetic wounds reportedly regulates local angiogenesis [74]. Recently, Ren et al. [56] demonstrated that ADSC-derived MVs (ADSC-MVs) promoted tube formation of HUVECs seeding in a transwell system. Similarly, ASC-MVs could also increase the establishment efficiency of newly formed vessels and mature vessels in vivo. Moreover, after the treatment of ADSC-MVs, the expression of many growth factors and receptors including platelet-derived growth factor subunit A(PDGFA), vascular endothelial growth factor A(VEGFA), bFGF, hypoxia-inducible factor-1α (HIF-1α), VEGF receptor 2 (VEGFR2), and platelet-derived growth factor receptor (PDGFR) were significantly upregulated in HUVECs. Further study showed that ASC-MVs could increase the activation level of ERK and AKT signaling pathways in HUVECs, which may be responsible for the angiogenesis effect of ASC-MVs on endothelial cells. It was observed that loss of Nrf2/ARE activity increases oxidative stress, which can aggravate endothelial dysfunction and abnormal angiogenesis occurring in diabetes [75]. The researchers demonstrated that ADSC-derived exosomes (ADSC-Exo) overexpressing Nrf2 increased granulation tissue formation, promoted tube formation, and accelerated angiogenesis, suggesting that ADSC-Exo can potentially promote angiogenesis. Therefore, transplantation of exosomes may be suitable for clinical applications to treat diabetic foot ulcers. In addition to this study, other scientists reported that treatment of HUVECs with ADSC-Exo can promote the expression of proangiogenesis genes Angpt1 and Flk1 and inhibit the anti-angiogenesis gene Vash1, which improved wound healing [57]. Angpt1 and Flk1 are key for promoting tube formation, whereas Vash1 inhibits tube formation. Further study found that, by activating PKA pathway signaling, hypoxia-exposed ADSC-Exo promoted proangiogenesis gene expression, downregulated anti-angiogenic gene expression (Angpt1 and Flk1), and promoted tube formation (Vash1). In addition, in vivo experiments assessing vascular formation yielded similar results to in vitro cell models, suggesting that hypoxia-exposed exosomes can indeed enhance angiogenesis [58]. A large number of studies have reported that under conditions of hypoxia and inflammation, most cells can secrete VEGF, which can specifically act on vascular endothelial cells and promote blood vessel formation in vivo. As VEGF is known to activate the PKA signaling pathway [76], researchers examined the PKA signaling pathway in HUVECs after exosome treatment. They found that PKA signaling was activated, which further promoted endogenous VEGF expression in HUVECs and synergistically regulated the expression of downstream proangiogenic genes Angpt1 and Flk1, and decreased the anti-angiogenic gene Vash1, thus promoting angiogenesis. Therefore, this finding may represent a novel therapy for hypoxic-condition wounds, as well as the treatment of ischemic diseases with stem cell-derived products.

UCB-EVs, UC-MSC-EVs, and PMSC-EVs

Fetal appendage-derived MSCs are obtained from both maternal and fetal origins, such as umbilical cord blood MSCs (UCB-MSCs), umbilical cord MSCs (UC-MSCs), and placenta tissue MSCs (PMSCs). Fetal appendage-derived MSCs are an attractive source of transplantable stem cells for wound repair because they have no risk to donors, easy accessibility, and a low incidence of graft-versus-host disease [77, 78]. Many studies have shown that direct use of fetal-derived MSCs or their conditioned media can significantly increase neovascularization and promote chronic wound healing. MSCs derived from fetal appendages secrete proangiogenic molecules including VEGF, hepatocyte growth factor (HGF), bFGF, TGF-β, and insulin-like growth factor-1 [79,80,81,82,83]. Hu et al. first demonstrated that local transplantation of umbilical cord blood-derived exosomes (UCB-Exo) induced prominent regenerative effects in wound healing, mainly through new blood vessel formation [59]. Further experiments demonstrated that UCB-Exo accelerated cutaneous wound healing through miR-21-3p-mediated promotion of angiogenesis. Zhang et al. later demonstrated that human UC-MSC-Exo improved the tube-formation ability of endothelial cells in vitro and promoted angiogenesis in a cutaneous burn model in vivo [60]. Further exploration of the underlying mechanism revealed that extracted exosomes contained wnt4 protein, which can promote β-catenin nuclear transfer, activate the wnt/β-catenin pathway in skin, and inhibit E-cadherin expression, thus promoting the angiogenesis of skin. Furthermore, exosomes derived from PMSC-conditioned media (PMSC-Exo) also contained angiogenic factors, which enhanced endothelial tube formation. In vivo laser Doppler blood flow analysis showed that PMSC-Exo also enhanced angiogenesis in a murine ischemic injury model [61].

iPSC-EVs

Induced pluripotent stem cells (iPSCs) are generated by reprogramming somatic cells into pluripotent stem cells with the characteristics and functions of embryonic stem cells (ESCs). iPSCs are an abundant potential source of autologous or donor-matched cells for therapy and, therefore, have emerged as a promising alternative to ESCs for stem cell transplantation therapy. Scientists found that use of iPSCs or iPSC-derived fibroblasts with three-dimensional structure could improve wound healing [84, 85]. Recently, EVs derived from iPSCs (iPSC-EVs) were also used to treat diabetic wounds in mice [62]. Newly formed vessels and average vessel density in the exosomes derived from iPSCs (iPSC-Exo)-treated group at day 7 were significantly higher, suggesting that iPSC-Exo can improve diabetic chronic wounds by enhancing vessel density and number. In a rat skin full-thickness defect model, exosomes were found to promote wound blood vessel regeneration and maturation. In addition, iPSC-Exo were also reported to promote HUVEC tube formation in vitro [63]. Although some studies have shown that iPSC-Exo has a great effect on wound angiogenesis, underlying mechanisms have not been clearly explained, which provides direction for our future research.

EPC-EVs

Endothelial progenitor cells (EPCs) are mainly found in the bone marrow, umbilical cord blood, and peripheral blood. Recent studies indicate that EPCs can promote diabetic wound repair by facilitating neovascularization and the therapeutic effects of EPCs were attributed to a paracrine mechanism [86, 87]. Studies found that EVs derived from EPCs (EPC-EVs) accelerated the healing of diabetic skin wounds by promoting the regeneration of blood vessels. Experiments in vitro showed that EPC-Exo increased the proliferation and migration of vascular endothelial cells and accelerated expression of vascular-related factors such as VEGF and HIF-1α [64]. Experiments in vivo demonstrated that transplantation of EPC-Exo could accelerate skin wound healing in diabetic rats by positively modulating vascular endothelial cell function [65]. Further research showed that Erk1/2 signaling pathway was the critical mediator during the angiogenic responses of endothelial cells induced by EPC-Exo [88]. However, what EPC-Exo components are transferred into vascular endothelial cells remains unclear.

EVs promote angiogenesis in myocardial ischemia

With advancements in basic and clinical research of cardiovascular disease, the current clinical treatment of myocardial ischemia involves interventions such as percutaneous transluminal coronary angioplasty and coronary artery bypass grafting, but only for patients whose arteries larger than 2 mm in diameter. Patients with arteries less than 2 mm in diameter, certain diffuse coronary artery lesions, history of multiple surgeries, or lack of arteriovenous grafts are unsuitable for such revascularization techniques [89]. Increasingly, acute myocardial infarction (AMI) leads to acute coronary block and reperfusion injury, which can cause acute ischemia and hypoxia in cardiomyocytes. Myocardial necrosis and apoptosis ultimately lead to myocardial remodeling. Therefore, many studies hope stem cells can proliferate and differentiate into new cardiomyocytes to replace damaged myocardial tissue and improve cardiac function after AMI [90, 91]. However, recent studies have shown that survival and differentiation rates of stem cells in transplanted hearts are very low. Therefore, stem cell transplantation may elicit benefits mainly through paracrine effects [92]. At present, different cell-derived EV therapies for AMI are considered to be the most promising method for repairing damaged myocardium and promoting myocardial vessel regeneration (Table 3).

Table 3 Extracellular vesicles derived from stem cells promote angiogenesis in myocardial ischemia

CDC-EVs

Cardiosphere-derived cells (CDCs) are cardiac progenitor cells that can differentiate into the three major cardiac cell types: cardiomyocytes, endothelial cells, and smooth muscle cells [107]. Previous studies demonstrated that CDCs stimulate angiogenesis and functional improvement by indirect mechanisms in the infarcted myocardium [108, 109]. Treating CDCs with exosome biosynthesis inhibitor GW4869 abolished their cardioprotective and regenerative properties. Subsequent studies paid more attention to the role of EVs derived from CDCs (CDC-EVs) and found CDC-EVs have similar therapeutic effects of CDCs in the treatment of myocardial ischemia [93]. When CDC-Exo were injected into the infarct border zone after AMI, the scar was reduced and necrotic myocardium was repaired with neovascularization. This effect of CDC-Exo was confirmed by Gallet et al. who observed a higher number of arterioles in both infarct and border zones of exosomes derived from CDCs (CDC-Exo)-treated pigs [94]. Further study demonstrated that the function of CDC-Exo in neovascularization of ischemic myocardium was related to the high content of miR-146a. Experiments in vitro also showed that CDC-Exo increased HUVEC tube formation and promoted angiogenesis [95]. In addition, the contents of CDC-Exo can be changed under given conditions. For example, exosomes isolated from CDCs cultured under hypoxia were enriched with proangiogenic miRNAs such as miR-126, miR-130a, and miR-210, which increased tube formation of HUVECs [96]. For treatment of ischemic heart disease, CDCs which are derived from myocardial tissue have lower immune responses compared with other stem cells. Furthermore, allogeneic CDC-Exo did not induce significant immune responses after repeated dosing [110].

MSC-EVs

As a treatment for ischemia, EVs play an important role as key transporters of paracrine factors during angiogenesis [111]. For example, scientists observed that BM-MSC-EVs can be internalized by endothelial cells and enhanced HUVEC tube formation. Moreover, fluorescence micrographs showed a large number of functional tubes forming in regions surrounding infarction areas. Subsequent in vivo experiments observed increased blood vessel density in hearts injected with BM-MSC-EVs [97]. CXCR4 serves as a major regulator of stem/progenitor cell activities. CXCR4-enriched BM-MSC-Exo activates PI3K/Akt signaling pathway, leading to an increase of VEGF and cardiomyocyte survival under hypoxic conditions [98]. In vitro models, BM-MSC-Exo stimulated endothelial cell migration and vessel formation via ERK/Akt signaling. To determine the angiogenic effect of BM-MSC-Exo in vivo, exosomes were added to the Matrigel plug and then implanted subcutaneously. The results suggested the enhancement of the influx of vascular cells and the blood vessel formation in the Matrigel plug. Analysis of proangiogenic factors revealed the level of extracellular matrix metalloproteinase inducer (EMMPRIN) was high in BM-MSC-Exo. Knockdown of EMMPRIN leads to, both in vitro and in vivo, a diminished proangiogenic effect [99]. The exosomes were isolated from UC-MSC. In vitro, UC-MSC-Exo could promote migration of endothelial cells and tube formation, which might be associated with the increased expression of Bcl-2 family [100]. Kang et al. observed that MVs from ADSCs, especially from endothelial differentiation medium-preconditioned ADSCs, also enhanced angiogenesis both in vitro and in vivo, but the molecular mechanism was different. The level of miR-31 was found to be upregulated in preconditioned ADSCs. Further study showed miR-31 targeted factor-inhibiting hypoxia-inducible factor 1 (FIH1) in vascular endothelial cells to mediate the proangiogenic effect of MVs [101]. Another research assessed therapeutic properties of BM-MSCs, ADSCs, and endometrium-derived mesenchymal stem cells (EnMSCs) in a rat model of AMI and found that EnMSCs supported enhanced microvessel density. Analyses of exosomal microRNAs revealed miR-21 was the potential mediator of EnMSC therapy via the phosphatase and tensin homolog (PTEN)/Akt pathway [102].

ESC-EVs

ESCs have the ability to produce exosomes which are capable of instigating cell analogous response in target cells. In order to assess the therapeutic efficacy of ESC-derived exosome (ESC-Exo) in post-infarct myocardium, ESC-Exo were intramyocardially injected in mice at the time of AMI. After 4 weeks, immunohistochemical analysis showed the capillary density was remarkably increased in ESC-Exo transplanted hearts, but the underlying basis for the effect is unknown [103].

iPSC-EVs

In recent years, iPSC researches have offered exciting opportunities for tissue restoration. Scientists compared the angiogenesis ability of iPSCs with that of iPSC-EVs in heart failure. The results demonstrated that both iPSCs and iPSC-EVs significantly promoted the migration and tube formation of murine cardiac endothelial cells (CECs). Further experimental analysis of capillary density in vivo was performed in the infarct zone, border zone, and non-ischemic zone of infarcted mouse hearts respectively. IPSC-EV injection resulted in greater number of capillaries in the infarct zone compared with iPSC injection [104]. Another study observed the EVs from cardiovascular progenitor cells derived from iPSCs (iPSC-CPC-EVs) promoted the migration and tube formation of HUVECs. Moreover, iPSC-CPC-EVs could significantly improve chronic heart failure through decreasing left ventricular volumes and increasing left ventricular ejection fraction [105].

CD34+ cell-EVs

CD34 is selectively expressed on the surface of hematopoietic stem/progenitor cells and gradually weakens or even tends to disappear with the maturation of the cells. After intramyocardial injection, autologous CD34+ cells can enhance myocardial perfusion and function of patients with AMI by promoting angiogenesis [112]. Sahoo et al. investigated the mechanism of CD34+ cell-induced proangiogenic paracrine effects and found that the exosomes from CD34+ cells (CD34+-Exo) have the same effects on endothelial cell viability, proliferation and tube formation on Matrigel as CD34+ cells have [106]. Further study showed the therapeutic efficacy of CD34+ cell treatment could be increased by secretion of sonic hedgehog (Shh) and exosome-mediated delivery of Shh to AMI represents a major mechanism [113].

EVs promote angiogenesis in stroke

Stroke is a group of diseases characterized by cerebral ischemic and hemorrhagic injury, with ischemic stroke accounting for 60–80% of strokes [114]. Ischemia and hypoxia cause neuronal degeneration and necrosis, leading to irreversible damage in the ischemic core region [115]. Current effective therapies include the use of tissue plasminogen activator thrombolysis and intravascular thrombectomy. However, the time window for application of these treatments is only a few hours [116]. Moreover, most patients suffer from a certain degree of neurological dysfunction even after receiving effective thrombolytic therapy. Because of these limitations, more than 90% of ischemic strokes cannot be treated promptly and effectively. Therefore, how to reduce ischemic injury and promote the recovery of nerve function in ischemic areas has become a research hotspot. In recent years, a deeper understating of EVs has confirmed that the intercellular information exchange process regulated by EVs is widely involved in angiogenic processes of the cerebrovascular system [117] (Table 4).

Table 4 Extracellular vesicles derived from stem cells promote angiogenesis in stroke

MSCs isolated from various tissues can promote angiogenesis not only in wound healing, but also in stoke [120]. In trying to understand the exact molecular mechanism by which different sources of MSCs exert protective roles in ischemic stroke, many studies have investigated the proangiogenesis ability of EVs. BM-MSC-Exos were used to treat middle cerebral artery occlusion of adult male Wistar rats. The results demonstrated that endothelial cell proliferation, compared with the PBS-treated control group, was significantly increased and new capillary network was formed, suggesting that BM-MSC-Exos promote angiogenesis post stroke [12, 118]. Another research also found that ADSC-Exos which contained miRNA-181b-5p could enhance the tube length of brain microvascular endothelial cells (BMECs) after oxygen-glucose deprivation in vitro [119]. Direct targets of miR-181b-5p were further confirmed. Yang et al. found that the mRNA and protein levels of transient receptor potential melastatin 7 (TRPM7) were declined, and meanwhile, HIF-1α and VEGF were upregulated in BMECs after being cultured with 181b-Exos. These researches suggest that exosomes from stem cells may represent a novel therapeutic approach for stroke recovery.

EVs promote angiogenesis in other ischemic disease (Table 5)

Therapeutic effect of EVs in peripheral arterial disease

Peripheral arterial obstructive disease, caused by atherosclerotic occlusion of the leg arteries, is often accompanied by moderate to severe ischemic pain in limbs, which directly affects the quality of life of patients and imposes a huge economic burden on society and families [128]. Many researches have shown that stem cells such as MSCs and EPCs contribute to angiogenesis after hindlimb ischemia and EVs have been emerging as an important paracrine regulator for stem cells to exert positive therapeutic effects. iPSC-derived mesenchymal stem cells (iMSCs) own powerful therapeutic effects through a paracrine mechanism. Hu et al. reported that exosomes derived from iMSCs (iMSCs-Exo) have the ability to promote angiogenesis after transplantation into ischemic limbs of mice [121]. In another study, exosomes were isolated from human PMSCs cultured with a nitric oxide releasing polymer and revealed superior angiogenic effects on hind limb ischemia in a murine model. Further analysis indicated that enhanced VEGF and miR-126 expressions in exosomes were responsible for exosome promoting angiogenic processes [122]. MVs derived from EPC also contained miR-126 and miR-296 which are known to be angiogenetic, suggesting a role of RNAs transferred by MVs in EPC-derived MVs treatment of severe hindlimb ischemia of mice [123]. CD34+ stem cells have been demonstrated to improve perfusion and function of the ischemic limb of patients. CD34+-Exo can directly transfer miR-126-3p and mimic the angiogenic activity of their parent cells. MiR-126-3p suppresses the expression of SPRED1 and simultaneously regulates the expression of genes which are involved in angiogenic pathways to promote angiogenesis [124]. In addition, administration of BM-MSC-EVs enhanced the formation of new blood vessels in the ischemic limb. The research on mechanisms revealed the enriched presence of miR-210-3p and VEGF protein in BM-MSC-EV and the high levels of VEGFR1 and VEGFR2 in endothelial cells [125]. The miR-210-3p induces expression of several proangiogenic mRNAs (VEGF and VEGFR2) [129]. Therefore, all above researches indicate that angiogenesis-related miRNAs and proteins are the main components in EVs to exert their proangiogenesis function.

Table 5 Extracellular vesicles derived from stem cells promote angiogenesis in other ischemic diseases

Therapeutic effect of EVs in flap graft

Skin flap transplantation is the most widely used treatment in orthopedic surgery and the most effective treatment for ischemic tissue damage. Adequate blood supply is the basis for improving the survival rate of transplanted flaps. Skin flap transplantation has certain limitations in specific clinical applications, as ischemic necrosis occurs at the distal end of the flap [130]. How to safely and effectively improve the survival rate of transplant flaps and ensure their blood supply has always been a difficult problem for burn orthopedics. Therefore, promoting the angiogenesis of flap grafts is key to solving this problem. With a flap ischemia-reperfusion injury (IRI) model, the capability of ADSCs to protect tissue against IRI were examined. Treatment with ADSCs remarkably increased flap survival when compared with the control group and enhanced expression of proangiogenic genes [131]. Further study demonstrated that ADSC-CM and ADSC-Exo increased tube formation after injection into the flaps and interleukin 6 (IL-6) contained in ADSC-Exo stimulated angiogenesis and led to recovery after IRI [126]. A specific micro-environment can be used for in vitro ADSC culture to develop the customized EVs. Compared with ADSC-Exo and control groups, exosomes isolated from ADSC exposed to low concentration of H2O2 generated more cord-like structures on Matrigel in vitro and increased blood perfusion and microvascular density in the flap in vivo [127]. These results suggest that low H2O2 micro-environment facilitates the customized exosome development for cell-free therapeutic applications during skin flap transplantation.

Future directions and potential limitations (Table 6)

EVs have opened a new promising avenue for the treatment of ischemic diseases. Angiogenesis-related miRNAs and proteins in EVs derived from MSCs (Fig. 2) and other stem cells (Fig. 3) have shown potential to treat ischemic diseases by directly or indirectly activating angiogenesis-related signaling pathways in target cells. Based on recent researches, many miRNAs including miRNA-21-5p, miRNA-31, miRNA-125a, miRNA-126, miRNA-130a, miRNA-132, miRNA-146a, miRNA-181-5p, miRNA-210, and miRNA-296 are found to promote angiogenesis in ischemic disease [57, 93, 95, 101, 102, 106, 119, 122,123,124,125, 135, 139]. VEGF, as a major mediator of angiogenesis, is the most common functional protein component in EVs [125]. In view of the complex components in EVs, other specific functional proteins and miRNAs that play an important role in angiogenesis need to be further identified.

Table 6 The advantages and potential limitations of EV therapy
Fig. 2
figure 2

The mechanisms of angiogenesis induced by MSC-derived EVs in ischemic diseases. EVs from BM-MSCs, ADSCs, UC-MSCs, and PMSCs play an important role in neovascularization of ischemic diseases. MSC-derived EVs are enriched with specific cargo molecules including proteins (pSTAT3, IL-6, Wnt 3a, Wnt 4, and CXCR4) and miRNAs (miRNA-31, miRNA-125a, miRNA-181b, miRNA-210, miRNA-126, and miRNA-21). These proteins and miRNAs activate their related signal pathway to regulate the expression of angiogenic factors in endothelial cells. Abbreviation: IL-6, interleukin-6; FIH1, hypoxia-inducible factor 1-alpha inhibitor; HIF-1α, hypoxia-inducible factor-1α; VEGF, vascular endothelial growth factor; PTEN, phosphatase and tensin homolog

Fig. 3
figure 3

The mechanisms of angiogenesis induced by EVs derived from EPCs, CDCs, and CD34+ stem cells in ischemic diseases. EPC-derived EVs promote angiogenesis through upregulating the expression of related transcription factors. CDC-derived EVs are enriched with miR-210, miR-126, and miR-130a, which promote the expression of angiogenic proteins in endothelial cells. EVs derived from CD34+ stem cells transfer miR-126 and miR-130 into endothelial cells to stimulate angiogenesis. “?” represents uncertained functional cargo molecules in EVs. Abbreviation: PDGF, platelet-derived growth factor subunit; ANG-1, angiopoietin-1; VEGFA, vascular endothelial growth factor A; VEGF, vascular endothelial growth factor; bFGF, basic fibroblast growth factor; HIF-1α, hypoxia-inducible factor-1α

EVs produced by stem cells would be expected to have many advantages in the ischemic environment. First of all, EVs could transfer signals more effectively to target cells because their lipid bilayer shell can avert proteolytic degradation. Scientists are trying to harness the natural ability of EVs to transfer therapeutic payloads into the desired cells. For example, siRNA was effectively delivered by plasma exosomes into the target cells, leading to selective gene silencing of MAPK-1 [132]. Secondly, EVs contain many potential regulatory components such as miRNAs, mRNAs, and proteins. These informational molecules could function simultaneously to generate a strong effect on the characteristics of recipient cells. MiR-210-3p and VEGF protein, as the effective components of BM-MSC-EV, have the same function to promote new blood vessel formation of endothelial cells [125]. Finally, EVs can be applied to personalized medicine [133, 134]. Gene editing in stem cells can produce the desired EVs with specific cell-surface molecules. EVs from gene-edited patient-specific stem cells will hold potential for treatment of ischemic diseases of each individual patient. Furthermore, EVs from iPSC-derivatives can be used for an autologous therapy by activating endogenous repair. We believe that EVs generated from patient-specific iPSC-derivatives probably have a higher angiogenic effect and provide a safer way than stem cell transplantation because EVs used as cell-free therapy are not affected by the ischemic and hypoxic micro-environment and have no tumorigenic risk.

Numerous attempts to treat ischemic diseases with EVs have been made and the results are quite encouraging. However, there are many limitations remaining to be solved. Firstly, EVs transplanted into the damaged tissues may have only short-term effects owing to their short half-life and rapid clearance by the innate immune system. Takahashi et al. showed exosomes from murine melanoma cells disappeared very quickly with a half-time of approximately 2 min from the blood circulation [136]. So how to maintain the retention and stability of EVs over time in vivo is a main challenge in clinical application. Zhang et al. demonstrated that chitosan hydrogel remarkably increased the retention of exosomes in vivo and enhanced the stability of miRNAs and proteins in exosomes, enhancing angiogenesis in ischemic site [135]. Secondly, the efficiency of EV uptake needs to be improved. Cellular uptake of large number of EVs by target cells may improve the effects of angiogenesis. The efficiency of EVs uptake has been found to be related to intracellular and micro-environmental acidity [137]. Finally, the administration routes of EVs must be appropriately selected. Some studies explored whether the angiogenesis effects of EVs are influenced by intravascular injection or local injection of ischemic tissue. Results showed that topical injection of EVs made a better therapeutic effect, while intravascular injection caused EVs to degrade rapidly [138]. In conclusion, we believe that through continued and collaborative efforts, EV-based therapy will yield satisfactory responses in patients with ischemic diseases.

Abbreviations

ADSC-Exo:

Adipose-derived stem cells -derived exosomes

ADSC-MVs:

Adipose-derived stem cells derived microvesicles

ADSCs:

Adipose-derived stem cells

AMI:

Acute myocardial ischemia

ApoBDs:

Apoptotic bodies

bFGF:

Basic fibroblast growth factor

BMECs:

Brain microvascular endothelial cells

BM-MSC-EVs:

Bone marrow-mesenchymal stem cells- derived extracellular vesicles

BM-MSCs:

Bone marrow-mesenchymal stem cells

CD34+-Exo:

Exosomes from CD34+ cells

CDC-EVs:

Extracellular vesicles derived from cardiosphere-derived cells

CDC-Exo:

Cardiosphere-derived cells–derived exosomes

CDCs:

Cardiosphere-derived cells

CECs:

Murine cardiac endothelial cells

EMMPRIN:

Extracellular matrix metalloproteinase inducer

EnMSC:

Human endometrium-derived mesenchymal stem cells

EPC-EVs:

Extracellular vesicles derived from endothelial progenitor cells

EPCs:

Endothelial progenitor cells

ESC-Exo:

Embryonic stem cell-derived exosomes

ESCRT-3:

Endosomal sorting complex required for transport

ESCs:

Embryonic stem cells

EVs:

Extracellular vesicles

FIH1:

Factor-inhibiting hypoxia-inducible factor 1

HIF-1α:

Hypoxia-inducible factor-1α

HUVECs:

Human umbilical vein endothelial cells

IL-6:

Interleukin 6

ILVs:

Intraluminal vesicles

iMSCs:

Induced pluripotent stem cells-derived mesenchymal stem cells

iMSCs-Exo:

Exosomes-derived from induced pluripotent stem cells-derived mesenchymal stem cells

iPSC-EVs:

Extracellular vesicles derived from induced pluripotent stem cells

iPSC-Exo:

Exosomes derived from induced pluripotent stem cells

iPSCs:

Induced pluripotent stem cells

IRI:

Ischemia-reperfusion injury

LncRNAs:

Long nc-RNAs

MiRNAs:

MicroRNAs

MVEs:

Multi-vesicular endosomes

MVs:

Microvesicles

Nc-RNAs:

Non-coding RNAs

PDGFA:

Platelet-derived growth factor subunit A

PDGFR:

Platelet-derived growth factor receptor

Pi-RNAs:

Piwi-RNA

PMSC-Exo:

PMSCs derived exosomes

PMSCs:

Placenta tissue mesenchymal stem cells

Shh:

Sonic hedgehog

SiRNA:

Small interfering RNA

SncRNAs:

Small nc-RNAs

SnoRNA:

Small nucleolar RNA

TGF-β:

Transforming growth factor beta

TRPM7:

Transient receptor potential melastatin 7

UCB-Exo:

Umbilical cord blood-derived exosomes

UCB-MSCs:

Umbilical cord blood mesenchymal stem cells

UC-MSCs:

Umbilical cord mesenchymal stem cells

VEGF:

Vascular endothelial growth factor

VEGFA:

Vascular endothelial growth factor A

VEGFR2:

Vascular endothelial growth factor receptor 2

References

  1. Pande RL, et al. A pooled analysis of the durability and predictors of treatment response of cilostazol in patients with intermittent claudication. Vasc Med. 2010;15:181–8. https://doi.org/10.1177/1358863X10361545.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Mellière D, et al. The underestimated advantages of iliofemoral endarterectomy. Ann Vasc Surg. 2000;14:343–9.

    Article  PubMed  Google Scholar 

  3. Trams EG, et al. Exfoliation of membrane ecto-enzymes in the form of micro-vesicles. Biochim Biophys Acta. 1981;645:63–70.

    Article  CAS  PubMed  Google Scholar 

  4. Wu R, et al. Drug-eluting balloon versus standard percutaneous transluminal angioplasty in infrapopliteal arterial disease: a meta-analysis of randomized trials. Int J Surg. 2016;35:88–94. https://doi.org/10.1016/j.ijsu.2016.09.014.

    Article  PubMed  Google Scholar 

  5. Li S, et al. Advances in the treatment of ischemic diseases by mesenchymal stem cells. Stem Cells Int. 2016;2016:5896061. https://doi.org/10.1155/2016/5896061.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Choi M, et al. Proangiogenic features of Wharton's jelly-derived mesenchymal stromal/stem cells and their ability to form functional vessels. Int J Biochem Cell Biol. 2013;45:560–70. https://doi.org/10.1016/j.biocel.2012.12.001.

    Article  CAS  PubMed  Google Scholar 

  7. Fouraschen SM, et al. Secreted factors of human liver-derived mesenchymal stem cells promote liver regeneration early after partial hepatectomy. Stem Cells Dev. 2012;21:2410–9. https://doi.org/10.1089/scd.2011.0560.

    Article  CAS  PubMed  Google Scholar 

  8. Ionescu L, et al. Stem cell conditioned medium improves acute lung injury in mice: in vivo evidence for stem cell paracrine action. Am J Physiol Lung Cell Mol Physiol. 2012;303:L967–77. https://doi.org/10.1152/ajplung.00144.2011.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Panfoli I, et al. Microvesicles as promising biological tools for diagnosis and therapy. Expert Rev Proteomics. 2018;15:801–8. https://doi.org/10.1080/14789450.2018.1528149.

    Article  CAS  PubMed  Google Scholar 

  10. Muralidharan-Chari V, et al. Microvesicles: mediators of extracellular communication during cancer progression. J Cell Sci. 2010;123:1603–11. https://doi.org/10.1242/jcs.064386.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Amini A, et al. Stereological and molecular studies on the combined effects of T photobiomodulation and human bone marrow mesenchymal stem cell conditioned medium on wound healing in diabetic rats. J Photochem Photobiol B. 2018;182:42–51. https://doi.org/10.1016/j.jphotobiol.2018.03.010.

    Article  CAS  PubMed  Google Scholar 

  12. Doeppner TR, et al. Extracellular vesicles improve post-stroke neuroregeneration and prevent postischemic immunosuppression. Stem Cells Transl Med. 2015;4:1131–43. https://doi.org/10.5966/sctm.2015-0078.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Riazifar M, et al. Stem cell extracellular vesicles: extended messages of regeneration. Annu Rev Pharmacol Toxicol. 2017;6:125–54. https://doi.org/10.1146/annurev-pharmtox-061616-030146.

    Article  CAS  Google Scholar 

  14. Johnstone RM, et al. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem. 1987;262:9412–20.

    CAS  PubMed  Google Scholar 

  15. Théry C, et al. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol. 2006;30:3.22–9. https://doi.org/10.1002/0471143030.cb0322s30.

    Article  Google Scholar 

  16. Qin J, Xu Q. Functions and application of exosomes. Acta Pol Pharm. 2014;71:537–43.

    PubMed  Google Scholar 

  17. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200:373–83. https://doi.org/10.1083/jcb.201211138.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Akers JC, et al. Biogenesis of extracellular vesicles (EV): exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J Neuro-Oncol. 2013;113:1–11. https://doi.org/10.1007/s11060-013-1084-8.

    Article  Google Scholar 

  19. Atkin-Smith GK, et al. A novel mechanism of generating extracellular vesicles during apoptosis via a beads-on-a-string membrane structure. Nat Commun. 2015;6:7439. https://doi.org/10.1038/ncomms8439.

    Article  PubMed  Google Scholar 

  20. Ferguson TA, et al. Uptake of apoptotic antigen-coupled cells by lymphoid dendritic cells and cross-priming of CD8(+) T cells produce active immune unresponsiveness. J Immunol. 2002;168:5589–95.

    Article  CAS  PubMed  Google Scholar 

  21. Poon IK, et al. Apoptotic cell clearance: basic biology and therapeutic potential. Nat Rev Immunol. 2014;14:166–80. https://doi.org/10.1038/nri3607.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hochreiter-Hufford A, Ravichandran K. Clearing the dead: apoptotic cell sensing, recognition, engulfment, and digestion. Cold Spring Harb Perspect Biol. 2013;5:a008748. https://doi.org/10.1101/cshperspect.a008748.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Choi DS, et al. Proteomics, transcriptomics and lipidomics of exosomes and ectosomes. Proteomics. 2013;13:1554–71. https://doi.org/10.1002/pmic.201200329.

    Article  CAS  PubMed  Google Scholar 

  24. Choi DS, et al. Proteomics of extracellular vesicles: exosomes and ectosomes. Mass Spectrom Rev. 2015;34:474–90. https://doi.org/10.1002/mas.21420.

    Article  CAS  PubMed  Google Scholar 

  25. Tauro BJ, et al. Two distinct populations of exosomes are released from LIM1863 colon carcinoma cell-derived organoids. Mol Cell Proteomics. 2013;12:587–98. https://doi.org/10.1074/mcp.M112.021303.

    Article  CAS  PubMed  Google Scholar 

  26. Choi DS, et al. Proteomic analysis of microvesicles derived from human colorectal cancer cells. J Proteome Res. 2007;6:4646–55. https://doi.org/10.1021/pr070192y.

    Article  CAS  PubMed  Google Scholar 

  27. Safdar A, et al. The potential of endurance exercise-derived exosomes to treat metabolic diseases. Nat Rev Endocrinol. 2016;12:504–17. https://doi.org/10.1038/nrendo.2016.76.

    Article  CAS  PubMed  Google Scholar 

  28. Record M, et al. Exosomes as new vesicular lipid transporters involved in cell-cell communication and various pathophysiologies. Biochim Biophys Acta. 2014;1841:108–20. https://doi.org/10.1016/j.bbalip.2013.10.004.

    Article  CAS  PubMed  Google Scholar 

  29. Shahabipour F, et al. Exosomes as nanocarriers for siRNA delivery: paradigms and challenges. Arch Med Sci. 2016;12:1324–6. https://doi.org/10.5114/aoms.2016.62911.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Hannafon BN, Ding WQ. Intercellular communication by exosome-derived microRNAs in cancer. Int J Mol Sci. 2013;14:14240–69. https://doi.org/10.3390/ijms140714240.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ratajczak J, et al. Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication. Leukemia. 2006;20:1487–95. https://doi.org/10.1038/sj.leu.2404296.

    Article  CAS  PubMed  Google Scholar 

  32. Guescini M, et al. Astrocytes and glioblastoma cells release exosomes carrying mtDNA. J Neural Transm (Vienna). 2010;117:1–4. https://doi.org/10.1007/s00702-009-0288-8.

    Article  CAS  Google Scholar 

  33. Valadi H, et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–9. https://doi.org/10.1038/ncb1596.

    Article  CAS  PubMed  Google Scholar 

  34. Xiong XD, et al. Long non-coding RNAs: an emerging powerhouse in the battle between life and death of tumor cells. Drug Resist Updat. 2016;26:28–42. https://doi.org/10.1016/j.drup.2016.04.001.

    Article  PubMed  Google Scholar 

  35. Ha M, Kim V. Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol. 2014;15:509–24. https://doi.org/10.1038/nrm3838.

    Article  CAS  PubMed  Google Scholar 

  36. Bellingham SA, et al. Small RNA deep sequencing reveals a distinct miRNA signature released in exosomes from prion-infected neuronal cells. Nucleic Acids Res. 2012;40:10937–49. https://doi.org/10.1093/nar/gks832.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Nolte-'t Hoen EN, et al. Deep sequencing of RNA from immune cell-derived vesicles uncovers the selective incorporation of small non-coding RNA biotypes with potential regulatory functions. Nucleic Acids Res. 2012;40:9272–85. https://doi.org/10.1093/nar/gks658.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ogawa Y, et al. Small RNA transcriptomes of two types of exosomes in human whole saliva determined by next generation sequencing. Biol Pharm Bull. 2013;36:66–75.

    Article  CAS  PubMed  Google Scholar 

  39. Gandellini P, et al. microRNAs as players and signals in the metastatic cascade: implications for the development of novel anti-metastatic therapies. Semin Cancer Biol. 2017;44:132–40. https://doi.org/10.1016/j.semcancer.2017.03.005.

    Article  CAS  PubMed  Google Scholar 

  40. Tosar JP, et al. Assessment of small RNA sorting into different extracellular fractions revealed by high-throughput sequencing of breast cell lines. Nucleic Acids Res. 2015;43. https://doi.org/10.1093/nar/gkv432.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Li B, et al. piRNA-823 delivered by multiple myeloma-derived extracellular vesicles promoted tumorigenesis through re-educating endothelial cells in the tumor environment. Oncogene. 2019. https://doi.org/10.1038/s41388-019-0788-4.

  42. Rimer JM, et al. Long-range function of secreted small nucleolar RNAs that direct 2′-O-methylation. J Biol Chem. 2018;293:13284–96. https://doi.org/10.1074/jbc.RA118.003410.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Pi F, et al. Nanoparticle orientation to control RNA loading and ligand display on extracellular vesicles for cancer regression. Nat Nanotechnol. 2018;13:82–9. https://doi.org/10.1038/s41565-017-0012-z.

    Article  CAS  PubMed  Google Scholar 

  44. Clinton A, Carter T. Chronic wound biofilms: pathogenesis and potential therapies. Lab Med. 2015;46:277–84. https://doi.org/10.1309/LMBNSWKUI4JPN7SO.

    Article  PubMed  Google Scholar 

  45. Bhate K, Williams HC. What's new in acne? An analysis of systematic reviews published in 2011-2012. Clin Exp Dermatol. 2014;39:273–7. https://doi.org/10.1111/ced.12270.

    Article  CAS  PubMed  Google Scholar 

  46. Lefrancois T, et al. Evidence based review of literature on detriments to healing of diabetic foot ulcers. Foot Ankle Surg. 2017;23:215–24. https://doi.org/10.1016/j.fas.2016.04.002.

    Article  PubMed  Google Scholar 

  47. Crawford JM, et al. Pathophysiology of venous ulceration. 9J Vasc Surg Venous Lymphat Disord. 2017;5:596–605. https://doi.org/10.1016/j.jvsv.2017.03.015.

    Article  Google Scholar 

  48. Lusis AJ. Atherosclerosis. Nature. 2000;407:233–41. https://doi.org/10.1038/35025203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Uccioli L, et al. Critical limb ischemia: current challenges and future prospects. Vasc Health Risk Manag. 2018;26:63–74. https://doi.org/10.2147/VHRM.S125065.

    Article  Google Scholar 

  50. Veeravagu A, et al. National trends in burn and inhalation injury in burn patients: results of analysis of the nationwide inpatient sample database. J Burn Care Res. 2015;36:258–65. https://doi.org/10.1097/BCR.0000000000000064.

    Article  PubMed  Google Scholar 

  51. Zafren K. Frostbite: prevention and initial management. High Alt Med Biol. 2013;14:9–12. https://doi.org/10.1089/ham.2012.1114.

    Article  PubMed  Google Scholar 

  52. Pazyar N, et al. Skin wound healing and phytomedicine: a review. Skin Pharmacol Physiol. 2014;27:303–10. https://doi.org/10.1159/000357477.

    Article  CAS  PubMed  Google Scholar 

  53. Tonnesen MG, et al. Angiogenesis in wound healing. J Investig Dermatol Symp Proc. 2000;5:40–6. https://doi.org/10.1046/j.1087-0024.2000.00014.x.

    Article  CAS  PubMed  Google Scholar 

  54. Shabbir A, et al. Mesenchymal stem cell exosomes induce proliferation and migration of normal and chronic wound fibroblasts, and enhance angiogenesis in vitro. Stem Cell Dev. 2015;24:1635–47.

    Article  CAS  Google Scholar 

  55. McBride JD, et al. Bone marrow mesenchymal stem cell-derived CD63+ exosomes transport Wnt3a exteriorly and enhance dermal fibroblast proliferation, migration and angiogenesis in vitro. Stem Cells Dev. 2017;26:1384–98. https://doi.org/10.1089/scd.2017.0087.

    Article  CAS  PubMed  Google Scholar 

  56. Ren S, et al. Microvesicles from human adipose stem cells promote wound healing by optimizing cellular functions via AKT and ERK signaling pathways. Stem Cell Res Ther. 2019;10:47. https://doi.org/10.1186/s13287-019-1152-x.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Liang X, et al. Exosomes secreted by mesenchymal stem cells promote endothelial cell angiogenesis by transferring miR-125a. J Cell Sci. 2016;129:2182–9. https://doi.org/10.1242/jcs.170373.

    Article  CAS  PubMed  Google Scholar 

  58. Xue C, et al. Exosomes derived from hypoxia-treated human adipose mesenchymal stem cells enhance angiogenesis through the PKA signaling pathway. Stem Cells Dev. 2018;27:456–65. https://doi.org/10.1089/scd.2017.0296.

    Article  CAS  PubMed  Google Scholar 

  59. Hu Y, et al. Exosomes from human umbilical cord blood accelerate cutaneous wound healing through miR-21-3p-mediated promotion of angiogenesis and fibroblast function. Theranostics. 2018;8:169–84. https://doi.org/10.7150/thno.21234.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Zhang B, et al. Human umbilical cord mesenchymal stem cell exosomes enhance angiogenesis through the Wnt4/β-catenin pathway. Stem Cells Transl Med. 2015;4:513–22. https://doi.org/10.5966/sctm.2014-0267.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Komaki M, et al. Exosomes of human placenta-derived mesenchymal stem cells stimulate angiogenesis. Stem Cell Res Ther. 2017;8:219. https://doi.org/10.1186/s13287-017-0660-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kobayashi H, et al. Effects of exosomes derived from the induced pluripotent stem cells on skin wound healing. Nagoya J Med Sci. 2018;80:141–53. https://doi.org/10.18999/nagjms.80.2.141.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Zhang JY, et al. 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. https://doi.org/10.1186/s12967-015-0417-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Li X, et al. Exosomes derived from endothelial progenitor cells attenuate vascular repair and accelerate reendothelialization by enhancing endothelial function. Cytotherapy. 2016;18:253–62. https://doi.org/10.1016/j.jcyt.2015.11.009.

    Article  CAS  PubMed  Google Scholar 

  65. Li X, et al. Human endothelial progenitor cells-derived exosomes accelerate cutaneous wound healing in diabetic rats by promoting endothelial function. J Diabetes Complicat. 2016;30:986–92. https://doi.org/10.1016/j.jdiacomp.2016.05.009.

    Article  Google Scholar 

  66. O'Loughlin A, et al. Topical administration of allogeneic mesenchymal stromal cells seeded in a collagen scaffold augments wound healing and increases angiogenesis in the diabetic rabbit ulcer. Diabetes. 2013;62:2588–94. https://doi.org/10.2337/db12-1822.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Vojtassák J, et al. Autologous biograft and mesenchymal stem cells in treatment of the diabetic foot. Neuro Endocrinol Lett. 2006;27:134–7.

    PubMed  Google Scholar 

  68. Shen L, et al. Neurotrophin-3 accelerates wound healing in diabetic mice by promoting a paracrine response in mesenchymal stem cells. Cell Transplant. 2013;22:1011–21. https://doi.org/10.3727/096368912X657495.

    Article  PubMed  Google Scholar 

  69. Kim CH, et al. Mesenchymal stem cells improve wound healing in vivo via early activation of matrix metalloproteinase-9 and vascular endothelial growth factor. J Korean Med Sci. 2011;26:726–33. https://doi.org/10.3346/jkms.2011.26.6.726.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Chen L, et al. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS One. 2008;3:e1886. https://doi.org/10.1371/journal.pone.0001886.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Choudhery MS, et al. Comparison of human mesenchymal stem cells derived from dental pulp, bone marrow, adipose tissue, and umbilical cord tissue by gene expression. Cytotherapy. 2013;15:330–43. https://doi.org/10.1016/j.jcyt.2012.11.010.

    Article  CAS  PubMed  Google Scholar 

  72. Kim SM, et al. The effect of diabetes on the wound healing potential of adipose-tissue derived stem cells. Int Wound J. 2016;13:33–41. https://doi.org/10.1111/iwj.12540.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Cramer C, et al. Persistent high glucose concentrations alter the regenerative potential of mesenchymal stem cells. Stem Cells Dev. 2010;19:1875–84. https://doi.org/10.1089/scd.2010.0009.

    Article  CAS  PubMed  Google Scholar 

  74. Kim EK, et al. The effect of human adipose-derived stem cells on healing of ischemic wounds in a diabetic nude mouse model. Plast Reconstr Surg. 2011;128:387–94. https://doi.org/10.1097/PRS.0b013e31821e6de2.

    Article  CAS  PubMed  Google Scholar 

  75. Kensler TW, et al. Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu Rev Pharmacol Toxicol. 2007;47:89–116. https://doi.org/10.1146/annurev.pharmtox.46.120604.141046.

    Article  CAS  PubMed  Google Scholar 

  76. Spirli C, et al. ERK1/2-dependent vascular endothelial growth factor signaling sustains cyst growth in polycystin-2 defective mice. Gastroenterology. 2010;138:360–71. https://doi.org/10.1053/j.gastro.2009.09.005.

    Article  CAS  PubMed  Google Scholar 

  77. Ballen KK, et al. Umbilical cord blood transplantation: the first 25 years and beyond. Blood. 2013;122:491–8. https://doi.org/10.1182/blood-2013-02-453175.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Doi H, et al. Potency of umbilical cord blood- and Wharton's jelly-derived mesenchymal stem cells for scarless wound healing. Sci Rep. 2016;6:18844. https://doi.org/10.1038/srep18844.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Lee C, et al. Human umbilical cord blood-derived mesenchymal stromal cells and small intestinal submucosa hydrogel composite promotes combined radiation-wound healing of mice. Cytotherapy. 2017;19:1048–59. https://doi.org/10.1016/j.jcyt.2017.06.007.

    Article  CAS  PubMed  Google Scholar 

  80. Kong P, et al. Placenta mesenchymal stem cell accelerates wound healing by enhancing angiogenesis in diabetic Goto-Kakizaki (GK) rats. Biochem Biophys Res Commun. 2013;438:410–9. https://doi.org/10.1016/j.bbrc.2013.07.088.

    Article  CAS  PubMed  Google Scholar 

  81. Liu L, et al. Human umbilical cord mesenchymal stem cells transplantation promotes cutaneous wound healing of severe burned rats. PLoS One. 2014;9:e88348. https://doi.org/10.1371/journal.pone.0088348.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Wang S, et al. Wound dressing model of human umbilical cord mesenchymal stem cells-alginates complex promotes skin wound healing by paracrine signaling. Stem Cells Int. 2016;2016:3269267. https://doi.org/10.1155/2016/3269267.

    Article  CAS  PubMed  Google Scholar 

  83. Liu Z, et al. Human umbilical cord mesenchymal stem cells improve irradiation-induced skin ulcers healing of rat models. Biomed Pharmacother. 2018;101:729–36. https://doi.org/10.1016/j.biopha.2018.02.093.

    Article  CAS  PubMed  Google Scholar 

  84. Kashpur O, et al. Differentiation of diabetic foot ulcer-derived induced pluripotent stem cells reveals distinct cellular and tissue phenotypes. FASEB J. 2018;33:1262–77. https://doi.org/10.1096/fj.201801059.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Martin PE, et al. The potential of human induced pluripotent stem cells for modelling diabetic wound healing in vitro. Clin Sci (Lond). 2018;132:1629–43. https://doi.org/10.1042/CS20171483.

    Article  CAS  Google Scholar 

  86. Kanzler I, et al. Differential roles of angiogenic chemokines in endothelial progenitor cell-induced angiogenesis. Basic Res Cardiol. 2013;108:310. https://doi.org/10.1007/s00395-012-0310-4.

    Article  CAS  PubMed  Google Scholar 

  87. Basile DP, Yoder MC. Circulating and tissue resident endothelial progenitor cells. J Cell Physiol. 2014;229:10–6. https://doi.org/10.1002/jcp.24423.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Zhang J, et al. Exosomes derived from human endothelial progenitor cells accelerate cutaneous wound healing by promoting angiogenesis through Erk1/2 signaling. Int J Biol Sci. 2016;12:1472–87. https://doi.org/10.7150/ijbs.15514.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Ertl G, Frantz S. Healing after myocardial infarction. Cardiovasc Res. 2005;66:22–32. https://doi.org/10.1016/j.cardiores.2005.01.011.

    Article  CAS  PubMed  Google Scholar 

  90. Kanashiro-Takeuchi RM, et al. Pharmacologic and genetic strategies to enhance cell therapy for cardiac regeneration. J Mol Cell Cardiol. 2011;51:619–25. https://doi.org/10.1016/j.yjmcc.2011.05.015.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Iglesias-García O, et al. Induced pluripotent stem cells as a new strategy for cardiac regeneration and disease modeling. J Mol Cell Cardiol. 2013;62:43–50. https://doi.org/10.1016/j.yjmcc.2013.04.022.

    Article  CAS  PubMed  Google Scholar 

  92. Makridakis M, et al. Stem cells: insights into the secretome. Biochim Biophys Acta. 2013;1834:2380–4. https://doi.org/10.1016/j.bbapap.2013.01.032.

    Article  CAS  PubMed  Google Scholar 

  93. Ibrahim AG, et al. Exosomes as critical agents of cardiac regeneration triggered by cell therapy. Stem Cell Reports. 2014;2:606–19. https://doi.org/10.1016/j.stemcr.2014.04.006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Lang JK, et al. Inhibiting extracellular vesicle release from human cardiosphere derived cells with lentiviral knockdown of nSMase2 differentially effects proliferation and apoptosis in cardiomyocytes, fibroblasts and endothelial cells in vitro. PLoS One. 2016;11:e0165926. https://doi.org/10.1371/journal.pone.0165926.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Namazi H, et al. Exosomes secreted by hypoxic cardiosphere-derived cells enhance tube formation and increase pro-angiogenic miRNA. J Cell Biochem. 2018;119:4150–60. https://doi.org/10.1002/jcb.26621.

    Article  CAS  PubMed  Google Scholar 

  96. Gallet R, et al. Exosomes secreted by cardiosphere-derived cells reduce scarring, attenuate adverse remodelling, and improve function in acute and chronic porcine myocardial infarction. Eur Heart J. 2017;38. https://doi.org/10.1093/eurheartj/ehw240.

  97. Teng XM, et al. Mesenchymal stem cell-derived exosomes improve the microenvironment of infarcted myocardium contributing to angiogenesis and anti-inflammation. Cell Physiol Biochem. 2015;37:2415–24. https://doi.org/10.1159/000438594.

    Article  CAS  PubMed  Google Scholar 

  98. Kang K, et al. Exosomes secreted from CXCR4 overexpressing mesenchymal stem cells promote cardioprotection via Akt signaling pathway following myocardial infarction. Stem Cells Int. 2015;2015:659890. https://doi.org/10.1155/2015/659890.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Vrijsen KR, et al. Exosomes from cardiomyocyte progenitor cells and mesenchymal stem cells stimulate angiogenesis via EMMPRIN. Adv Healthc Mater. 2016;5:2555–65. https://doi.org/10.1002/adhm.201600308.

    Article  CAS  PubMed  Google Scholar 

  100. Zhao YY, et al. Exosomes derived from human umbilical cord mesenchymal stem cells relieve acute myocardial ischemic injury. Stem Cells Int. 2015;2015:761643. https://doi.org/10.1155/2015/761643.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Kang T, et al. Adipose-derived stem cells induce angiogenesis via microvesicle transport of miRNA-31. Stem Cells Transl Med. 2016;5:440–50. https://doi.org/10.5966/sctm.2015-0177.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Wang K, et al. Enhanced cardioprotection by human endometrium mesenchymal stem cells driven by exosomal microRNA-21. Stem Cells Transl Med. 2017;6:209–22. https://doi.org/10.5966/sctm.2015-0386.

    Article  CAS  PubMed  Google Scholar 

  103. Khan M, et al. Embryonic stem cell-derived exosomes promote endogenous repair mechanisms and enhance cardiac function following myocardial infarction. Circ Res. 2015;117:52–64. https://doi.org/10.1161/CIRCRESAHA.117.305990.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Adamiak M, et al. Induced pluripotent stem cell (iPSC)-derived extracellular vesiclesare safer and more effective for cardiac repair than iPSCs. Circ Res. 2018;122:296–309. https://doi.org/10.1161/CIRCRESAHA.117.311769.

    Article  CAS  PubMed  Google Scholar 

  105. El Harane N, et al. Acellular therapeutic approach for heart failure: in vitro production of extracellular vesicles from human cardiovascular progenitors. Eur Heart J. 2018;39:1835–47. https://doi.org/10.1093/eurheartj/ehy012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Sahoo S, et al. Exosomes from human CD34(+) stem cells mediate their proangiogenic paracrine activity. Circ Res. 2011;109:724–8. https://doi.org/10.1161/CIRCRESAHA.111.253286.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Marbán E. Breakthroughs in cell therapy for heart disease: focus on cardiosphere-derived cells. Mayo Clin Proc. 2014;89:850–8. https://doi.org/10.1016/j.mayocp.2014.02.014.

    Article  PubMed  Google Scholar 

  108. Smith RR, et al. Regenerative potential of cardiosphere-derived cells expanded from percutaneous endomyocardial biopsy specimens. Circulation. 2007;115. https://doi.org/10.1161/CIRCULATIONAHA.106.655209.

    Article  PubMed  Google Scholar 

  109. Kreke M, et al. Cardiospheres and cardiosphere-derived cells as therapeutic agents following myocardial infarction. Expert Rev Cardiovasc Ther. 2012;10:1185–94. https://doi.org/10.1586/erc.12.102.

    Article  CAS  PubMed  Google Scholar 

  110. Mirotsou M, et al. Repeated doses of cardiosphere-derived cell extracellular vesicles are hypo-immunogenic. Proceedings of the Abstracts from the 4th International Meeting of ISEV (ISEV '15); April 2015; Washington, DC, USA.

  111. Lai RC, et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010;4:214–22. https://doi.org/10.1016/j.scr.2009.12.003.

    Article  CAS  PubMed  Google Scholar 

  112. Kawamoto A, et al. CD34-positive cells exhibit increased potency and safety for therapeutic neovascularization after myocardial infarction compared with total mononuclear cells. Circulation. 2006;114:2163–9. https://doi.org/10.1161/CIRCULATIONAHA.106.644518.

    Article  PubMed  Google Scholar 

  113. Mackie AR, et al. Sonic hedgehog - modified human CD34+ cells preserve cardiac function following acute myocardial infarction. Circ Res. 2012;111:312–21. https://doi.org/10.1161/CIRCRESAHA.112.266015.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Navaratna D, et al. Mechanisms and targets for angiogenic therapy after stroke. Clin Lung Cancer. 2009;3:216–23. https://doi.org/10.3816/CLC.2009.n.006.

    Article  CAS  Google Scholar 

  115. Sharp FR, et al. Multiple molecular penumbras after focal cerebral ischemia. J Cereb Blood Flow Metab. 2000;20:1011–32. https://doi.org/10.1097/00004647-200007000-00001.

    Article  CAS  PubMed  Google Scholar 

  116. Hack W, et al. Thrombolysis with alteplase 3 to 4.5 hours after acute ischemic stroke. N Engl J Med. 2008;359:1317–29. https://doi.org/10.1056/NEJMoa0804656.

    Article  Google Scholar 

  117. Xian X, et al. Exosomes with highly angiogenic potential for possible use in pulp regeneration. J Endod. 2018;44:751–8. https://doi.org/10.1016/j.joen.2017.12.024.

    Article  PubMed  Google Scholar 

  118. Xin H, et al. Systemic administration of exosomes released from mesenchymal stromal cells promote functional recovery and neurovascular plasticity after stroke in rats. J Cereb Blood Flow Metab. 2013;33:1711–5. https://doi.org/10.1038/jcbfm.2013.152.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Yang Y, et al. Exosomes secreted by adipose-derived stem cells contribute to angiogenesis of brain microvascular endothelial cells following oxygen–glucose deprivation in vitro through microRNA-181b/TRPM7 axis. J Mol Neurosci. 2018;65:74–83. https://doi.org/10.1007/s12031-018-1071-9.

    Article  CAS  PubMed  Google Scholar 

  120. Li Y, Chopp M. Marrow stromal cell transplantation in stroke and traumatic brain injury. Neurosci Lett. 2009;456:120–3. https://doi.org/10.1016/j.neulet.2008.03.096.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Hu GW, et al. Exosomes secreted by human-induced pluripotent stem cell-derived mesenchymal stem cells attenuate limb ischemia by promoting angiogenesis in mice. Stem Cell Res Ther. 2015;6:10. https://doi.org/10.1186/scrt546.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Du W, et al. Enhanced proangiogenic potential of mesenchymal stem cell-derived exosomes stimulated by a nitric oxide releasing polymer. Biomaterials. 2017;133:70–81. https://doi.org/10.1016/j.biomaterials.2017.04.030.

    Article  CAS  PubMed  Google Scholar 

  123. Ranghino A, et al. Endothelial progenitor cell-derived microvesicles improve neovascularization in a murine model of hindlimb ischemia. Int J Immunopathol Pharmacol. 2012;25:75–85.

    Article  CAS  PubMed  Google Scholar 

  124. Mathiyalagan P, et al. Angiogenic mechanisms of human CD34+ stem cell exosomes in the repair of Iischemic hindlimb. Circ Res. 2017;120:1466–76. https://doi.org/10.1161/CIRCRESAHA.116.310557.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Gangadaran P, et al. Extracellular vesicles from mesenchymal stem cells activates VEGF receptors and accelerates recovery of hindlimb ischemia. J Control Release. 2017;264:112–26. https://doi.org/10.1016/j.jconrel.2017.08.022.

    Article  CAS  PubMed  Google Scholar 

  126. Pu CM, et al. Adipose-derived stem cells protect skin flaps against ischemia/reperfusion injury via interleukin-6 expression. J Invest Dermatol. 2017;137:1353–62. https://doi.org/10.1016/j.jid.2016.12.030.

    Article  CAS  PubMed  Google Scholar 

  127. Bai Y, et al. Adipose mesenchymal stem cell-derived exosomes stimulated by hydrogen peroxide enhanced skin flap recovery in ischemia-reperfusion injury. Biochem Biophys Res Commun. 2018;500:310–7. https://doi.org/10.1016/j.bbrc.2018.04.065.

    Article  CAS  PubMed  Google Scholar 

  128. Chen JC, et al. Risks of peripheral arterial occlusive disease in patients with obstructive sleep apnoea: a population-based case-control study. Clin Otolaryngol. 2015;40:437–42. https://doi.org/10.1111/coa.12393.

    Article  PubMed  Google Scholar 

  129. Liu F, et al. Upregulation of microRNA-210 regulates renal angiogenesis mediated by activation of VEGF signaling pathway under ischemia/perfusion injury in vivo and in vitro. Kidney Blood Press Res. 2012;35:182–91. https://doi.org/10.1159/000331054.

    Article  CAS  PubMed  Google Scholar 

  130. Akhavani MA, et al. Angiogenesis and plastic surgery. J Plast Reconstr Aesthet Surg. 2008;61:1425–37. https://doi.org/10.1016/j.bjps.2008.05.041.

    Article  PubMed  Google Scholar 

  131. Reichenberger MA, et al. Adipose derived stem cells protect skin flaps against ischemia-reperfusion injury. Stem Cell Rev. 2012;8:854–62. https://doi.org/10.1007/s12015-012-9368-5.

    Article  CAS  Google Scholar 

  132. Wahlgren J, et al. Plasma exosomes can deliver exogenous short interfering RNA to monocytes and lymphocytes. Nucleic Acids Res. 2012;40:e130. https://doi.org/10.1093/nar/gks463.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Contreras-Naranjo JC, et al. Microfluidics for exosome isolation and analysis: enabling liquid biopsy for personalized medicine. Lab Chip. 2017;17:3558–77. https://doi.org/10.1039/c7lc00592j.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Srivastava A, et al. Exploitation of exosomes as nanocarriers for gene-, chemo-, and immune-therapy of cancer. J Biomed Nanotechnol. 2016;12:1159–73.

    Article  CAS  PubMed  Google Scholar 

  135. Zhang K, et al. Enhanced therapeutic effects of mesenchymal stem cell-derived exosomes with an injectable hydrogel for hindlimb ischemia treatment. ACS Appl Mater Interfaces. 2018;10:30081–91. https://doi.org/10.1021/acsami.8b08449.

    Article  CAS  PubMed  Google Scholar 

  136. Takahashi Y, et al. Visualization and in vivo tracking of the exosomes of murine melanoma B16-BL6 cells in mice after intravenous injection. J Biotechnol. 2013;165:77–84. https://doi.org/10.1016/j.jbiotec.2013.03.013.

    Article  CAS  PubMed  Google Scholar 

  137. Tian T, et al. Visualizing of the cellular uptake and intracellular trafficking of exosomes by live-cell microscopy. J Cell Biochem. 2010;111:488–96. https://doi.org/10.1002/jcb.22733.

    Article  CAS  PubMed  Google Scholar 

  138. Arderiu G, et al. Tissue factor regulates microvessel formation and stabilization by induction of chemokine (C-C motif) ligand 2 expression. Arterioscler Thromb Vasc Biol. 2011;31:2607–15. https://doi.org/10.1161/ATVBAHA.111.233536.

    Article  CAS  PubMed  Google Scholar 

  139. Barile L, et al. Extracellular vesicles from human cardiac progenitor cells inhibit cardiomyocyte apoptosis and improve cardiac function after myocardial infarction. Cardiovasc Res. 2014;103:530–41.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported in part by the National Nature Science Foundation of China (81830064, 81721092, 81571905), the National Key Research Development Plan (2017YFC1103300), and the Military Logistics Research Key Project (AWS17J005).

Availability of data and materials

All data and material are included in this published article.

Author information

Authors and Affiliations

Authors

Contributions

BX and ZC conceived the manuscript. BX and MK wrote the manuscript and designed the figures. FX and ZC revised and edited the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Cuiping Zhang or Xiaobing Fu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bian, X., Ma, K., Zhang, C. et al. Therapeutic angiogenesis using stem cell-derived extracellular vesicles: an emerging approach for treatment of ischemic diseases. Stem Cell Res Ther 10, 158 (2019). https://doi.org/10.1186/s13287-019-1276-z

Download citation

  • Published:

  • DOI: https://doi.org/10.1186/s13287-019-1276-z

Keywords