Skip to main content

Engineering exosomes and biomaterial-assisted exosomes as therapeutic carriers for bone regeneration

Abstract

Mesenchymal stem cell-based therapy has become an effective therapeutic approach for bone regeneration. However, there are still limitations in successful clinical translation. Recently, the secretome of mesenchymal stem cells, especially exosome, plays a critical role in promoting bone repair and regeneration. Exosomes are nanosized, lipid bilayer-enclosed structures carrying proteins, lipids, RNAs, metabolites, growth factors, and cytokines and have attracted great attention for their potential application in bone regenerative medicine. In addition, preconditioning of parental cells and exosome engineering can enhance the regenerative potential of exosomes for treating bone defects. Moreover, with recent advancements in various biomaterials to enhance the therapeutic functions of exosomes, biomaterial-assisted exosomes have become a promising strategy for bone regeneration. This review discusses different insights regarding the roles of exosomes in bone regeneration and summarizes the applications of engineering exosomes and biomaterial-assisted exosomes as safe and versatile bone regeneration agent delivery platforms. The current hurdles of transitioning exosomes from bench to bedside are also discussed.

Introduction

Bone defects are often the consequence of high-energy trauma, infection, tumor excision or congenital defects. In addition, they are frequently accompanied by soft tissue damage, including injuries to muscles, tendons and joints. Bone healing might be initiated immediately following bone loss, which is a highly complex, well-orchestrated regenerative process of forming new bones and is involved in a series of fundamental cellular and molecular events [1]. Unfortunately, the intrinsic regenerative capacity of bone tissues fails to repair massive bone defects above a critical size [2]. Large bone defects require additional clinical intervention, and multiple options are currently available in clinical practice. The defective area is generally reconstructed and filled with grafts of an autogenous, allogeneic, or alloplastic nature to achieve satisfactory bone regeneration [3, 4]. Despite the clinical success of bone grafting, it is limited by major drawbacks, such as invasive surgical procedures, pain, secondary complications and disease transmission [5]. In addition, an ideal bone graft substitute should possess the following characteristics: osteoconduction, osteoinduction, osteoincorporation, osteointegration and osteogenesis [3]. However, to date, no single material has all the necessary properties to efficiently promote bone regeneration. In recent decades, MSC-based tissue engineering has become an effective therapeutic approach in the field of regenerative medicine, which has opened a new path for bone repair and regeneration [6, 7].

MSCs hold great promise for wide-ranging clinical applications and regenerative medicine, as they retain the potential to differentiate into multilineages under defined conditions and the ability to self-renew [8, 9]. In addition, MSCs can be expanded rapidly and easily in vitro, and the supernatants, growth factors and cytokines derived from MSCs are also cell-free sources for tissue regeneration [10]. It has been well documented that MSCs are of great importance during the bone formation or healing process [11, 12]. They have the capacity of migration and homing into or around the injured sites, which are the essential steps for achieving bone repair and regeneration. Then, MSCs are able to commit to osteogenic/chondrogenic progenitors and eventually differentiate into osteocytes/chondrocytes for bone tissue reconstruction after bone defects [13]. In addition, due to the immunomodulatory properties of MSCs, they escape immune recognition and protect against the cytotoxic effects of the host’s immune system [14]. Moreover, MSCs might promote bone regeneration by modulating the microenvironment. For instance, human adipose tissue-derived MSCs (hASCs) and their conditioned medium (CM) enhanced bone regeneration in a rabbit model of surgical bone lesions, suggesting that hASCs promoted bone regeneration mainly by releasing paracrine factors [15].

Considering the critical role of MSCs in bone regeneration, as expected, MSCs have been shown to be an efficacious treatment for bone defects [16]. For example, transplanted MSCs have been shown to not only survive and proliferate in injured sites but also differentiate into osteoblasts, leading to bone tissue reconstruction in a model of osteonecrosis of the femoral head [17]. Similarly, bone marrow MSC (BMSC) transplantation significantly accelerated bone regeneration in a rat mandibular osteodistraction model [18]. Percutaneous injection of MSCs isolated from bone marrow alone or with bioscaffolds such as biphasic hydroxyapatite/β-calcium-triphosphate granules has been shown to promote bone repair in patients with delayed healing of long bone fractures or avascular necrosis of the femoral head [19, 20]. Despite the vast number of MSC-based clinical trials that have been completed, none of the approaches have been approved for the treatment of bone defects. There are several significant barriers to the clinical application of MSC-based bone regeneration [21]. First, standardized protocols need to be established to avoid unwanted MSC differentiation during ex vivo expansion [22]. Second, the beneficial effects of MSC osteogenic differentiation for bone repair and regeneration are significantly impaired due to the poor engraftment and survival of MSCs in the injury sites [23]. Third, the number of administered MSCs is not sufficient for treating large bone defects in the clinical setting, and long-term in vitro expansion might affect the biological functions of MSCs [24]. Other limitations in MSC-based bone regeneration concern ethical issues and the possible risk of tumorigenicity [25].

The key success factor of MSC-based bone regeneration is because MSCs are able to differentiate into bone-forming osteoblasts and cartilage-forming chondrocytes [26]. It is expected that the implanted or injected MSCs will commit to a particular lineage at the site of injury for tissue repair and regeneration. However, the survival status and the differentiation capacity of the implanted/injected MSCs are unfavorable, thereby casting doubt on its correct mechanism of action. Interestingly, growing evidence has demonstrated that MSCs might exert their therapeutic effects mainly through paracrine effects [19, 27, 28]. The MSC-derived secretome includes vesicles, growth factors, cytokines, chemokines, extracellular matrix (ECM) and metabolites and plays an indispensable role in modulating tissue repair and regeneration [29]. For example, after incorporating human umbilical cord MSC (hucMSC)-derived extracellular vesicles into tubular epithelial cells, the complex was injected into the injured kidneys, and surprisingly, kidney function and morphology were significantly improved [30]. MSC-derived immunomodulators and trophic factors, including vascular endothelial growth factor (VEGF), transforming growth factor-β, and hepatic growth factor, contribute to the positive effects of the BMSC secretome in the bone regenerative process [31]. The secretome in the CM from MSCs mimicked the native bone healing procedures to enhance bone regeneration [6].

As this review aims to summarize the recent progress on engineering exosomes and biomaterial-assisted exosomes as therapeutic carriers for bone regeneration, we mainly referred to the research articles published in recent 5 years. The databases we mainly used included PubMed, Google scholar, Scopus, Web of Science and ProQuest by using the following keywords: “exosomes”, “mesenchymal stem cells”, “preconditioning”, “exosome engineering”, “biomaterial scaffolds”, and “bone regeneration”. The articles were further screened to identify their relevance to this review.

Exosome concept, biogenesis, content and application

Exosomes are nanosized, lipid bilayer-enclosed structures carrying proteins, lipids, RNAs, metabolites, growth factors and cytokines and have attracted great attention for their critical role in mediating cell–cell communication locally and systemically [32]. Exosomes are derived from the endocytic pathway of most cells and released from multivesicular bodies (MVBs) into nearly all biological fluids, such as blood, saliva and urine [33]. The formation of exosomes can be classified into four processes: invagination, endosome formation, fusion and secretion. Initially, early endosomes are formed by invagination of the plasma membrane, which are subsequently transformed into late endosomes containing MVBs. Following the fusion of MVBs with the plasma membrane, the exosomes are consecutively released from the cells [34]. The abundance, composition and functional properties of exosomes depend on the origin of the cell/tissue, the physiological/pathological state and even the microenvironment in which the parental cells reside [35]. The main role of exosomes lies in their capability to deliver information to adjacent cells and even to those cells that are remote from the exosome cellular origin, thereby influencing their function [36]. The contents of circulating exosomes are different between patients and healthy individuals, which can reflect the real-time state. Therefore, exosomes are emerging as robust and promising noninvasive biomarkers for the diagnosis, prognosis and prediction of treatment efficacy in various diseases [37]. More importantly, exosomes have been developed as therapeutic carriers for treating diseases. The advantage of using exosomes as delivery vehicles is their capacity to load both hydrophobic and hydrophilic items either inside or in the lipid bilayer [38]. The cargos are shielded by the bilayer membranes, which effectively protects the cargos from enzymatic degradation [38, 39]. In addition, the long circulation property of MSC exosomes prolongs their residence time in the blood and reduces the clearance rate of drugs they carry. MSC-produced exosomes can target specific cell types through their surface receptors [40, 41]. Exosomes have also been proven to be able to cross biological barriers and travel into deep tissues [40, 42]. Moreover, exosomes are efficacious for promoting regeneration, inducing stem cell differentiation and triggering particular immunological reactions [43]. Furthermore, they also exhibit great biocompatibility, biodegradability and stability, as well as low immunogenicity (Fig. 1) [43]. Small molecules, nucleic acid drugs or bioactive molecules can be functionally incorporated into exosome-based nanocarriers and then transported to targeted sites to achieve therapeutic outcomes [44]. Pascucci et al. [45] reported that MSC-derived exosomes loaded with chemotherapeutic drugs trafficked to tumor tissues and exerted antitumorigenic effects. In addition, MSC-derived exosomes also exhibit promising therapeutic potential for bone regeneration. As a critical mediator of intercellular communication, exosomes play a prominent role in a variety of biological processes including bone formation. MSC-derived exosomes exert prolific therapeutic efficacy in bone defects as they are capable of targeting bone tissues to induce osteogenic differentiation, thereby accelerating the process of bone regeneration [46]. For example, in a rat model of disuse osteoporosis, the diseased animals treated with hucMSC-derived exosomes formed new bone, and the bone structural parameters were significantly improved [47]. Exosomes secreted from human-induced pluripotent stem cells (hiPSC) enhanced bone regeneration in the osteoporotic rat model [48]. BMSC-derived exosomes stimulate osteoblast proliferation and osteogenic differentiation, thereby promoting osteogenesis in the rabbit model of osteonecrosis of the femoral head [49]. In addition, exosomes have a certain degree of natural targeting capacity. The targeting specificity of exosomes is mainly achieved by surface receptor–ligand binding to target specific cells, and then, exosomes can stimulate intracellular signaling in recipient cells or deliver their components to stimulate osteoblast proliferation and differentiation [50]. However, the therapeutic efficacy of natural exosomes for bone regeneration is limited [51]. MSC-derived exosomes can be captured by the reticuloendothelial system or cleared by the mononuclear phagocyte system, which might prevent their accumulation in the injured sites to exert desirable regenerative effects [52, 53]. Additionally, the natural targeting property of MSC exosomes is not accurate enough for delivering the indicated cargos to the specific recipient cells or tissues. Engineering or manipulating exosomes can optimize their targeting capability, providing potential wide clinical applications for treating diseases, including bone defects [54]. Here, we summarize recent research progress on naturally derived exosomes and engineered exosomes in bone repair and regeneration. In addition, as biomaterial scaffolds are the basic material for bone tissue engineering, the effects of biomaterial-assisted exosomes on bone regeneration have also been reviewed. The practical challenges associated with exosome-based bone regeneration are also discussed.

Fig. 1
figure 1

Exosomes derived from MSCs as a delivery vehicle have the advantages of low immunogenicity, easily uptaken by cells, easily crossing biological barriers, cargo protection from degradation, easily loading therapeutics, high cargo release stability, high biocompatibility, long circulation, and having the ability of tissue targeting. Therefore, they have been developed as a novel strategy for treating various diseases

Mechanism of MSC-derived exosomes for bone regeneration

MSCs-derived exosomes promote bone regeneration by directly transferring their internal cargos and subsequently controlling downstream signaling pathways in the targeted cells [55, 56]. Exosomes also regulate immune responses, inhibit osteoclast activities and induce osteogenesis and angiogenesis [57]. Various signaling cascades, including BMP/Smad, Wnt/β-catenin and PI3K/AKT, are activated by MSC exosomes, resulting in bone regeneration by promoting osteoblast proliferation and differentiation as well as the recruitment of endogenous MSCs to bone defect sites [57,58,59]. In addition, MSC exosomes repair bone defects by enhancing local angiogenesis and suppressing bone resorption, and activation of AKT/mTOR signaling pathway might be responsible for the osteogenesis-promoting effects [60, 61]. Moreover, MSC exosomes are effective in improving overall wound healing by inhibiting inflammatory responses and preventing osteoclast activities [62, 63]. The release of BMP-2 and other osteogenic growth factors from macrophages and non-stem cells were markedly elevated following the addition of MSC exosomes, which subsequently accelerated the healing of damaged tissues [46, 62]. It has also been demonstrated that MSC exosomes promote angiogenesis in bone defect area, which play a crucial role in bone regeneration [60, 61]. Vasculature serves as the main transport conduit for hormones and growth factors to supply the bone defect area with oxygen, nutrients and metabolites, which are essential for bone growth and regeneration [64]. A number of exosomal miRNAs such as miRNA-135b, miRNA-204 and miRNA-196a have been shown to play a critical role in regulating the bone regeneration process [65,66,67]. Interestingly, the immune microenvironment formed by immune cells and their metabolites is also indispensable for bone regeneration. The immune microenvironment not only controls the activities of osteoblasts and osteoclasts, but also the secretion of chemokines, growth factors and inflammatory factors, which in turn modulates the formation of new bone and the strength of existing bones [68, 69]. MSC exosomes are crucial in modulating immune cell activities in the complicated internal environment [46, 62]. For instance, by delivering the intracellular cargos, MSC exosomes induce the polarization of macrophages from the M1 to the M2 phenotype, contributing to the establishment of anti-inflammatory microenvironment during bone defect repair [70].

Modification of MSC-derived exosomes for bone regeneration

Due to the limitations of natural exosomes, such as lack of sufficient production, instability in circulation and poor targeting capacity, various strategies have been developed to modify MSC-derived exosomes and enhance their therapeutic potential [71]. The avenues for the modification of exosomes are mainly divided into two aspects: preconditioning of parental cells and exosome engineering (Fig. 2).

Fig. 2
figure 2

The avenues for the modification of exosomes are mainly divided into two aspects: preconditioning of parental cells and exosome engineering. Preconditioning of parental cells for bone regeneration mainly included hypoxic preconditioning, cytokine preconditioning and chemical preconditioning. Exosome engineering is mainly divided into two categories: cargo packaging into exosomes and surface modification of exosomes

Preconditioning of parental cells

Although exosomes derived from MSCs have been widely explored for cell-free therapy of various diseases, limitations, including low production of MSC exosomes, the demand for large amounts of exosomes, heterogeneity and low targetability impede the clinical trials of MSC-derived exosomes [51]. Growing evidence indicates that the preconditioning of parental cells is an adaptive strategy to enhance the therapeutic efficacy and yield of MSC exosomes [51]. Preconditioning of MSCs overcomes the limitations of natural exosomes by enhancing MSC paracrine activities to increase the production of MSC exosomes [72, 73]. In addition, pretreatment can promote exosomes from MSCs to regulate both the innate and adaptive immune responses of recipient cells [72, 73]. The approaches for preconditioning of MSCs mainly include hypoxic preconditioning, cytokine preconditioning and chemical preconditioning.

Hypoxic preconditioning

Hypoxia refers to the cell culture conditions under 0–10% oxygen tension. Oxygen tension plays an essential role in maintaining bone homeostasis [74]. The 1% oxygen tension in physiological cartilage and bone marrow is far lower than the 21% O2 routinely used for MSC culture [75]. A hypoxic environment can lead to cell death; however, hypoxic preconditioning can reduce hypoxia-induced apoptosis by elevating the expression of pro-survival signaling [76]. Interestingly, recent studies showed that the therapeutic effects of MSC-derived exosomes after hypoxic preconditioning were significantly enhanced for treating a wide array of diseases, including bone fracture healing [77, 78], acute kidney injury [79], spinal cord injury [80], diabetic wounds [81], myocardial infarction [82,83,84] and insufficient vessel growth [85].

Hypoxic preconditioning can enhance the cytoprotective and regenerative effects of MSCs to promote the secretion and therapeutic effects of their exosomes in injury sites such as bone defects and osteoarthritis [86, 87]. Mechanistically, hypoxic preconditioning increases the levels of cytoprotective molecules and maintains the multipotent and proliferative capabilities of stem cells [88,89,90]. An increasing number of studies have demonstrated that hypoxic preconditioning may improve the osteogenic differentiation and proliferation of MSCs by elevating the expression of growth factors and stemness-related markers such as SOX2, OCT4, NANOG and KLF4 [91, 92]. In addition, hypoxia preconditioning contributes to the activation of hypoxia-inducible factor (HIF-1α), which subsequently promotes the expression of angiogenic factors [93]. Similarly, hypoxic preconditioning can increase the relative expression levels of molecule contained in MSC exosomes in the same way.

MSC-derived exosomes from hypoxic conditioning have shown superior abilities in enhancing osteogenesis and angiogenesis, and the secretion of exosomes was found to be significantly improved after hypoxic preconditioning [77, 78]. For example, the focal adhesion pathway, thyroid hormone synthesis and VEGF signaling pathway were significantly upregulated in exosomes from stem cells from human-exfoliated deciduous teeth (SHEDs) after hypoxic preconditioning [77]. Thus, exosomes derived from hypoxic-preconditioned SHEDs showed superior potential for cellular osteogenesis and angiogenesis in a rat calvarial defect model [77]. In addition, exosomes derived from hypoxia-preconditioned hucMSC healed bone fractures via the SPRED1/Ras/Erk signaling pathway [78]. In addition, hypoxic preconditioning activated HIF-1α to promote the secretion of exosomal miRNA-126 for bone fracture healing by enhancing proliferation, angiogenesis and migration to a greater extent [78]. Exosomes from BMSCs after hypoxic preconditioning promoted the proliferation and migration of chondrocytes and inhibited the apoptosis of chondrocytes via the miRNA-181c-5p/MAPK or miRNA-18-3p/JAK/STAT signaling pathways [86]. MicroRNAome analysis revealed that hsa-miRNA-18a-3p, hsa-miRNA-181c-5p, hsa-miRNA-337-5p and hsa-miRNA-376a-5p were differentially expressed between normoxia-preconditioned and hypoxia-preconditioned BMSC-derived exosomes [86]. In contrast, oxidative stress caused by H2O2 inhibited the release of exosomes from human periodontal ligament cells (hPDLCs) through a large accumulation of MVB caused by reducing the expression of Rab11a and Rab27a [94]. In general, hypoxia preconditioning is a promising strategy for enhancing the effectiveness of MSC-exosome-based therapy in bone regeneration.

Cytokine preconditioning

Accumulating evidence has demonstrated that preconditioning MSCs with inflammatory cytokines promotes the secretion of immunoregulatory factors [95]. The immune responses regulated by exosomes from cytokine-preconditioned MSCs include transferring MSCs to a more anti-inflammatory phenotype and inducing the polarization of anti-inflammatory M2 macrophages [96,97,98,99]. MSC-chemokine preconditioning can concentrate numerous immunocytes for their migration and homing into or around the injured sites to combat inflammatory responses. This shows that MSC-chemokine preconditioning is a promising strategy for bone regeneration by decreasing inflammation in bone defects [98,99,100,101].

Tumor necrosis factor-alpha (TNF-α) is one of the most studied inflammatory cytokines. An increasing number of studies have shown that TNF-α preconditioning enhances the therapeutic effects of exosomes derived from MSCs in various diseases, including bone defects [100], periodontitis [98], retinal ganglion injury [102], acute liver failure [96] and urethral stricture [97]. TNF-α promoted the secretion of a greater number of anti-inflammatory MSC exosomes by upregulating inflammatory suppression-related miRNAs, including miRNA-146a and miRNA-299-3p [96, 97]. These anti-inflammatory MSC exosomes contributed to the transfer of MSCs to a more anti-inflammatory phenotype [96, 97]. Preconditioning MSCs with TNF-α not only promoted the anti-inflammatory ability of MSC exosomes but also enhanced the osteogenic differentiation potential of MSC exosomes. hASCs preconditioned with TNF-α regulated the proliferation and osteogenic differentiation of human primary osteoblastic cells through Wnt signaling [100]. Exosomes derived from gingival tissue-derived MSCs (GMSCs) preconditioned with TNF-α regulated inflammation and osteoclastogenesis in a ligature-induced periodontitis mouse model by promoting the secretion of exosomes, upregulation of the exosomal CD73 expression, and induction of the polarization of anti-inflammatory M2 macrophages [98]. In addition, preconditioning GMSCs with TNF-α promoted the therapeutic effects of GMSC exosomes, including decreasing the number of tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts and suppressing periodontal bone resorption [98].

Interleukin-1β (IL-1β) has been shown to promote the anti-inflammatory effects of MSC exosomes in osteoarthritis by increasing the expression of miRNA-147b [101]. In addition, exosomes derived from BMSCs preconditioned with IL-1β inhibited the inflammation of hippocampal astrocytes and status epilepticus mice through suppression of the NRF2 signaling pathway [103]. Exosomes derived from transforming growth factor-β1 (TGF-β1)-preconditioned BMSCs attenuated cartilage damage in osteoarthritis rat models by upregulating miRNA-135b, which promoted M2 polarization of synovial macrophages by targeting MAPK6 [99]. In summary, preconditioning of certain cytokines and growth factors promotes the osteogenic potential and anti-inflammatory function of MSC exosomes. It should be pointed out some potential cytokines and growth factors are promising candidates for MSC preconditioning. For example, preconditioning of MSCs with IFN-γ showed the potential of anti-inflammatory and angiogenic abilities [104]. In addition, growth factors such as FGF 2, IGF-1, IGF-2, TGF-β1 and PDGF are efficacious in promoting soft tissue healing, and the effects of MSC preconditioning with these growth factors on bone regeneration warrant further investigation [104, 105].

Chemical preconditioning

Chemical signals can be rapidly detected by MSCs and may significantly alter their phenotypes [51]. In addition, the influence also changes the secretion and content of MSC exosomes, which show promising potential in immune regulation and tissue regeneration [106]. For example, exosomes derived from kartogenin-preconditioned hucMSCs induced chondrogenic differentiation by promoting the secretion of exosomal miRNA-381-3p through targeting TAOK1 [107]. Similarly, kartogenin-reconditioned mouse BMSC (mBMSC)-exosomes have a better ability to promote chondral matrix regeneration than exosomes derived from untreated mBMSCs [108]. In addition, exosomes derived from curcumin-preconditioned MSCs attenuated osteoarthritis by modulating the miRNA-124/NF-kB and miRNA-143/ROCK1/TLR9 signaling pathways [109].

Exosome engineering

Exosome engineering has been explored to enhance the therapeutic potential of MSC exosomes [110]. Exosome engineering is mainly divided into two categories: cargo packaging into exosomes and surface modification of exosomes [51].

Cargo packaging into exosomes

Packaging cargo into exosomes can promote the therapeutic effects of MSC exosomes. The methods of packaging cargo into exosomes can be divided into two main categories: endogenous and exogenous cargo loading methods [51]. Endogenous cargo loading is the modification of parental cells with viral vectors and plasmids, while exogenous cargo loading is the direct loading of drugs into the extracted MSC exosomes [51]. Endogenous cargo methods are usually used to load endogenous proteins and nucleotides with therapeutic effects, while exogenous cargo loading methods are usually used to load small molecule drugs [111].

Endogenous cargo loading

Accumulating evidence shows that the functional biomolecules of MSC exosomes, such as nucleotides and proteins, play essential roles in osteogenesis, angiogenesis, immunomodulation and tissue regeneration in different kinds of disease models [112]. Genetic engineering tools, such as viral vectors and plasmids, can genetically manipulate the endogenous molecule expression levels in MSCs.

Growing evidence indicates that miRNA delivery plays a crucial role in improving the therapeutic potential of MSC exosomes in various disease models. Exosomes from miRNA-375-overexpressing hASCs embedded with hydrogel promoted the bone regeneration of BMSCs by inhibiting the expression of IGFBP3 [113]. Exosomes from miRNA-181b-overexpressing BMSCs enhanced osteogenesis in vitro and osteointegration in vivo by secreting VEGF and bone morphogenetic protein 2 (BMP2) to promote M2 polarization and inhibit inflammation by activating the PRKCD/AKT signaling pathway [114]. Exosomes from miRNA-140-5p-overexpressing synovial mesenchymal stem cells promoted the migration and proliferation of articular chondrocytes in an osteoarthritis rat model [115]. Exosomes from BMP2-overexpressing BMSCs enhanced osteogenic regeneration and osteoinduction in a rat calvarial defect model [116, 117]. Exosomes derived from BMSCs modified with mutant HIF-1α (BMSC-ExosMU) significantly promoted the osteogenic differentiation ability of BMSCs in vivo, while exosomes derived from BMSCs modified with wild-type HIF-1α (BMSC-ExosWT) did not show osteogenic differentiation ability [118]. In addition, BMSC-ExosMU markedly increased angiogenesis and bone regeneration in the necrotic regions by increasing microvascular density and trabecular reconstruction [118].

Exogenous cargo loading

Physical methods are usually employed to encapsulate drugs into engineered exosomes, such as permeabilization with saponins, freeze‒thaw cycles, extrusion, or sonication, for treating various diseases [119, 120]. For example, sonication was used to fractionate exosome membranes into small vesicles, and then, drugs were extruded into the vesicles [119, 120]. Other methods for exogenous cargo loading usually included permeabilization with saponin, freeze‒thaw cycles and incubation at room temperature for treating various diseases [119]. BMP2 was directly loaded into engineered extracellular vesicles of BMSCs, which induced osteogenic regeneration in vitro and in vivo, while BMP2 was absent in the exosomes of untreated group [121]. Compared to naturally occurring extracellular vesicles, engineered BMP2-extracellular vesicles protected BMP2 from proteolysis [121]. ATDC5 was encapsulated with the VEGF gene into exosomes, which induced vascularized bone regeneration [122]. Mechanistically, exosomes loading VEGF gene promoted the osteogenic differentiation ability of MSCs by controllably releasing the vascularized gene VEGF to remodel the vascular system and combining the 3D-printed porous bone scaffolds with engineered exosome nanoparticles (NPs) [122]. In addition, the exosomes encapsulated with the VEGF gene showed the best bone regeneration ability and induced the formation of a large amount of new bone in a rat radial defect repair model [122].

Surface modification

Surface modification is used to enhance the targeting ability of MSC exosomes to promote their therapeutic effects by direct modification of exosome surface molecules [111]. Targeting ligands can be added onto the surface of exosomes to precisely deliver therapeutic drugs to the indicated lesions. For example, an aptamer was combined with exosomes derived from bone marrow stromal cells (STExos) to generate the STExo-aptamer complex [123]. The complex enhanced the bone mass in an ovariectomy-induced postmenopausal osteoporosis mouse model and promoted bone healing in a femur fracture mouse model after intravenous injection [123]. C-X-C motif chemokine receptor 4 (CXCR4) was added to the surface of exosomes derived from genetically engineered NIH-3T3 cells [124]. The complex was then combined with liposomes carrying antagomir-188 to generate hybrid NPs. The hybrid NPs recovered the loss of age-related trabecular bone and reduced cortical bone porosity in mice by specifically gathering in the bone marrow and controllably releasing antagomir-188 to inhibit adipogenesis and promote osteogenesis of BMSCs [124].

Collectively, the modification of exosomes enhances the osteogenic and angiogenic potential of MSC exosomes by increasing the secretion of MSC exosomes and regulating the expression levels of osteogenic and angiogenic genes, which are promising therapeutic strategies for bone regeneration. We summarized the methods of exosome modification in Table 1.

Table 1 Modification of exosomes for improving osteogenic therapy potential

Biomaterial-based MSC-derived exosomes for bone regeneration

Despite advancements in natural exosomes for bone regeneration applications, there are still certain limitations in complex composition properties, limited cargo loading efficiency and undesirable retention stability. However, exosomes can bind to biomaterial scaffolds to enhance bone regeneration mainly by inducing osteogenesis and angiogenesis (Fig. 3). A range of biomaterial scaffolds endows exosomes with desirable pharmaceutical acceptability by extending exosome storage time and altering the release properties, thereby overcoming the drawbacks of natural exosomes. The use of biomaterial-assisted exosomes has proven to be a novel strategy for regenerating bone tissues [125]. Biomaterials employed for scaffolds can be divided into metal materials, bioactive ceramics, hydrogels and synthetic polymers. We have summarized the biomaterial-based exosomes for bone regeneration applications in Table 2.

Fig. 3
figure 3

Exosomes can bind to biomaterial scaffolds, including metal materials, bioactive ceramics, hydrogels and synthetic polymers, and then target injured sites to enhance bone regeneration mainly by inducing osteogenesis and angiogenesis

Table 2 Biomaterials scaffold for bone regeneration application

Metal materials

Biodegradable metal materials are widely used biomaterials in exosome engineering for repairing bone defects. Their excellent biocompatibility, easy production and processing, mild stretchability, and corrosion resistance make them suitable for use as exosome-loaded scaffolds to promote bone regeneration [126]. For example, MSC-derived exosomes incorporated into silver NPs hybrid scaffolds induced osteogenesis, thus serving as a promising cell-free therapeutic for bone regeneration [127]. In addition, titanium alloys are also scaffolds for bone defect treatment with desirable biocompatibility, excellent friction coefficient, high porosity and corrosion resistance. Exosomes derived from human MSCs (hMSCs) loaded into 3D-printed titanium alloy scaffolds can be used to achieve cell-free bone regeneration. The exosomes released by 3D-printed titanium alloy-decorated hMSCs induced osteogenic differentiation of hMSCs and regenerated bone tissues by upregulating osteogenic miRNAs, downregulating anti-osteogenic miRNAs and activating the PI3K/Akt and MAPK signaling pathways [128].

Bioactive ceramic

Bioactive ceramic biomaterials, including bioactive glass and tricalcium phosphate, are also desirable alternatives for repairing bone tissue that have good toughness and biocompatibility and different characteristics [129].

Mesoporous bioactive glass

Mesoporous bioactive glass (MBG) is a hierarchical structure scaffold that has been proven to be beneficial for bone repair with positive biological effects on osteogenesis [130]. For example, optimized osteogenic BMSC-derived exosomes were loaded into MBG scaffolds to realize bioactivity maintenance and sustained release of exosomes, thereby efficiently promoting bone-forming ability and enhancing rapid initiation of bone regeneration [131].

Tricalcium phosphate

β-Tricalcium phosphate (β-TCP), resembling the human autologous bone component, is one of the most commonly used bone tissue engineering biomaterials. β-TCP scaffolds with advanced osteotransductive ability and high osteoinductive potential are able to maintain the balance between material degradation and osteogenesis by releasing calcium ions and sulfates [132]. Therefore, β-TCP is an optimal scaffold for bone regeneration. Porous β-TCP scaffolds can be used as exosome carriers and implanted into the bone defect area, thereby promoting bone regeneration after being loaded into MSC-derived exosomes in a concentration-dependent manner. For instance, exosomes derived from MSCs in combination with β-TCP induced new bone formation. This MSC-exosome-β-TCP scaffold complex improved the osteoinductivity of β-TCP and possessed better osteogenesis activity than β-TCP alone. Moreover, gene expression profiling and bioinformatics analyses revealed that the MSC-exosome-β-TCP scaffold promoted bone regeneration mainly by activating the PI3K/AKT signaling pathway [133]. Exosomes released by hiPSC (hiPSC-Exos) combined with β-TCP scaffolds can potentially be used for bone repair. The β-TCP scaffolds incorporated into hiPSC-Exos were able to significantly promote bone regeneration in an osteoporotic rat skull defect model, and the therapeutic effect was increased with increasing concentrations of hiPSC-Exos. In addition to osteogenesis, the hiPSC-Exos + β-TCP scaffolds also markedly enhanced angiogenesis in the area of the calvarial defect, and the restoration of blood flow was capable of providing nutrients and renewable autologous cells to repair critical-sized bone defects [48]. BMSC-Exos-HIF1α loaded onto the β-TCP scaffolds implanted in the bone defect area repaired bone defects by promoting new bone regeneration in cranial critical-sized bone defect rats [134]. Additionally, β-TCP scaffolds are also employed to repair bone defects in alveolar regions for treating periodontitis. For example, exosomes secreted by SHED-derived exosomes combined with β-TCP scaffolds induced alveolar bone regeneration and neovascularization by promoting osteogenesis-related gene expression and phosphorylation of AMPK [135]. Exosomes released from human periodontal ligament stem cells (hPDLSCs) were loaded with β-TCP scaffolds and then applied to repair bone defects, leading to increased formation of alveolar bone in rat models of periodontitis [136].

Bioactive ceramic supplemented with bone substitute

However, the limited bioactivity of ceramics may influence their regenerative effects. Therefore, bioactive materials such as bone substitutes can be added to promote their performance. Using a calcium sulfate-nanohydroxyapatite nanocement (NC) bone filler as the MSC-exosome carrier provides a newly improved approach for bone regeneration. After implantation in rat tibia critical defect models, this complex enhanced bone regeneration by inducing bone mineralization [137].

Hydrogels

Currently, hydrogels scaffolds have been extensively utilized in the field of bone regeneration, as they are compatible biomaterials acting as exosome carriers and exosome delivery reservoirs. For bone regeneration, hydrogels may be synthesized from various biodegradable polymers, such as hyaluronic acid (HA), chitosan (CS), silk fibroin (SF), alginate (ALG) and polyethylene glycol (PEG) [138].

Hydrogels are three-dimensional network structure polymer chains with superior mechanical strength and can mimic the natural ECM of bone tissue. Therefore, hydrogels provide nutrient environments suitable for endogenous cell proliferation, thus presenting a prospective ability of exosome encapsulation for bone regeneration [139]. Due to the 3D network structure and physiochemical properties of the hydrogels, the encapsulated bioactive molecules are confined in the meshes. In addition, exosomes formulated in hydrogel scaffolds may reduce the degradation rate and easily maintain the release of exosomes as needed, contributing to the high local concentrations of desired pharmacologically important molecules contained in exosomes to achieve desirable therapeutic effects. These properties make hydrogels a promising scaffold for cell-free therapy in targeting bone tissue sites and facilitate localized delivery of therapeutic agents, thereby promoting bone regeneration for treating bone defects [138].

Natural hydrogels

Hydrogels prepared by natural polysaccharides and proteins, including hyaluronic acid, hydrogel alginate, chitosan and gelatin, can be used as scaffolds for exosome carriers with desirable biodegradability and proper cell interactions in the treatment of bone defects.

HA is a nonimmunogenic natural polymer that is the major component of the ECM structure and critical to tissue regeneration [140]. Umbilical MSC-derived exosomes (uMSCEXOs) combined with HA hydrogels (HA-Gel) and then customized nanohydroxyapatite/poly-ε-caprolactone (nHP) scaffolds were shown to repair cranial defects in rats. The uMSCEXOs/HA-Gel/nHP complex markedly accelerated the bone regeneration process [141]. Yang et al. combined HA-ALG hydrogels and hydroxyapatite (HAP) with hucMSC-derived exosomes targeting bone defect sites in a calvarial defect rat model for bone regeneration. The composite hydrogel system durably retained exosomes at the targeting sites and significantly promoted the healing of damaged bone by enhancing osteogenic abilities [142]. CS is another polysaccharide with a linear structure composed of β-(1,4)-linked D-glucosamine and N-acetyl-D-glucosamine units with intrinsic bone healing properties. BMSC-derived exosomes were added to the CS/β-glycerophosphate hydrogel for bone regeneration in rats with calvarial defects. These exosome-release hydrogels were biocompatible and exhibited excellent therapeutic effects for repairing bone tissues [143]. Gelatin-based hydrogels have also attracted attention as potentially implantable materials in bone engineering applications due to their functionality. For example, hASC-derived exosomes combined with gelatin NPs hydrogel scaffolds accurately transported to the target sites and exerted a stronger bone repair capacity [70]. Natural hydrogel scaffolds have also shed light on the therapy of bone loss in periodontitis. For example, BMSC-derived exosomes were isolated and then loaded into a natural hydrogel for injection into rats with experimental periodontitis. The BMSC-Exo-hydrogel system promoted the migration, proliferation, and osteogenic differentiation of hPDLSCs for periodontal regeneration [62]. Exosomes derived from dental pulp stem cells (DPSCs) encapsulated into CS hydrogel effectively accelerated the regeneration of alveolar bone in a mouse model of periodontitis [144].

Moreover, the secretome of MSCs is able to provide specific peptides, and hydrogel-assisted exosomes combined with these peptides can improve the effects of bone regeneration. For example, small intestinal submucosa (SIS) hydrogels incorporated with 3-(3,4-dihydroxyphenyl) propionic acid (CA) modified by fusion peptides and exosomes derived from BMSCs were considered desirable for bone regeneration due to the positive effect of the exosomes in promoting the osteogenic differentiation of BMSCs. The combination of SIS hydrogels and CA significantly improved the mechanical properties of the hydrogels, and fusion peptides were designed to enhance the retention and stability of exosomes. Therefore, these scaffolds strengthen the therapeutic effect of MSC-derived exosomes on bone defects [145].

Synthetic hydrogels

Despite the advantages of using natural hydrogels in bone repair, the drawbacks of natural materials for hydrogel preparation are less stability, and undesirable mechanical characteristics. The incorporation of MSC-derived exosomes into synthetic hydrogels may be used to improve bone regeneration effects. Hydrogels can be synthesized from various polymeric biodegradable materials. Synthetic hydrogels present a longer shelf life, reliable mechanical properties and lower risk of immunogenicity, making them suitable for bone tissue regeneration. For instance, stem cells from apical papilla-derived exosomes (SCAP-Exos) loaded into a bioresponsive PEG/DNA hybrid hydrogel facilitated bone regeneration for diabetic bone defects by the controlled release of SCAP-Exos [146]. A coralline hydroxyapatite/SF/glycol CS/difunctionalized PEG self-healing hydrogel was synthesized, and hucMSC-derived exosomes were loaded into the hydrogel. This synthetic hydrogel containing exosomes effectively promoted bone regeneration [147].

Hydrogels combined with bone engineering materials

The use of hydrogels alone may cause problems, due to their poor mechanical and chemical properties. The emergence of composite bone engineering materials, such as metal–organic frameworks (MOFs) and β-TCP scaffolds, can promote the widespread use of hydrogel composite scaffolds in bone regeneration. MOFs are promising platforms for biomedical applications because of their structural diversity, high surface areas, adjustable porosity, simple surface functionalization and tunable biocompatibility. The application of MOF scaffolds as exosome carriers is a novel strategy for bone regeneration. For instance, hADSC-derived exosomes and Mg-gallic acid (GA) MOF were combined for bone regeneration applications, taking advantage of hADSC-Exos, Mg2+, GA and MOF scaffolds. As the scaffolds degraded, exosomes were released continuously and then internalized by hBMSCs to enhance their osteogenic effects by stabilizing the bone graft environment, promoting osteogenic differentiation and stimulating bone reconstruction. Furthermore, exosomes secreted from the composite scaffolds were also proven to induce new bone formation and osseointegration in the rat calvarial defect model [148]. β-TCP can also be combined with hydrogels to enhance bone regeneration. Zhang et al. extracted exosomes originating from rat BMSCs (rBMSCs) and then cocultured them with polyethylene glycol maleate citrate (PEGMC) hydrogels and β-TCP to promote bone regeneration. The PEGMC/β-TCP-MSC-Exos showed great potential in rapid osteogenesis [149].

Synthetic polymers

Synthetic polymers with tunable mechanical properties, such as poly-lactic-co-glycolic acid (PLGA) and polylactic acid (PLA), have also been used as scaffolds for the development of biocompatible and biodegradable polymeric structures to regenerate bone tissues.

PLGA with excellent mechanical strength and biodegradation properties is another widely used biocompatible scaffold for bone regeneration. For instance, hASC exosomes immobilized on polydopamine-coated PLGA scaffolds effectively promoted bone healing in mouse skull defect models, and polydopamine (pDA) was used to provide a more efficient coating on the PLGA substrate. hASC-derived exosomes combined with PLGA/pDA scaffolds were capable of consistently releasing exosomes, resulting in enhanced bone regeneration through their osteoinductive effects. In addition, this exosome-composite scaffold also promoted MSC migration and homing in newly formed bone tissues [150]. PLGA and PEG were engineered with human DPSC (hDPSC)-derived exosomes to improve the controlled release of exosomes for bone regeneration. These engineered scaffolds constantly released osteogenic hDPSC-derived exosomes that facilitated the osteogenic differentiation of BMSCs, leading to mineralization and accelerated bone healing [151]. PLA is a versatile and biodegradable scaffold widely used in repairing tissue defects. Exosomes were isolated from MSCs and coated with porous 3D PLA scaffolds to potentiate osteogenic differentiation. PLA-MSC-Exos significantly improved osteogenesis, which holds great potential for applications in bone tissue regeneration [152]. Polyetheretherketone (PEEK) is a noncytotoxic, highly biocompatible and chemically stable aromatic polymer material that can be used for bone regeneration. BMSC-derived exosomes packaged with tannic acid-modified sulfonated PEEK continually released exosomes and exerted osteoimmunomodulation effects to enhance osteogenesis by promoting macrophage M2 polarization [153].

Limitations and future perspectives

Despite the promising application of exosomes in bone regeneration, there are certain limitations with the use of MSC exosomes for bone regeneration [154]. The main bottleneck in clinical application is the lack of standardized isolation and purification strategies for exosomes. Exosomes, as carriers of elements of cellular communication and interaction, can be detected in nearly all biological fluids including blood, saliva, urine and cerebrospinal fluid [155]. Effective extraction and separation of exosomes from different sources for bone regeneration are challenging. Ultrafiltration is the most commonly used method to isolate exosomes, with the advantages of easy operation and screening based on size [156]. However, it still has problems with large-scale production. The production of both high-quality and high-quantity exosomes remains challenging for the translation of these nanosystems into the clinic [44, 157].

Although the therapeutic benefits of modifications and exosome engineering have been demonstrated in various bone diseases, some challenges in preconditioning methods may limit their applications. MSC-exosome preconditioning can promote the anti-inflammatory and/or osteogenic potential of exosomes by regulating the secretion and content of MSC exosomes. However, the optimal time and intensity of different preconditioning still need to be further explored because of the heterogeneity of MSCs from different sources [51]. In addition, the long-term effects of preconditioning on the properties of MSCs and the evidence of consistently obtaining desirable exosomes from engineering methods still need to be evaluated in clinical [158, 159]. Accordingly, addressing these problems may increase their possibility of regenerative applications.

The emergence of biomaterials provides various options for exosome-based therapy; encapsulating exosomes into biomaterial scaffolds can optimize their application in bone regeneration, but there are still some limitations. For instance, it is difficult to maintain exosome release by scaffolds. Although the slow release is achieved, strategies for the consistently stable release of exosomes are not available, and the most suitable release rate for bone regeneration has not yet been explored [160]. Additionally, there was insufficient evidence to support the scalable manufacturing of scaffolds and efficient delivery of exosomes, and thus, further studies need to be performed to address these issues [138]. The promising cell-free therapy will surely attract deeper investigations to improve the production efficiency and quality of exosomes, the preconditioning or engineering methods, and the release of exosomes from the matched scaffold at the most appropriate rate, thereby maximizing exosome function in bone regeneration.

It should be noted that there is still a long way before exosome-based therapy can be comprehensively applied for bone regeneration in clinical settings. Many hurdles should be overcome before the clinical translation of MSC-derived exosomes. Firstly, the loading efficiency for exosomes is relatively low as their naturally complex contents limit the space for the loading of exogenous therapeutics into exosomes [161, 162]. More effective modification methodologies are required to develop to improve the drug loading efficiency. Secondly, the original sources, physiological states and cultural conditions of MSC may impact the targeting and biological properties of isolated exosomes [163,164,165]. Therefore, widely applicable standards for the isolation of MSC-derived exosomes should be established. Thirdly, several factors, such as the dosage of MSC exosomes and the use of scaffolds, may influence the potency of MSC exosomes for bone regeneration. A particular source and concentration of exosomes, a specific scaffold or delivery route and the frequency of treatment need to be identified for optimization in different treatments of specific bone defect/disease indications [16]. Exosomes provide a new direction for bone defect treatments. However, the regenerative efficacy of MSC exosomes for treating bone defects is still in infancy, as the current research is limited to small animal models. Efforts to progress to clinically related large animal models and ultimately to clinical trials would likely be helpful to advance the field of MSC exosomes for bone regeneration.

Conclusion

The process of natural bone healing lacks the capacity to repair massive bone defects beyond a critical size, which makes the treatment of bone defects an intractable clinical problem [166]. Currently, progress in the use of bone grafting for clinical applications has many challenges, such as invasive surgical procedures, pain, secondary complications and disease transmission. However, cell-free therapy has great potential for bone regeneration, with MSCs as the most commonly used cell-free source [167]. MSC-derived exosomes are able to promote bone regeneration, which is of great significance for the effective application of bone defect repair. Moreover, modifications of exosomes, exosomes engineering and combined with biomaterial scaffolds markedly enhance their therapeutic effects [57]. Although there are currently limitations in MSC-derived exosome-based cell-free therapy, this is a promising field in bone regeneration, which can attract further investigations to confirm the clinical effect of treating bone defects.

Availability of data and materials

Not applicable.

Abbreviations

MSC:

Mesenchymal stem cell

hASCs:

Human adipose tissue-derived mesenchymal stem cells

CM:

Conditioned medium

BMSC:

Bone marrow mesenchymal stem cell

hucMSC:

Human umbilical cord mesenchymal stem cell

VEGF:

Vascular endothelial growth factor

MVBs:

Multivesicular bodies

hiPSC:

Human-induced pluripotent stem cells

HIF-1α:

Hypoxia-inducible factor

SHEDs:

Stem cells from human exfoliated deciduous teeth

hPDLCs:

Human periodontal ligament cells

TNF-α:

Tumor necrosis factor-alpha

GMSCs:

Gingival tissue-derived mesenchymal stem cells

TRAP:

Tartrate-resistant acid phosphatase

IL-1β:

Interleukin-1β

TGF-β1:

Transforming growth factor-β1

mBMSC:

Mouse bone marrow mesenchymal stem cell

BMP2:

Bone morphogenetic protein 2

NPs:

Nanoparticles

STExos:

Exosomes derived from bone marrow stromal cells

CXCR4:

C-X-C motif chemokine receptor 4

hMSCs:

Human mesenchymal stem cells

MBG:

Mesoporous bioactive glass

β-TCP:

β-Tricalcium phosphate

hiPSC-Exos:

Exosomes released by human-induced pluripotent stem cells

hPDLSCs:

Human periodontal ligament stem cells

HA:

Hyaluronic acid

CS:

Chitosan

SF:

Silk fibroin

ALG:

Alginate

PEG:

Polyethylene glycol

ECM:

Extracellular matrix

uMSCEXOs:

Umbilical mesenchymal stem cell-derived exosomes

HA-Gel:

Hyaluronic acid hydrogels

nHP:

Nanohydroxyapatite/poly-ε-caprolactone

DPSCs:

Dental pulp stem cells

SIS:

Small intestinal submucosa

CA:

3-(3,4-Dihydroxyphenyl) propionic acid

SCAP-Exos:

Stem cells from apical papilla-derived exosomes

MOFs:

Metal–organic frameworks

GA:

Gallic

PEGMC:

Polyethylene glycol maleate citrate

PLGA:

Polylactic-co-glycolic acid

PLA:

Polylactic acid

pDA:

Polydopamine

PEEK:

Polyetheretherketone

References

  1. Dimitriou R, Jones E, McGonagle D, Giannoudis PV. Bone regeneration: current concepts and future directions. BMC Med. 2011;9:66.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Walmsley GG, Ransom RC, Zielins ER, Leavitt T, Flacco JS, Hu MS, Lee AS, Longaker MT, Wan DC. Stem cells in bone regeneration. Stem Cell Rev Rep. 2016;12(5):524–9.

    Article  CAS  PubMed  Google Scholar 

  3. Wang W, Yeung KWK. Bone grafts and biomaterials substitutes for bone defect repair: a review. Bioact Mater. 2017;2(4):224–47.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Fillingham Y, Jacobs J. Bone grafts and their substitutes. Bone Jt J. 2016;98(B(1 Suppl A)):6–9.

    Article  Google Scholar 

  5. Sanz-Sanchez I, Sanz-Martin I, Ortiz-Vigon A, Molina A, Sanz M. Complications in bone-grafting procedures: classification and management. Periodontol 2000. 2022;88(1):86–102.

    Article  PubMed  Google Scholar 

  6. Marolt Presen D, Traweger A, Gimona M, Redl H. Mesenchymal stromal cell-based bone regeneration therapies: from cell transplantation and tissue engineering to therapeutic secretomes and extracellular vesicles. Front Bioeng Biotechnol. 2019;7:352.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Kuo CK, Tuan RS. Tissue engineering with mesenchymal stem cells. IEEE Eng Med Biol Mag. 2003;22(5):51–6.

    Article  PubMed  Google Scholar 

  8. Han Y, Li X, Zhang Y, Han Y, Chang F, Ding J. Mesenchymal stem cells for regenerative medicine. Cells. 2019;8(8):886.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kangari P, Talaei-Khozani T, Razeghian-Jahromi I, Razmkhah M. Mesenchymal stem cells: amazing remedies for bone and cartilage defects. Stem Cell Res Ther. 2020;11(1):492.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Konala VB, Mamidi MK, Bhonde R, Das AK, Pochampally R, Pal R. The current landscape of the mesenchymal stromal cell secretome: a new paradigm for cell-free regeneration. Cytotherapy. 2016;18(1):13–24.

    Article  CAS  PubMed  Google Scholar 

  11. Lin W, Xu L, Zwingenberger S, Gibon E, Goodman SB, Li G. Mesenchymal stem cells homing to improve bone healing. J Orthop Translat. 2017;9:19–27.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Qin Y, Guan J, Zhang C. Mesenchymal stem cells: mechanisms and role in bone regeneration. Postgrad Med J. 2014;90(1069):643–7.

    Article  PubMed  Google Scholar 

  13. Shang F, Yu Y, Liu S, Ming L, Zhang Y, Zhou Z, Zhao J, Jin Y. Advancing application of mesenchymal stem cell-based bone tissue regeneration. Bioact Mater. 2021;6(3):666–83.

    Article  CAS  PubMed  Google Scholar 

  14. Song N, Scholtemeijer M, Shah K. Mesenchymal stem cell immunomodulation: mechanisms and therapeutic potential. Trends Pharmacol Sci. 2020;41(9):653–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Linero I, Chaparro O. Paracrine effect of mesenchymal stem cells derived from human adipose tissue in bone regeneration. PLoS ONE. 2014;9(9):e107001.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Tan SHS, Wong JRY, Sim SJY, Tjio CKE, Wong KL, Chew JRJ, Hui JHP, Toh WS. Mesenchymal stem cell exosomes in bone regenerative strategies-a systematic review of preclinical studies. Mater Today Bio. 2020;7:100067.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Yan Z, Hang D, Guo C, Chen Z. Fate of mesenchymal stem cells transplanted to osteonecrosis of femoral head. J Orthop Res. 2009;27(4):442–6.

    Article  PubMed  Google Scholar 

  18. Qi M, Hu J, Zou S, Zhou H, Han L. Mandibular distraction osteogenesis enhanced by bone marrow mesenchymal stem cells in rats. J Craniomaxillofac Surg. 2006;34(5):283–9.

    Article  PubMed  Google Scholar 

  19. Wu J, Guo H, Liu X, Li M, Cao Y, Qu X, Zhou H, Weng L. Percutaneous autologous bone marrow transplantation for the treatment of delayed union of limb bone in children. Ther Clin Risk Manag. 2018;14:219–24.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Rosset P, Deschaseaux F, Layrolle P. Cell therapy for bone repair. Orthop Traumatol Surg Res. 2014;100(1 Suppl):S107–12.

    Article  CAS  PubMed  Google Scholar 

  21. Zhou T, Yuan Z, Weng J, Pei D, Du X, He C, Lai P. Challenges and advances in clinical applications of mesenchymal stromal cells. J Hematol Oncol. 2021;14(1):24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Jung S, Panchalingam KM, Rosenberg L, Behie LA. Ex vivo expansion of human mesenchymal stem cells in defined serum-free media. Stem Cells Int. 2012;2012:123030.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Li M, Jiang Y, Hou Q, Zhao Y, Zhong L, Fu X. Potential pre-activation strategies for improving therapeutic efficacy of mesenchymal stem cells: current status and future prospects. Stem Cell Res Ther. 2022;13(1):146.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Bijonowski BM, Yuan X, Jeske R, Li Y, Grant SC. Cyclical aggregation extends in vitro expansion potential of human mesenchymal stem cells. Sci Rep. 2020;10(1):20448.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Neri S. Genetic stability of mesenchymal stromal cells for regenerative medicine applications: a fundamental biosafety aspect. Int J Mol Sci. 2019;20(10):2406.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Lin H, Sohn J, Shen H, Langhans MT, Tuan RS. Bone marrow mesenchymal stem cells: aging and tissue engineering applications to enhance bone healing. Biomaterials. 2019;203:96–110.

    Article  CAS  PubMed  Google Scholar 

  27. Ahn SY, Chang YS, Sung DK, Yoo HS, Sung SI, Choi SJ, Park WS. Cell type-dependent variation in paracrine potency determines therapeutic efficacy against neonatal hyperoxic lung injury. Cytotherapy. 2015;17(8):1025–35.

    Article  CAS  PubMed  Google Scholar 

  28. Park WS, Ahn SY, Sung SI, Ahn JY, Chang YS. Strategies to enhance paracrine potency of transplanted mesenchymal stem cells in intractable neonatal disorders. Pediatr Res. 2018;83(1–2):214–22.

    Article  CAS  PubMed  Google Scholar 

  29. Marote A, Teixeira FG, Mendes-Pinheiro B, Salgado AJ. MSCs-derived exosomes: cell-secreted nanovesicles with regenerative potential. Front Pharmacol. 2016;7:231.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Zhou Y, Xu H, Xu W, Wang B, Wu H, Tao Y, Zhang B, Wang M, Mao F, Yan Y, et al. Exosomes released by human umbilical cord mesenchymal stem cells protect against cisplatin-induced renal oxidative stress and apoptosis in vivo and in vitro. Stem Cell Res Ther. 2013;4(2):34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Katagiri W, Osugi M, Kawai T, Hibi H. First-in-human study and clinical case reports of the alveolar bone regeneration with the secretome from human mesenchymal stem cells. Head Face Med. 2016;12:5.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Batrakova EV, Kim MS. Using exosomes, naturally-equipped nanocarriers, for drug delivery. J Control Release. 2015;219:396–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Demirer GS, Zhang H, Matos JL, Goh NS, Cunningham FJ, Sung Y, Chang R, Aditham AJ, Chio L, Cho MJ, et al. High aspect ratio nanomaterials enable delivery of functional genetic material without DNA integration in mature plants. Nat Nanotechnol. 2019;14(5):456–64.

    Article  CAS  PubMed  Google Scholar 

  34. Elsharkasy OM, Nordin JZ, Hagey DW, de Jong OG, Schiffelers RM, Andaloussi SE, Vader P. Extracellular vesicles as drug delivery systems: why and how? Adv Drug Deliv Rev. 2020;159:332–43.

    Article  CAS  PubMed  Google Scholar 

  35. Yanez-Mo M, Siljander PR, Andreu Z, Zavec AB, Borras FE, Buzas EI, Buzas K, Casal E, Cappello F, Carvalho J, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles. 2015;4:27066.

    Article  PubMed  Google Scholar 

  36. Farooqi AA, Desai NN, Qureshi MZ, Librelotto DRN, Gasparri ML, Bishayee A, Nabavi SM, Curti V, Daglia M. Exosome biogenesis, bioactivities and functions as new delivery systems of natural compounds. Biotechnol Adv. 2018;36(1):328–34.

    Article  CAS  PubMed  Google Scholar 

  37. Samal S, Dash P, Dash M. Drug delivery to the bone microenvironment mediated by exosomes: an axiom or enigma. Int J Nanomed. 2021;16:3509–40.

    Article  Google Scholar 

  38. Ren J, He W, Zheng L, Duan H. From structures to functions: insights into exosomes as promising drug delivery vehicles. Biomater Sci. 2016;4(6):910–21.

    Article  CAS  PubMed  Google Scholar 

  39. Barile L, Vassalli G. Exosomes: therapy delivery tools and biomarkers of diseases. Pharmacol Ther. 2017;174:63–78.

    Article  CAS  PubMed  Google Scholar 

  40. Tang XJ, Sun XY, Huang KM, Zhang L, Yang ZS, Zou DD, Wang B, Warnock GL, Dai LJ, Luo J. Therapeutic potential of CAR-T cell-derived exosomes: a cell-free modality for targeted cancer therapy. Oncotarget. 2015;6(42):44179–90.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Tian X, Zhu M, Nie G. How can nanotechnology help membrane vesicle-based cancer immunotherapy development? Hum Vaccin Immunother. 2013;9(1):222–5.

    Article  CAS  PubMed  Google Scholar 

  42. Sterzenbach U, Putz U, Low LH, Silke J, Tan SS, Howitt J. Engineered exosomes as vehicles for biologically active proteins. Mol Ther. 2017;25(6):1269–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Zhu Q, Heon M, Zhao Z, He M. Microfluidic engineering of exosomes: editing cellular messages for precision therapeutics. Lab Chip. 2018;18(12):1690–703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Luan X, Sansanaphongpricha K, Myers I, Chen H, Yuan H, Sun D. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacol Sin. 2017;38(6):754–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pascucci L, Cocce V, Bonomi A, Ami D, Ceccarelli P, Ciusani E, Vigano L, Locatelli A, Sisto F, Doglia SM, et al. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: a new approach for drug delivery. J Control Release. 2014;192:262–70.

    Article  CAS  PubMed  Google Scholar 

  46. Wang X, Thomsen P. Mesenchymal stem cell-derived small extracellular vesicles and bone regeneration. Basic Clin Pharmacol Toxicol. 2021;128(1):18–36.

    Article  CAS  PubMed  Google Scholar 

  47. Yang BC, Kuang MJ, Kang JY, Zhao J, Ma JX, Ma XL. Human umbilical cord mesenchymal stem cell-derived exosomes act via the miR-1263/Mob1/Hippo signaling pathway to prevent apoptosis in disuse osteoporosis. Biochem Biophys Res Commun. 2020;524(4):883–9.

    Article  CAS  PubMed  Google Scholar 

  48. Qi X, Zhang J, Yuan H, Xu Z, Li Q, Niu X, Hu B, Wang Y, Li X. Exosomes secreted by human-induced pluripotent stem cell-derived mesenchymal stem cells repair critical-sized bone defects through enhanced angiogenesis and osteogenesis in osteoporotic rats. Int J Biol Sci. 2016;12(7):836–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Liao W, Ning Y, Xu HJ, Zou WZ, Hu J, Liu XZ, Yang Y, Li ZH. BMSC-derived exosomes carrying microRNA-122-5p promote proliferation of osteoblasts in osteonecrosis of the femoral head. Clin Sci (Lond). 2019;133(18):1955–75.

    Article  CAS  PubMed  Google Scholar 

  50. He J, Ren W, Wang W, Han W, Jiang L, Zhang D, Guo M. Exosomal targeting and its potential clinical application. Drug Deliv Transl Res. 2022;12(10):2385–402.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chen S, Sun F, Qian H, Xu W, Jiang J. Preconditioning and engineering strategies for improving the efficacy of mesenchymal stem cell-derived exosomes in cell-free therapy. Stem Cells Int. 2022;2022:1779346.

    Article  PubMed  PubMed Central  Google Scholar 

  52. 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(13):4430–8.

    Article  CAS  PubMed  Google Scholar 

  53. Imai T, Takahashi Y, Nishikawa M, Kato K, Morishita M, Yamashita T, Matsumoto A, Charoenviriyakul C, Takakura Y. Macrophage-dependent clearance of systemically administered B16BL6-derived exosomes from the blood circulation in mice. J Extracell Vesicles. 2015;4:26238.

    Article  PubMed  Google Scholar 

  54. Al-Sowayan B, Alammari F, Alshareeda A. Preparing the bone tissue regeneration ground by exosomes: from diagnosis to therapy. Molecules. 2020;25(18):4205.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Xie Y, Chen Y, Zhang L, Ge W, Tang P. The roles of bone-derived exosomes and exosomal microRNAs in regulating bone remodelling. J Cell Mol Med. 2017;21(5):1033–41.

    Article  PubMed  Google Scholar 

  56. Gao M, Gao W, Papadimitriou JM, Zhang C, Gao J, Zheng M. Exosomes-the enigmatic regulators of bone homeostasis. Bone Res. 2018;6:36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Wang D, Cao H, Hua W, Gao L, Yuan Y, Zhou X, Zeng Z. Mesenchymal stem cell-derived extracellular vesicles for bone defect repair. Membranes (Basel). 2022;12(7):716.

    Article  CAS  PubMed  Google Scholar 

  58. Yang H, Cong M, Huang W, Chen J, Zhang M, Gu X, Sun C, Yang H. The effect of human bone marrow mesenchymal stem cell-derived exosomes on cartilage repair in rabbits. Stem Cells Int. 2022;2022:5760107.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Yahao G, Xinjia W. The role and mechanism of exosomes from umbilical cord mesenchymal stem cells in inducing osteogenesis and preventing osteoporosis. Cell Transplant. 2021;30:9636897211057464.

    Article  PubMed  Google Scholar 

  60. Liang B, Liang JM, Ding JN, Xu J, Xu JG, Chai YM. 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(1):335.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Maity S, Das F, Ghosh-Choudhury N, Kasinath BS, Ghosh CG. High glucose increases miR-214 to power a feedback loop involving PTEN and the Akt/mTORC1 signaling axis. FEBS Lett. 2019;593(16):2261–72.

    Article  CAS  PubMed  Google Scholar 

  62. Liu L, Guo S, Shi W, Liu Q, Huo F, Wu Y, Tian W. Bone marrow mesenchymal stem cell-derived small extracellular vesicles promote periodontal regeneration. Tissue Eng Part A. 2021;27(13–14):962–76.

    Article  CAS  PubMed  Google Scholar 

  63. Zhang Y, Xiong Y, Chen X, Chen C, Zhu Z, Li L. Therapeutic effect of bone marrow mesenchymal stem cells pretreated with acetylsalicylic acid on experimental periodontitis in rats. Int Immunopharmacol. 2018;54:320–8.

    Article  CAS  PubMed  Google Scholar 

  64. Filipowska J, Tomaszewski KA, Niedzwiedzki L, Walocha JA, Niedzwiedzki T. The role of vasculature in bone development, regeneration and proper systemic functioning. Angiogenesis. 2017;20(3):291–302.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Huang J, Zhao L, Xing L, Chen D. MicroRNA-204 regulates Runx2 protein expression and mesenchymal progenitor cell differentiation. Stem Cells. 2010;28(2):357–64.

    Article  PubMed  Google Scholar 

  66. Xu S, Cecilia Santini G, De Veirman K, Vande Broek I, Leleu X, De Becker A, Van Camp B, Vanderkerken K, Van Riet I. Upregulation of miR-135b is involved in the impaired osteogenic differentiation of mesenchymal stem cells derived from multiple myeloma patients. PLoS ONE. 2013;8(11):e79752.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Kim YJ, Bae SW, Yu SS, Bae YC, Jung JS. miR-196a regulates proliferation and osteogenic differentiation in mesenchymal stem cells derived from human adipose tissue. J Bone Miner Res. 2009;24(5):816–25.

    Article  CAS  PubMed  Google Scholar 

  68. Loi F, Cordova LA, Pajarinen J, Lin TH, Yao Z, Goodman SB. Inflammation, fracture and bone repair. Bone. 2016;86:119–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Chen Z, Chen L, Liu R, Lin Y, Chen S, Lu S, Lin Z, Chen Z, Wu C, Xiao Y. The osteoimmunomodulatory property of a barrier collagen membrane and its manipulation via coating nanometer-sized bioactive glass to improve guided bone regeneration. Biomater Sci. 2018;6(5):1007–19.

    Article  CAS  PubMed  Google Scholar 

  70. Li R, Li D, Wang H, Chen K, Wang S, Xu J, Ji P. Exosomes from adipose-derived stem cells regulate M1/M2 macrophage phenotypic polarization to promote bone healing via miR-451a/MIF. Stem Cell Res Ther. 2022;13(1):149.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Wu P, Zhang B, Ocansey DKW, Xu W, Qian H. Extracellular vesicles: a bright star of nanomedicine. Biomaterials. 2021;269:120467.

    Article  CAS  PubMed  Google Scholar 

  72. Saparov A, Ogay V, Nurgozhin T, Jumabay M, Chen WC. Preconditioning of human mesenchymal stem cells to enhance their regulation of the immune response. Stem Cells Int. 2016;2016:3924858.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Gorgun C, Ceresa D, Lesage R, Villa F, Reverberi D, Balbi C, Santamaria S, Cortese K, Malatesta P, Geris L, et al. Dissecting the effects of preconditioning with inflammatory cytokines and hypoxia on the angiogenic potential of mesenchymal stromal cell (MSC)-derived soluble proteins and extracellular vesicles (EVs). Biomaterials. 2021;269:120633.

    Article  CAS  PubMed  Google Scholar 

  74. Ejtehadifar M, Shamsasenjan K, Movassaghpour A, Akbarzadehlaleh P, Dehdilani N, Abbasi P, Molaeipour Z, Saleh M. The effect of hypoxia on mesenchymal stem cell biology. Adv Pharm Bull. 2015;5(2):141–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Das R, Jahr H, van Osch GJ, Farrell E. The role of hypoxia in bone marrow-derived mesenchymal stem cells: considerations for regenerative medicine approaches. Tissue Eng Part B Rev. 2010;16(2):159–68.

    Article  CAS  PubMed  Google Scholar 

  76. Yan F, Yao Y, Chen L, Li Y, Sheng Z, Ma G. Hypoxic preconditioning improves survival of cardiac progenitor cells: role of stromal cell derived factor-1alpha-CXCR4 axis. PLoS ONE. 2012;7(7):e37948.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Gao Y, Yuan Z, Yuan X, Wan Z, Yu Y, Zhan Q, Zhao Y, Han J, Huang J, Xiong C, et al. Bioinspired porous microspheres for sustained hypoxic exosomes release and vascularized bone regeneration. Bioact Mater. 2022;14:377–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Liu W, Li L, Rong Y, Qian D, Chen J, Zhou Z, Luo Y, Jiang D, Cheng L, Zhao S, et al. Hypoxic mesenchymal stem cell-derived exosomes promote bone fracture healing by the transfer of miR-126. Acta Biomater. 2020;103:196–212.

    Article  CAS  PubMed  Google Scholar 

  79. Tang TT, Wang B, Wu M, Li ZL, Feng Y, Cao JY, Yin D, Liu H, Tang RN, Crowley SD, et al. Extracellular vesicle-encapsulated IL-10 as novel nanotherapeutics against ischemic AKI. Sci Adv. 2020;6(33):eaaz748.

    Article  Google Scholar 

  80. Liu W, Rong Y, Wang J, Zhou Z, Ge X, Ji C, Jiang D, Gong F, Li L, Chen J, et al. Exosome-shuttled miR-216a-5p from hypoxic preconditioned mesenchymal stem cells repair traumatic spinal cord injury by shifting microglial M1/M2 polarization. J Neuroinflammation. 2020;17(1):47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Wang J, Wu H, Peng Y, Zhao Y, Qin Y, Zhang Y, Xiao Z. Hypoxia adipose stem cell-derived exosomes promote high-quality healing of diabetic wound involves activation of PI3K/Akt pathways. J Nanobiotechnol. 2021;19(1):202.

    Article  CAS  Google Scholar 

  82. Zhu LP, Tian T, Wang JY, He JN, Chen T, Pan M, Xu L, Zhang HX, Qiu XT, Li CC, et al. Hypoxia-elicited mesenchymal stem cell-derived exosomes facilitates cardiac repair through miR-125b-mediated prevention of cell death in myocardial infarction. Theranostics. 2018;8(22):6163–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Zhang CS, Shao K, Liu CW, Li CJ, Yu BT. Hypoxic preconditioning BMSCs-exosomes inhibit cardiomyocyte apoptosis after acute myocardial infarction by upregulating microRNA-24. Eur Rev Med Pharmacol Sci. 2019;23(15):6691–9.

    PubMed  Google Scholar 

  84. Cheng H, Chang S, Xu R, Chen L, Song X, Wu J, Qian J, Zou Y, Ma J. Hypoxia-challenged MSC-derived exosomes deliver miR-210 to attenuate post-infarction cardiac apoptosis. Stem Cell Res Ther. 2020;11(1):224.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Gonzalez-King H, Garcia NA, Ontoria-Oviedo I, Ciria M, Montero JA, Sepulveda P. Hypoxia inducible factor-1alpha potentiates jagged 1-mediated angiogenesis by mesenchymal stem cell-derived exosomes. Stem Cells. 2017;35(7):1747–59.

    Article  CAS  PubMed  Google Scholar 

  86. Zhang B, Tian X, Qu Z, Hao J, Zhang W. Hypoxia-preconditioned extracellular vesicles from mesenchymal stem cells improve cartilage repair in osteoarthritis. Membranes (Basel). 2022;12(2):225.

    Article  PubMed  Google Scholar 

  87. Yuan N, Ge Z, Ji W, Li J. Exosomes secreted from hypoxia-preconditioned mesenchymal stem cells prevent steroid-induced osteonecrosis of the femoral head by promoting angiogenesis in rats. Biomed Res Int. 2021;2021:6655225.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Lan YW, Choo KB, Chen CM, Hung TH, Chen YB, Hsieh CH, Kuo HP, Chong KY. Hypoxia-preconditioned mesenchymal stem cells attenuate bleomycin-induced pulmonary fibrosis. Stem Cell Res Ther. 2015;6:97.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Hawkins KE, Sharp TV, McKay TR. The role of hypoxia in stem cell potency and differentiation. Regen Med. 2013;8(6):771–82.

    Article  CAS  PubMed  Google Scholar 

  90. Berniakovich I, Giorgio M. Low oxygen tension maintains multipotency, whereas normoxia increases differentiation of mouse bone marrow stromal cells. Int J Mol Sci. 2013;14(1):2119–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Ahmed NE, Murakami M, Kaneko S, Nakashima M. The effects of hypoxia on the stemness properties of human dental pulp stem cells (DPSCs). Sci Rep. 2016;6:35476.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Hung SP, Ho JH, Shih YR, Lo T, Lee OK. Hypoxia promotes proliferation and osteogenic differentiation potentials of human mesenchymal stem cells. J Orthop Res. 2012;30(2):260–6.

    Article  PubMed  Google Scholar 

  93. Semenza GL. Targeting hypoxia-inducible factor 1 to stimulate tissue vascularization. J Investig Med. 2016;64(2):361–3.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Chen Z, Lu M, Zhang Y, Wang H, Zhou J, Zhou M, Zhang T, Song J. Oxidative stress state inhibits exosome secretion of hPDLCs through a specific mechanism mediated by PRMT1. J Periodontal Res. 2022;57(6):1101–15.

    Article  CAS  PubMed  Google Scholar 

  95. Ferreira JR, Teixeira GQ, Santos SG, Barbosa MA, Almeida-Porada G, Goncalves RM. Mesenchymal stromal cell secretome: influencing therapeutic potential by cellular pre-conditioning. Front Immunol. 2018;9:2837.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Zhang S, Jiang L, Hu H, Wang H, Wang X, Jiang J, Ma Y, Yang J, Hou Y, Xie D, et al. Pretreatment of exosomes derived from hUCMSCs with TNF-alpha ameliorates acute liver failure by inhibiting the activation of NLRP3 in macrophage. Life Sci. 2020;246:117401.

    Article  CAS  PubMed  Google Scholar 

  97. Liang YC, Wu YP, Li XD, Chen SH, Ye XJ, Xue XY, Xu N. TNF-alpha-induced exosomal miR-146a mediates mesenchymal stem cell-dependent suppression of urethral stricture. J Cell Physiol. 2019;234(12):23243–55.

    Article  CAS  PubMed  Google Scholar 

  98. Nakao Y, Fukuda T, Zhang Q, Sanui T, Shinjo T, Kou X, Chen C, Liu D, Watanabe Y, Hayashi C, et al. Exosomes from TNF-alpha-treated human gingiva-derived MSCs enhance M2 macrophage polarization and inhibit periodontal bone loss. Acta Biomater. 2021;122:306–24.

    Article  CAS  PubMed  Google Scholar 

  99. Wang R, Xu B. TGF-beta1-modified MSC-derived exosomal miR-135b attenuates cartilage injury via promoting M2 synovial macrophage polarization by targeting MAPK6. Cell Tissue Res. 2021;384(1):113–27.

    Article  CAS  PubMed  Google Scholar 

  100. 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(21–22):1212–20.

    Article  CAS  PubMed  Google Scholar 

  101. Kim M, Shin DI, Choi BH, Min BH. Exosomes from IL-1beta-primed mesenchymal stem cells inhibited IL-1beta- and TNF-alpha-mediated inflammatory responses in osteoarthritic SW982 cells. Tissue Eng Regen Med. 2021;18(4):525–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Mead B, Chamling X, Zack DJ, Ahmed Z, Tomarev S. TNFalpha-mediated priming of mesenchymal stem cells enhances their neuroprotective effect on retinal ganglion cells. Invest Ophthalmol Vis Sci. 2020;61(2):6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Liu K, Cai GL, Zhuang Z, Pei SY, Xu SN, Wang YN, Wang H, Wang X, Cui C, Sun MC, et al. Interleukin-1beta-treated mesenchymal stem cells inhibit inflammation in hippocampal astrocytes through Exosome-activated Nrf-2 signaling. Int J Nanomed. 2021;16:1423–34.

    Article  Google Scholar 

  104. Wang S, Umrath F, Cen W, Salgado AJ, Reinert S, Alexander D. Pre-conditioning with IFN-gamma and hypoxia enhances the angiogenic potential of iPSC-derived MSC secretome. Cells. 2022;11(6):988.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Cecerska-Heryc E, Goszka M, Serwin N, Roszak M, Grygorcewicz B, Heryc R, Dolegowska B. Applications of the regenerative capacity of platelets in modern medicine. Cytokine Growth Fact Rev. 2022;64:84–94.

    Article  CAS  Google Scholar 

  106. Mirabel C, Puente-Massaguer E, Del Mazo-Barbara A, Reyes B, Morton P, Godia F, Vives J. Stability enhancement of clinical grade multipotent mesenchymal stromal cell-based products. J Transl Med. 2018;16(1):291.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Jing H, Zhang X, Luo K, Luo Q, Yin M, Wang W, Zhu Z, Zheng J, He X. miR-381-abundant small extracellular vesicles derived from kartogenin-preconditioned mesenchymal stem cells promote chondrogenesis of MSCs by targeting TAOK1. Biomaterials. 2020;231:119682.

    Article  CAS  PubMed  Google Scholar 

  108. Liu C, Li Y, Yang Z, Zhou Z, Lou Z, Zhang Q. Kartogenin enhances the therapeutic effect of bone marrow mesenchymal stem cells derived exosomes in cartilage repair. Nanomedicine (Lond). 2020;15(3):273–88.

    Article  CAS  PubMed  Google Scholar 

  109. Qiu B, Xu X, Yi P, Hao Y. Curcumin reinforces MSC-derived exosomes in attenuating osteoarthritis via modulating the miR-124/NF-kB and miR-143/ROCK1/TLR9 signalling pathways. J Cell Mol Med. 2020;24(18):10855–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Joo HS, Suh JH, Lee HJ, Bang ES, Lee JM. Current knowledge and future perspectives on mesenchymal stem cell-derived exosomes as a new therapeutic agent. Int J Mol Sci. 2020;21(3):727.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Cheng J, Sun Y, Ma Y, Ao Y, Hu X, Meng Q. Engineering of MSC-derived exosomes: a promising cell-free therapy for osteoarthritis. Membranes (Basel). 2022;12(8):739.

    Article  CAS  PubMed  Google Scholar 

  112. Hassanzadeh A, Rahman HS, Markov A, Endjun JJ, Zekiy AO, Chartrand MS, Beheshtkhoo N, Kouhbanani MAJ, Marofi F, Nikoo M, et al. Mesenchymal stem/stromal cell-derived exosomes in regenerative medicine and cancer; overview of development, challenges, and opportunities. Stem Cell Res Ther. 2021;12(1):297.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Chen S, Tang Y, Liu Y, Zhang P, Lv L, Zhang X, Jia L, Zhou Y. Exosomes derived from miR-375-overexpressing human adipose mesenchymal stem cells promote bone regeneration. Cell Prolif. 2019;52(5):e12669.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Liu W, Yu M, Chen F, Wang L, Ye C, Chen Q, Zhu Q, Xie D, Shao M, Yang L. A novel delivery nanobiotechnology: engineered miR-181b exosomes improved osteointegration by regulating macrophage polarization. J Nanobiotechnol. 2021;19(1):269.

    Article  CAS  Google Scholar 

  115. Tao SC, Yuan T, Zhang YL, Yin WJ, Guo SC, Zhang CQ. Exosomes derived from miR-140-5p-overexpressing human synovial mesenchymal stem cells enhance cartilage tissue regeneration and prevent osteoarthritis of the knee in a rat model. Theranostics. 2017;7(1):180–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Huang CC, Kang M, Lu Y, Shirazi S, Diaz JI, Cooper LF, Gajendrareddy P, Ravindran S. Functionally engineered extracellular vesicles improve bone regeneration. Acta Biomater. 2020;109:182–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Li F, Wu J, Li D, Hao L, Li Y, Yi D, Yeung KWK, Chen D, Lu WW, Pan H, et al. Engineering stem cells to produce exosomes with enhanced bone regeneration effects: an alternative strategy for gene therapy. J Nanobiotechnol. 2022;20(1):135.

    Article  CAS  Google Scholar 

  118. Li H, Liu D, Li C, Zhou S, Tian D, Xiao D, Zhang H, Gao F, Huang J. Exosomes secreted from mutant-HIF-1alpha-modified bone-marrow-derived mesenchymal stem cells attenuate early steroid-induced avascular necrosis of femoral head in rabbit. Cell Biol Int. 2017;41(12):1379–90.

    Article  CAS  PubMed  Google Scholar 

  119. Haney MJ, Klyachko NL, Zhao Y, Gupta R, Plotnikova EG, He Z, Patel T, Piroyan A, Sokolsky M, Kabanov AV, et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J Control Release. 2015;207:18–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Piffoux M, Volatron J, Cherukula K, Aubertin K, Wilhelm C, Silva AKA, Gazeau F. Engineering and loading therapeutic extracellular vesicles for clinical translation: a data reporting frame for comparability. Adv Drug Deliv Rev. 2021;178:113972.

    Article  CAS  PubMed  Google Scholar 

  121. Yerneni SS, Adamik J, Weiss LE, Campbell PG. Cell trafficking and regulation of osteoblastogenesis by extracellular vesicle associated bone morphogenetic protein 2. J Extracell Vesicles. 2021;10(12):e12155.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Zha Y, Li Y, Lin T, Chen J, Zhang S, Wang J. Progenitor cell-derived exosomes endowed with VEGF plasmids enhance osteogenic induction and vascular remodeling in large segmental bone defects. Theranostics. 2021;11(1):397–409.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Luo ZW, Li FX, Liu YW, Rao SS, Yin H, Huang J, Chen CY, Hu Y, Zhang Y, Tan YJ, et al. Aptamer-functionalized exosomes from bone marrow stromal cells target bone to promote bone regeneration. Nanoscale. 2019;11(43):20884–92.

    Article  CAS  PubMed  Google Scholar 

  124. Hu Y, Li X, Zhang Q, Gu Z, Luo Y, Guo J, Wang X, Jing Y, Chen X, Su J. Exosome-guided bone targeted delivery of Antagomir-188 as an anabolic therapy for bone loss. Bioact Mater. 2021;6(9):2905–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Zhang M, Li Y, Feng T, Li R, Wang Z, Zhang L, Yin P, Tang P. Bone engineering scaffolds with exosomes: a promising strategy for bone defects repair. Front Bioeng Biotechnol. 2022;10:920378.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Glenske K, Donkiewicz P, Kowitsch A, Milosevic-Oljaca N, Rider P, Rofall S, Franke J, Jung O, Smeets R, Schnettler R, et al. Applications of metals for bone regeneration. Int J Mol Sci. 2018;19(3):826.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Lu H, Zhang Y, Xiong S, Zhou Y, Xiao L, Ma Y, Xiao Y, Wang X. Modulatory role of silver nanoparticles and mesenchymal stem cell-derived exosome-modified barrier membrane on macrophages and osteogenesis. Front Chem. 2021;9:699802.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Zhai M, Zhu Y, Yang M, Mao C. Human mesenchymal stem Cell derived exosomes enhance cell-free bone regeneration by altering their miRNAs profiles. Adv Sci (Weinh). 2020;7(19):2001334.

    Article  CAS  PubMed  Google Scholar 

  129. Fernandes JS, Gentile P, Pires RA, Reis RL, Hatton PV. Multifunctional bioactive glass and glass-ceramic biomaterials with antibacterial properties for repair and regeneration of bone tissue. Acta Biomater. 2017;59:2–11.

    Article  CAS  PubMed  Google Scholar 

  130. Baino F, Fiorilli S, Vitale-Brovarone C. Bioactive glass-based materials with hierarchical porosity for medical applications: review of recent advances. Acta Biomater. 2016;42:18–32.

    Article  CAS  PubMed  Google Scholar 

  131. Liu A, Lin D, Zhao H, Chen L, Cai B, Lin K, Shen SG. Optimized BMSC-derived osteoinductive exosomes immobilized in hierarchical scaffold via lyophilization for bone repair through Bmpr2/Acvr2b competitive receptor-activated Smad pathway. Biomaterials. 2021;272:120718.

    Article  CAS  PubMed  Google Scholar 

  132. Bohner M, Santoni BLG, Dobelin N. beta-tricalcium phosphate for bone substitution: synthesis and properties. Acta Biomater. 2020;113:23–41.

    Article  CAS  PubMed  Google Scholar 

  133. 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):136.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Ying C, Wang R, Wang Z, Tao J, Yin W, Zhang J, Yi C, Qi X, Han D. BMSC-exosomes carry mutant HIF-1alpha for improving angiogenesis and osteogenesis in critical-sized calvarial defects. Front Bioeng Biotechnol. 2020;8:565561.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Wu J, Chen L, Wang R, Song Z, Shen Z, Zhao Y, Huang S, Lin Z. Exosomes secreted by stem cells from human exfoliated deciduous teeth promote alveolar bone defect repair through the regulation of angiogenesis and osteogenesis. ACS Biomater Sci Eng. 2019;5(7):3561–71.

    Article  CAS  PubMed  Google Scholar 

  136. Lei F, Li M, Lin T, Zhou H, Wang F, Su X. Treatment of inflammatory bone loss in periodontitis by stem cell-derived exosomes. Acta Biomater. 2022;141:333–43.

    Article  CAS  PubMed  Google Scholar 

  137. Teotia AK, Qayoom I, Singh P, Mishra A, Jaiman D, Seppala J, Lidgren L, Kumar A. Exosome-functionalized ceramic bone substitute promotes critical-sized bone defect repair in rats. ACS Appl Bio Mater. 2021;4(4):3716–26.

    Article  CAS  PubMed  Google Scholar 

  138. Pishavar E, Luo H, Naserifar M, Hashemi M, Toosi S, Atala A, Ramakrishna S, Behravan J. Advanced hydrogels as exosome delivery systems for osteogenic differentiation of MSCs: application in bone regeneration. Int J Mol Sci. 2021;22(12):6203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Khayambashi P, Iyer J, Pillai S, Upadhyay A, Zhang Y, Tran SD. Hydrogel encapsulation of mesenchymal stem cells and their derived exosomes for tissue engineering. Int J Mol Sci. 2021;22(2):684.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Zhai P, Peng X, Li B, Liu Y, Sun H, Li X. The application of hyaluronic acid in bone regeneration. Int J Biol Macromol. 2020;151:1224–39.

    Article  CAS  PubMed  Google Scholar 

  141. Zhang Y, Xie Y, Hao Z, Zhou P, Wang P, Fang S, Li L, Xu S, Xia Y. Umbilical mesenchymal stem cell-derived exosome-encapsulated hydrogels accelerate bone repair by enhancing angiogenesis. ACS Appl Mater Interfaces. 2021;13(16):18472–87.

    Article  CAS  PubMed  Google Scholar 

  142. Yang S, Zhu B, Yin P, Zhao L, Wang Y, Fu Z, Dang R, Xu J, Zhang J, Wen N. Integration of human umbilical cord mesenchymal stem cells-derived exosomes with hydroxyapatite-embedded hyaluronic acid-alginate hydrogel for bone regeneration. ACS Biomater Sci Eng. 2020;6(3):1590–602.

    Article  CAS  PubMed  Google Scholar 

  143. Wu D, Qin H, Wang Z, Yu M, Liu Z, Peng H, Liang L, Zhang C, Wei X. Bone mesenchymal stem cell-derived sEV-encapsulated thermosensitive hydrogels accelerate osteogenesis and angiogenesis by release of exosomal miR-21. Front Bioeng Biotechnol. 2021;9:829136.

    Article  PubMed  Google Scholar 

  144. Shen Z, Kuang S, Zhang Y, Yang M, Qin W, Shi X, Lin Z. Chitosan hydrogel incorporated with dental pulp stem cell-derived exosomes alleviates periodontitis in mice via a macrophage-dependent mechanism. Bioact Mater. 2020;5(4):1113–26.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Ma S, Wu J, Hu H, Mu Y, Zhang L, Zhao Y, Bian X, Jing W, Wei P, Zhao B, et al. Novel fusion peptides deliver exosomes to modify injectable thermo-sensitive hydrogels for bone regeneration. Mater Today Bio. 2022;13:100195.

    Article  CAS  PubMed  Google Scholar 

  146. Jing X, Wang S, Tang H, Li D, Zhou F, Xin L, He Q, Hu S, Zhang T, Chen T, et al. Dynamically bioresponsive DNA hydrogel incorporated with dual-functional stem cells from apical papilla-derived exosomes promotes diabetic bone regeneration. ACS Appl Mater Interfaces. 2022;14(14):16082–99.

    Article  CAS  PubMed  Google Scholar 

  147. Wang L, Wang J, Zhou X, Sun J, Zhu B, Duan C, Chen P, Guo X, Zhang T, Guo H. A new self-healing hydrogel containing hucMSC-derived exosomes promotes bone regeneration. Front Bioeng Biotechnol. 2020;8:564731.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Kang Y, Xu C, Meng L, Dong X, Qi M, Jiang D. Exosome-functionalized magnesium-organic framework-based scaffolds with osteogenic, angiogenic and anti-inflammatory properties for accelerated bone regeneration. Bioact Mater. 2022;18:26–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Zhang B, Huang J, Liu J, Lin F, Ding Z, Xu J. Injectable composite hydrogel promotes osteogenesis and angiogenesis in spinal fusion by optimizing the bone marrow mesenchymal stem cell microenvironment and exosomes secretion. Mater Sci Eng C Mater Biol Appl. 2021;123:111782.

    Article  CAS  PubMed  Google Scholar 

  150. 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(6):5240–54.

    Article  CAS  PubMed  Google Scholar 

  151. Swanson WB, Zhang Z, Xiu K, Gong T, Eberle M, Wang Z, Ma PX. Scaffolds with controlled release of pro-mineralization exosomes to promote craniofacial bone healing without cell transplantation. Acta Biomater. 2020;118:215–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Zhang Y, Huo M, Wang Y, Xiao L, Wu J, Ma Y, Zhang D, Lang X, Wang X. A tailored bioactive 3D porous poly(lactic-acid)-exosome scaffold with osteo-immunomodulatory and osteogenic differentiation properties. J Biol Eng. 2022;16(1):22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Fan L, Guan P, Xiao C, Wen H, Wang Q, Liu C, Luo Y, Ma L, Tan G, Yu P, et al. Exosome-functionalized polyetheretherketone-based implant with immunomodulatory property for enhancing osseointegration. Bioact Mater. 2021;6(9):2754–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Meng W, He C, Hao Y, Wang L, Li L, Zhu G. Prospects and challenges of extracellular vesicle-based drug delivery system: considering cell source. Drug Deliv. 2020;27(1):585–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Zhang Y, Liu Y, Liu H, Tang WH. Exosomes: biogenesis, biologic function and clinical potential. Cell Biosci. 2019;9:19.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Momen-Heravi F, Balaj L, Alian S, Mantel PY, Halleck AE, Trachtenberg AJ, Soria CE, Oquin S, Bonebreak CM, Saracoglu E, et al. Current methods for the isolation of extracellular vesicles. Biol Chem. 2013;394(10):1253–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Li P, Kaslan M, Lee SH, Yao J, Gao Z. Progress in exosome isolation techniques. Theranostics. 2017;7(3):789–804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Phan J, Kumar P, Hao D, Gao K, Farmer D, Wang A. Engineering mesenchymal stem cells to improve their exosome efficacy and yield for cell-free therapy. J Extracell Vesicles. 2018;7(1):1522236.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Herrmann IK, Wood MJA, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nat Nanotechnol. 2021;16(7):748–59.

    Article  CAS  PubMed  Google Scholar 

  160. Wang W, Liang X, Zheng K, Ge G, Chen X, Xu Y, Bai J, Pan G, Geng D. Horizon of exosome-mediated bone tissue regeneration: The all-rounder role in biomaterial engineering. Mater Today Bio. 2022;16:100355.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Vader P, Mol EA, Pasterkamp G, Schiffelers RM. Extracellular vesicles for drug delivery. Adv Drug Deliv Rev. 2016;106(Pt A):148–56.

    Article  CAS  PubMed  Google Scholar 

  162. Lai RC, Yeo RW, Tan KH, Lim SK. Exosomes for drug delivery - a novel application for the mesenchymal stem cell. Biotechnol Adv. 2013;31(5):543–51.

    Article  CAS  PubMed  Google Scholar 

  163. Li X, Zheng Y, Hou L, Zhou Z, Huang Y, Zhang Y, Jia L, Li W. Exosomes derived from maxillary BMSCs enhanced the osteogenesis in iliac BMSCs. Oral Dis. 2020;26(1):131–44.

    Article  PubMed  Google Scholar 

  164. Xu T, Luo Y, Wang J, Zhang N, Gu C, Li L, Qian D, Cai W, Fan J, Yin G. Exosomal miRNA-128-3p from mesenchymal stem cells of aged rats regulates osteogenesis and bone fracture healing by targeting Smad5. J Nanobiotechnol. 2020;18(1):47.

    Article  CAS  Google Scholar 

  165. Borger V, Bremer M, Ferrer-Tur R, Gockeln L, Stambouli O, Becic A, Giebel B. Mesenchymal stem/stromal cell-derived extracellular vesicles and their potential as novel immunomodulatory therapeutic agents. Int J Mol Sci. 2017;18(7):1450.

    Article  PubMed  PubMed Central  Google Scholar 

  166. Henkel J, Woodruff MA, Epari DR, Steck R, Glatt V, Dickinson IC, Choong PF, Schuetz MA, Hutmacher DW. Bone regeneration based on tissue engineering conceptions: a 21st century perspective. Bone Res. 2013;1(3):216–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Merimi M, El-Majzoub R, Lagneaux L, Moussa Agha D, Bouhtit F, Meuleman N, Fahmi H, Lewalle P, Fayyad-Kazan M, Najar M. The therapeutic potential of mesenchymal stromal cells for regenerative medicine: current knowledge and future understandings. Front Cell Dev Biol. 2021;9:661532.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (81901006), Guangdong Basic and Applied Basic Research Foundation (2020A1515110051), Scientific Research Talent Cultivation Project of Stomatological Hospital, Southern Medical University (RC202005) and Science Research Cultivation Program of Stomatological Hospital, Southern Medical University (PY2020002).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the conception and the main idea of the work. XZ, LC, YL and ZM drafted the manuscript. YL and ZM designed the tables and figures. XZ and LC supervised and revised the work. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Li Cui or Xinyuan Zhao.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they are no competing interests.

Additional information

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 licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. 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 in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lu, Y., Mai, Z., Cui, L. et al. Engineering exosomes and biomaterial-assisted exosomes as therapeutic carriers for bone regeneration. Stem Cell Res Ther 14, 55 (2023). https://doi.org/10.1186/s13287-023-03275-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-023-03275-x

Keywords