Open access peer-reviewed chapter

Potential Therapeutic Applications of Exosomes in Bone Regenerative Medicine

Written By

Jiazhao Yang, Wanbo Zhu, Jinsen Lu, Kai Xie, Shiyuan Fang and Lixin Kan

Submitted: 08 June 2018 Reviewed: 22 August 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.81069

From the Edited Volume

Osteogenesis and Bone Regeneration

Edited by Haisheng Yang

Chapter metrics overview

1,311 Chapter Downloads

View Full Metrics

Abstract

The ability of bone regeneration is relatively robust, which is crucial for fracture healing, but delayed healing and nonunion are still common problems in clinical practice. Fortunately, exciting results have been achieved for regenerative medicine in recent years, especially in the area of stem cell-based treatment, but all these cell-based approaches face challenging problems, including immune rejection. For this reason, exosomes, stem cell-derived small vesicles of endocytic origin, have attracted the attention of many investigators in the field of bone regeneration. One of the attractive features of exosomes is that they are small and can travel between cells and deliver bioactive products, including miRNA, mRNA, proteins, and various other factors, to promote bone regeneration, with undetectable immune rejection. In this chapter, we intend to briefly update the recent progressions, and discuss the potential challenges in the target areas. Hopefully, our discussion would be helpful not only for the clinicians and the researchers in the specific disciplines but also for the general audiences as well.

Keywords

  • exosome
  • stem cell
  • fracture healing
  • osteogenesis
  • bone regeneration

1. Introduction

Fractures are common traumatic injuries during the entire human history. Both traditional and modern medicine have kept on exploring and researching on many potential treatments. Despite these efforts and relatively robust regenerative capacity of bone, currently, there are still about 5–10% fracture patients face delayed fracture healing and even nonunion, which has a great negative impact on the quality of life of patients as well as their families [1]. Surgical intervention with autologous bone graft seems to be the preferred method for such complication, but the secondary trauma and the limited resources of grafting bone make this approach still unsatisfactory [2, 3]. Other methods, including active substance injection and bone marrow transplantation, are also used clinically but they face their own challenges, including the effectiveness, safety and immune rejection [4, 5]. Therefore, how to promote fracture healing efficiently and safely is still the major focus of recent research in regenerative medicine for bone.

Normal bone regeneration is a complex but well-orchestrated physiological process that includes the initiation of ossification, osteoinduction, and osteogenesis [6, 7, 8, 9]. Specifically, when bone injury occurs, a series of signaling pathways is activated, which, in turn, leads to angiogenesis and other downstream events, and these together establish a favorable microenvironment, which set the stage for stem cell based fracture healing/regeneration [10]. Within this microenvironment, abundant blood vessels accelerate the metabolism while bringing a large number of multipotential stem cells [11, 12]. On the other hand, the mononuclear phagocyte system from the blood differentiates into osteoclasts in the newly established microenvironment, and the bone resorption, in turn, specifically stimulates the bone re-modeling process [13, 14]. During the stereotyped osteogenesis process, stem cells proliferate and differentiate into osteoblasts and migrate to areas of bone defects and bone resorption, secreting collagen matrices [7, 15, 16, 17], and then immature osteoblasts produce bone matrix containing calcium and phosphate to promote mineralization [18]. Of note, new blood vessels in the fracture microenvironment can also bring essential nutrients and mineral salt for fracture healing, improving the efficiency of osteogenic differentiation and bone regeneration [19].

Embryonic stem cell transplantation was considered as a potential promising treatment for tissue repair; however, due to the limitation of donor cells and biosafety issues, its clinical application has not been widely accepted [20, 21, 22, 23]. Recently, it has been recognized that adult bone marrow-derived mesenchymal stem cells (BMSCs) might be a better alternative, and moreover, researchers found that BMSCs play an important role in promoting tissue regeneration through paracrine signaling [24, 25], in addition to directly differentiation into bony tissue. This paracrine effect, mediated by signaling molecules, transcription factors, and other proteins, regulates a series of signaling pathways involved in bone regeneration.

Interestingly, extracellular vesicle derived from stem cells under specific stimulation can carry specific substances produced by paracrine secretion and transmit to target organs/cells to act as an intercellular communicator [26, 27]. Among all the extracellular vesicles, the particles with the diameter around 40–100 nm are commonly called exosomes. Further study found, that in addition to stem cells, many other cells, such as osteoblasts, can also produce exosomes [28]. The key unanswered question is: could these different cell-derived exosomes promote bone regeneration and accelerate fracture healing? This chapter will focus on this important question.

Advertisement

2. A brief overview of exosome

In 1983, Harding found a lysosomal-like vesicle in reticulocytes of rats. It was found that transferrin was internalized by this vesicle and its receptors also recycled back to the plasma membrane through endocytosis [29]. In 1987, such vesicle-like structures were also found in the culture medium of sheep red blood cells cultured in vitro by Johnstone, and the vesicles were later named as exosomes [30]. It is now accepted that the extracellular vesicles secreted by cells could be generally classified as microvesicles, apoptotic bodies, and exosomes, on the basis of the size, cellular origin, content, and biological function [31, 32]. Currently, the exosomes are extensively studied. Exosomes, normally 40–100 nm in diameter, have been defined as a type of extracellular vesicles with unique biological features and morphology (flat or cup-shape under electron microscope) [33, 34](Figure 1). The formation of exosome is essentially the encapsulation of bioactive substances, including proteins and nucleic acids, into multivesicular bodies with the help of endosomal sorting complex in the cells [35, 36]. The newly formed exosomes inside the cell are transported and fused with the plasma membrane and eventually released into the extracellular matrix [37, 38].

Figure 1.

Electron-microscopic observation of whole-mounted exosomes purified from mouse dendritic cells. Arrows indicate exosomes, arrowheads point to smaller nonexosomal vesicles. Scale bar = 100 nm. (Quote from Théry et al. [33].)

It is now known that numerous different type of cells, including dendritic cells, mast cells, lymphocytes, neurons, and endothelial cells secrete exosomes [39, 40, 41, 42, 43], which are found in blood, amniotic fluid, urine, malignant ascites, and other body fluids such as bile [44, 45, 46, 47]. The key features of exosomes as intercellular communicators is due to the fact that they are able to selectively carry the contents of the parent cells and act on target cells [31, 38]. In 2007, Valadi found that exosomes contain RNA, which indicated exosomes might regulate genetic information flow [48]. In recent years, many studies have found that a variety of cell-derived exosomes contain mRNA and miRNA and play an important role in cell-to-cell signaling [48, 49, 50]. Therefore, the transport of RNA and active proteins through exosomes provides a novel pathway for activating target cell and initiating and propagating downstream signaling pathways. For example, in 2012, Cantaluppi discovered that microvesicles from epithelial progenitor-derived cell initiated renal-regeneration procedures by carrying miRNAs and acting on target cells, reversing focal ischemic lesions [51].

The regenerative effects of exosomes have been validated in other tissues and organs, including the heart, lungs, kidneys, and brain [52, 53, 54]. For example, in a mouse model of myocardial infarction, treatment of exosomes can improve cardiac epicardial remodeling and increase left ventricular ejection fraction [55]. In hypoxic-induced pulmonary hypertension, exosome treatment inhibits disease progression and protects the lungs from hypertension [56]. In addition, exosome treatment can improve renal function in a mouse model of acute kidney injury [57]. These studies indicate that exosomes have the capacity to promote tissue regeneration, which provides a basis for their potential application in bone regeneration [58].

Advertisement

3. Exosomes in bone regeneration

3.1 The exosomes derived from different cells promote bone regeneration

The mechanism of stem cells in the treatment of diseases has not been fully elucidated; however, it is now commonly accepted that there are two recognized mechanisms: differentiation and paracrine. In fact, it is becoming clearer that paracrine mechanism could be a more important mechanism; therefore, exosomes, as important mediators in paracrine mechanism, have attracted researchers.

Embryonic stem cells are considered to be the ideal materials for regenerative medicine because of their ability of pluripotent differentiation. But later study found that bone marrow mesenchymal stem cell (BMSC) could be a better alternative, i.e., BMSCs are self-renewing mesodermal pluripotent stem cells that can differentiate into osteoblasts, fat cells, nerve cells, and myoblasts [24, 59]. Recent study also found that BMSCs’ roles in inducing angiogenesis, regulating inflammation, inhibiting apoptosis, and regulating osteogenesis differentiation make them desirable for bone regeneration applications [59].

Similarly, the adipose-derived stem cells (ADSCs) can also be osteogenic differentiated to promote bone regeneration, when they have been applied to the bone defects using a composite biological scaffold [60]. In addition, endothelial progenitor cells (EPCs) can differentiate into vascular endothelial cells to generate blood vessels, and promote MSCs osteogenesis in a specific microenvironment [61, 62]. Also, differentiated cells, such as osteoblasts and osteoclasts, also have the ability to promote bone regeneration [15, 26].

More importantly, numerous studies suggest that the above-mentioned cell-derived exosomes all have a certain ability to promote bone regeneration, through regulating bone regeneration procedures such as angiogenesis, osteogenic differentiation, and bone mineral deposition. However, the capacities and regeneration mechanisms of exosomes from different derived cells are somewhat inconsistent, likely due to their different contents.

3.2 Genetic materials carried by exosomes regulate bone regeneration

It was reported that stem cell-derived exosomes can carry genetic materials such as miRNA and mRNA, and share these genetic information between mature bone cells and stem/progenitor cells, which is an important way to promote bone regeneration [63]. MicroRNAs (miRNAs) are thought to be important posttranscriptional regulators of osteoblast-associated osteogenesis and bone remodeling, enabling a range of bone regenerative responses [64, 65]. Interestingly, miRNAs, inside the lipid membrane of exosomes, can avoid the decomposition of immune system; therefore, they exert their effects more efficiently [66].

Many researchers reported that some stem cell-derived exosomal miRNAs have the ability to activate osteogenic differentiation and angiogenesis of target cells and promote bone formation. For example, Xu first found that exosomal miRNA is a regulator of osteoblast differentiation [67]. Similarly, a series of miRNAs, such as let-7a, which could enhance the osteogenic differentiation of stem cells and promote bone regeneration, are significantly upregulated [68]. These data all demonstrated that stem cell-derived exosomes could promote bone regeneration by carrying specific miRNAs (Table 1).

miRNADerived cellsExpress levelTarget cellsIn vivo evaluationIn vitro evaluationInvolved pathway
Let-7a [67]BMSCsUpregulatedMSCsPromote bone formationPromote osteogenesis and suppress adipogenesis [68]AXIN2
HMGA2
miR-218 [67]BMSCsUpregulatedSMSCsNoneInhibit osteogenic differentiationNone
miR-203 [67]BMSCsUpregulatedBMSCsNonePromote osteoblastic differentiationNone
miR-196a [59]BMSCsUpregulatedBMSCsStimulate bone formationPositively regulated osteogenic genes and osteoblastic differentiation but did not inhibit proliferationNone
miR-27a [59]BMSCsUpregulatedMSCsOverexpression promoted osteogenic differentiationNonePPARγ
miR-206 [59]BMSCsUpregulatedNoneNoneNoneNone
miR-21 [69]BMSCsUpregulatedBMSCs/MSCsAccelerate fracture healingPromote osteogenic differentiationPI3K/AKT
miR-125b [69]BMSCsUpregulatedBMSCsNoneSuppresses the proliferation and osteogenic differentiation of BMSCsNone
miR-10b [72]BMSCsUpregulatedMSCsNonePromote the migration of MSCsNone
miR-221 [72]BMSCs/MSCsDownregulatedMSCsAnti-miR-221 enhances bone healingDownregulation of miR-221 triggers osteogenic differentiationNone
miR-155 [67]BMSCsDownregulatedNoneNoneNoneNone
miR-31 [72]MSCsDownregulatedBMSCsInhibition of miR-31 in MSCs increased bone volume and bone mineral densityInhibit the osteogenic differentiation of MSCsWnt
miR-144 [72]MSCsDownregulatedMSCsNoneInhibit the osteogenic differentiation of MSCsNone

Table 1.

Summary of Exosomal miRNAs and their potential effects on bone metabolism.

Furthermore, many recent studies focus on MSCs-derived exosomes (BMSC-Exo) for bone regeneration. For examples, in CD9−/− mice, BMSC-Exo isolated from culture medium can accelerate fracture healing compared with the control group [69]. In vitro analysis of the exosomes revealed that miR-21, miR-4332 and other osteogenic differentiation-related miRNAs are highly expressed compared to other cell-derived exosomes. Interestingly, mononuclear cell chemotactic protein MCP-1/-3 and stromal cell-derived factor SDF-1, were lower in BMSC-Exo than in the control group [70, 71]. This might suggest that differential distribution of osteogenic differentiation and angiogenesis-related miRNAs in BMSC-Exo. In another study, BMSC-Exo group showed a significant increase in bone formation and repair rate in the model of mouse skull repair, compared with the control group. Similarly, in vitro experiments, BMSC-Exo was showed to activate osteogenic differentiation, increase osteoblast activity, and promote bone formation without inhibiting stem cell proliferation [59].

Further study found that the ability of exosomes to promote bone formation is different even when the parent cells are in different differentiation stages. For example, in vitro experiments demonstrated that the human mesenchymal stem cell-derived exosomes (hMSCs-Exo) from the late differential stage have the strongest osteogenic differentiation ability [67, 72]. Consistently, MiR-31, miR-221, and miR-144 that inhibit osteogenic differentiation have significant decreased levels in late differential stage of hMSCs-Exo, while miR-21, miR-10b, and other miRNAs that contribute to osteogenesis is significantly upregulated [73, 74, 75, 76]. It should be noted that the exosome miRNA’s ability to regulate cell function could be context dependent, especially in the present of inhibitory miRNAs [67, 77]. Therefore, to promote bone regeneration using stem cell-derived exosomes, silencing inhibitory miRNAs may be a problem to be solved.

In addition, some miRNAs carried by other cell-derived exosomes also have the ability to promote bone regeneration. For example, the mineralization-related miR-503-3p is highly expressed in the miRNAs carried by osteoblast-derived exosomes. Interestingly, miR-503-3p also inhibits osteoclast differentiation by mediating RANK expression [78]. Osteoblast-derived exosomes and pre-osteoblasts-derived cells can also carry miRNAs such as let-7a and miR-96a, which have been previously confirmed to be involved in bone remodeling [79]. Similarly, the miR-27a-3p carried by myogenic cell-derived exosomes can also enhance osteogenic differentiation of pre-osteoblasts [80]. In contrast, osteoclast-derived exosomes can carry miRNAs such as miR-214 that inhibit osteogenic differentiation of osteoblasts [81]. Interestingly, in vitro experiments have found that human adipose stem cell-derived exosomes (ASCs-Exo) can increase the osteogenic capacity of target cells by upregulating the mRNA expression of osteogenesis-related genes RUNX2, ALP, and COL1A1, and promote bone formation [82]. In addition, the mRNA of RAB13, an osteoclastic membrane trafficking protein required for bone resorption, is also overrepresented in osteoblast-derived exosome [49].

Overall, cell-derived exosomal miRNAs and mRNAs likely play important roles in bone regeneration, through promoting osteogenic differentiation, angiogenesis and other processes. However, it is unclear whether protein factors are eventually needed to mediate their final effects.

3.3 Key protein factors carried by exosomes regulate bone regeneration

Key factors in stem cell-derived exosomes are known to mediate a series of conserved signaling pathways.

RUNX2 is an important transcription factor that can regulate osteogenesis differentiation, through promoting the differentiation of pluripotent stem cells into osteoblasts and inhibit osteoblast maturation [83]. Consistently, in vivo experiment found that human induced pluripotent mesenchymal stem cell-derived exosomes (hiPS-MSC-Exo) stimulated osteogenic differentiation, promoted angiogenesis, and improved fracture healing rate in animals with the upregulated transcription factors such as RUNX2 [84]. It was also reported that cell derived exosomal miRNAs are critical for upregulation of RUNX2 [85, 86]. Interestingly, RUNX2 directly represses miR-31 expression, which significantly inhibits expression of the osteogenic transcription factors OPN, BSP, Osterix (OSX), and OCN [87].

PI3K-AKT signaling pathway is thought to play an important role in exosomes-mediated bone regeneration because it stimulates osteogenic differentiation and promotes osteogenesis [88, 89]. Consistently, Shabbir et al. found that BMSCs-Exo activates multiple signaling pathways including Akt, Erk1/2, and STAT3 to induce angiogenic responses in fibroblasts [90]. In vitro experiment also found that hiPS-MSC-Exo downregulates inhibitory factor (GSK3β and PTEN) by upregulating PI3K-AKT target genes PDGFA and FGFR1 [91], and activation of PI3K-AKT cascade induces stem cell proliferation and differentiation into osteoblasts, and enhances ALP expression and calcium salt deposition, promoting bone regeneration. In the context of long-term nonunion of the femoral neck fracture or intertrochanteric fracture induces femoral head necrosis, Liu et al. found that iPS-MSCs-Exo activates the PI3K/Akt signaling pathway to increase angiogenesis and reduce bone loss [94].

miRNAs are also important molecules that regulate the PI3K-AKT signaling pathway. For example, miR-21, highly expressed in BMSCs-Exo, is one of the major regulators in stem cell-derived exosomes, which promotes osteogenic differentiation not only by inhibiting SOX2 [92], but also regulating the PI3K-AKT-GSK3β signaling pathway, which, in turn, activates the transcription of RUNX2, and stimulate osteogenic differentiation [93].

Wnt pathway is an important signaling pathway related to bone repair. In this regard, ASCs-Exo pretreated with TNF-α could upregulate Wnt3 expression in stem cells and promote bone regeneration [95, 96]. Zhang et al. also found that human umbilical cord stem cell-derived exosomes induce Wnt4-mediated β-catenin nuclear transport, and induce endothelial cell proliferation, differentiation, and neovascularization [97]. Similarly, BMSCs-Exo also activates the Wnt3a-β-catenin pathway and induces angiogenic capacity of fibroblast [98].

RANKL-RANK signaling is known to be responsible for homeostasis of bone metabolism, which is determined by a dynamic balance between osteoclasts and osteoblast [99]. Interestingly, Nuclear factor kappaB ligand (RANKL) can be encapsulated into osteoblast exosomes, while osteoclast exosomes are enriched with RANK [100]. When RANKL binds to RANK in pre-osteoclasts, TNF receptor-related factors (TRAF) 2, 3, 5, and 6 are recruited, leading to activation of multiple signaling pathways including MAPK and NF-κB, promoting osteoclast differentiation and bone resorption [101]. Moreover, level of RANK-containing exosomes increases in the late stage of osteoclast differentiation, which negatively feedbacks on RANKL-RANK signaling to inhibit osteoclast differentiation [99]. Therefore, RANKL-RANK loop contributes to the homeostasis of bone metabolism and bone regeneration.

Other proteins and cytokines in the exosomes are also involved in promoting bone regeneration process. For example, Martins et al. found that hBMSCs-Exo induced BMP2 upregulation, and BMP2 in turn, promoted stem cell osteogenic differentiation and osteogenesis by cascade activation of transcription factor OSX instead of RUNX2 [65]. Similarly, SPE1 (secreted phosphoprotein 1), integrin-binding sialoprotein and bone gland protein BGLAP (bone g-carboxyglutamate (gla) protein) were also upregulated, which facilitated bone mineralization and other bone regeneration processes. MSCs-Exo is also known to induce high expression of BMP9, transforming growth factor β1 (TGFβ1), vascular endothelial growth factor (VEGF), and platelet-derived growth factor (PDGF) [102]. BMP9 is considered to be an osteogenic factor stronger than BMP2. TGFβ1 and PDGF are known to play roles both in osteogenic differentiation and angiogenesis [103, 104, 105]. Qi et al. also found that hiPSC-MSC-Exo induced high expression of osteopontin, osteocalcin, and type I collagen (COL1), and enhanced bone mineralization [84, 106]. Meanwhile, high expression of phosphorylated protein and bone matrix acidic protein (DMP1) was found in the extracellular matrix (ECM) containing MSCs-Exo, suggesting MSCs-Exo promotes calcium phosphate recruitment and bone mineralization [107].

In addition, exosomes from osteoblast carry transforming growth factor beta receptor II interacting protein1 (TRIP-1), a regulator of osteoblast function. TRIP-1 from the exosomes can bind to type I collagen and promote its mineralized extracellular matrix, therefore bone mineralizing [108]. Sema4D is an osteoclast membrane protein that can be carried in exosomes derived from osteoclasts and acts on the receptor Plexin B on osteoblasts [81]. The Sema4D-Plexin B interaction promotes the release the content of exosomes and accelerates bone formation.

It is worthy to mention that some proteins, though are highly expressed in stem cell-derived exosomes and have the potential for bone regeneration, do not seem to play important roles in exosomes mediated osteogenic or chondrogenic differentiation in different contexts. For example, heat-shock protein 70 (HSP70), which can be used as a marker of BMSCs-Exo, is downregulated in human MSC-Exos and negatively affects osteogenic and chondrogenic differentiation. Similarly, down-regulation of heat shock protein B8 (HSPB8) can reduce the formation of dental pulp stem cells, and osteogenic differentiation ability [109, 110, 111]. Overall, the specific biological mechanisms of some functional proteins to promote fracture healing are not fully understood, and further detailed researches will be needed.

Advertisement

4. Clinical therapeutic applications and limitations

Many studies have shown that stem-derived exosomes in vitro and in vivo activate a series of bone regeneration programs through their selective bioactive substances, which are mainly through osteogenic differentiation, angiogenesis, and bone mineralization. In these applications, the high extracellular matrix binding affinity of stem cell-derived exosomes is a big plus for their clinical application. Recently, some scholars have found that human adipose-derived stem cell-derived exosomes promote fracture healing in animals by binding to polylactic acid-glycolic acid scaffolds [82]. At the same time, the immunomodulatory and anti-inflammatory properties of stem cell-derived exosomes have also attracted the attention of researchers, which could be the potential biological mechanisms for clinical treatment to promote bone regeneration [112, 113].

However, so far there are few examples of clinical trials using exosomes as clinical treatments. At present, exosome clinical application has only been reported in the fields of treatment of chronic kidney disease, type 1 diabetes mellitus (clinical trialNCT02138331), and skin damage (clinical trial NCT02565264) [114]. In the field of bone regeneration, to our best knowledge, there is not any clinical trial, either ongoing or finished. The main reasons for this delay could be logistic, since the separation, acquisition, purification, and identification of exosomes are still in the laboratory stage, and large scale manufacture is still a major practical challenge. In addition, the healing of the fracture will take several months, and how to make the exosomes available constantly in the fracture site for such a long time is also a problem.

Cell culture: The acquisition of a large number of exosomes requires a large number of cells [115]. However, large scale stem cells culture may alter the cell phenotype [116]. Existing cell culture techniques such as bioreactors have expanded the surface area of cell growth, but it is still difficult to perfectly control the conditions of cell growth [117]. As mentioned above, exosomes from different stages of derived cells have different bone regeneration capabilities. However, there are still limitations on how to obtain batch production from the specific stage of the cells.

Purification: Ultracentrifugation and ultrafiltration can be used to obtain purified exosomes in the laboratory, but this technology is difficult to apply on a large scale [118]. The nonspecific precipitation method using polyethyleneglycol (PEG) can solve this problem well, but PEG needs to be removed again in the product, which is technical challenging [119]. The tangential-flow filtration technology based on cell size separation is currently considered promising; however, it is expensive to use and does not separate some biological materials such as DNA [118, 120].

Identification and quality control: Current laboratory identification and quality control methods include direct observation under electron microscopy and biomarkers observation, but none of them can be scaled up easily. The identification and quality control using immunomagnetic capture of exosomal biomarkers through microfluidic technology can speed up the identification process, but it also has a long way to go before this method can be commonly accepted [118, 121].

In summary, the existing technology still has great challenges for large-scale acquisition of purified exosomes.

Advertisement

5. Existing disputes and problems

Whether promoting bone regeneration will indirectly lead to tumor production is a controversy that needs to be tested seriously. In fact, there are some studies have shown that exosomes can promote tumor growth and malignant transformation or inhibit tumor survival [122, 123]. For example, Qi et al. found that BMSCs-Exo can induce osteosarcoma growth by activating the Hedgehog signaling pathway [124]. BMSCs-Exo can induce drug resistance even on the basis of promoting the proliferation and differentiation of myeloma cells and the survival of migration [125, 126]. How to limit the potential tumor-promoting ability of stem cell-derived exosomes is a problem that must be solved before clinical application. However, miR-340 carried by early BMSCs-Exo can inhibit the angiogenic ability of myeloma thus significantly limiting tumor growth [127].

In clinical applications, while the short term activity of pro-osteogenic differentiation in vitro or promotion of bone regeneration is observed by exosomes treatment, the long-term activity that affects the quality of fracture healing or osteophyte formation is unknown. It is also unclear how to stop the biological effects of exosomes when the satisfactory therapeutic effect is achieved. To clarify these issues, at present, it is urgently needed to test exosomes in animal model before we can move on to clinical study.

Advertisement

6. Conclusion

In summary, exosomes with their carried bioactive contents have a capacity to promote bone regeneration through osteogenic differentiation, angiogenesis, and bone mineralization (Figure 2). Hence, exosomes are identified as potential new “acellular” therapeutic application in bone regenerative medicine. However, clinical application of exosomes still faces controversies and challenges, and further researches are needed to elucidate the signaling pathway, molecular mechanism, and long-term clinical effect.

Figure 2.

Main biological mechanism of therapeutic application exosomes in bone regenerative medicine.

Advertisement

Acknowledgments

We appreciate the help from many members of the Kessler Lab. LK was supported in part by national natural science foundation of China (81472087) and natural science foundation of Anhui province (1508085MC45).

Advertisement

Conflict of interest

The authors declare no competing interests.

References

  1. 1. Huang W, Zhang K, Zhu Y, Wang Z, Li Z, Zhang J. Genetic polymorphisms of NOS2 and predisposition to fracture non-union: A case control study based on Han Chinese population. PLoS One. 2018;13:e193673. DOI: 10.1371/journal.pone.0193673
  2. 2. Ehrler DM, Vaccaro AR. The use of allograft bone in lumbar spine surgery. Clinical Orthopaedics and Related Research. 2000:38-45
  3. 3. Stafford PR, Norris BL. Reamer-irrigator-aspirator bone graft and bi Masquelet technique for segmental bone defect nonunions: A review of 25 cases. Injury. 2010;41:S72-S77. DOI: 10.1016/S0020-1383(10)70014-0
  4. 4. Hernigou P, Poignard A, Beaujean F, Rouard H. Percutaneous autologous bone-marrow grafting for nonunions. Influence of the number and concentration of progenitor cells. The Journal of Bone and Joint Surgery. American Volume. 2005;87:1430-1437. DOI: 10.2106/JBJS.D.02215
  5. 5. Kawaguchi H, Oka H, Jingushi S, Izumi T, Fukunaga M, Sato K, et al. A local application of recombinant human fibroblast growth factor 2 for tibial shaft fractures: A randomized, placebo-controlled trial. Journal of Bone and Mineral Research. 2010;25:2735-2743. DOI: 10.1002/jbmr.146
  6. 6. Cornell CN, Lane JM. Current understanding of osteoconduction in bone regeneration. Clinical Orthopaedics and Related Research. 1998;Res:S267-S273. PMID: 9917646
  7. 7. Javed A, Chen H, Ghori FY. Genetic and transcriptional control of bone formation. Oral and Maxillofacial Surgery Clinics of North America. 2010;22:283-293. DOI: 10.1016/j.coms.2010.05.001
  8. 8. Salgado CL, Grenho L, Fernandes MH, Colaco BJ, Monteiro FJ. Biodegradation, biocompatibility, and osteoconduction evaluation of collagen-nanohydroxyapatite cryogels for bone tissue regeneration. Journal of Biomedical Materials Research. Part A. 2016;104:57-70. DOI: 10.1002/jbm.a.35540
  9. 9. Albrektsson T, Johansson C. Osteoinduction, osteoconduction and osseointegration. European Spine Journal. 2001;10(Suppl 2):S96-S101. DOI: 10.1007/s005860100282
  10. 10. Percival CJ, Richtsmeier JT. Angiogenesis and intramembranous osteogenesis. Developmental Dynamics. 2013;242:909-922. DOI: 10.1002/dvdy.23992
  11. 11. Stegen S, van Gastel N, Carmeliet G. Bringing new life to damaged bone: The importance of angiogenesis in bone repair and regeneration. Bone. 2015;70:19-27. DOI: 10.1016/j.bone.2014.09.017
  12. 12. Ando Y, Matsubara K, Ishikawa J, Fujio M, Shohara R, Hibi H, et al. Stem cell-conditioned medium accelerates distraction osteogenesis through multiple regenerative mechanisms. Bone. 2014;61:82-90. DOI: 10.1016/j.bone.2013.12.029
  13. 13. Chambers TJ. The birth of the osteoclast. Annals of the New York Academy of Sciences. 2010;1192:19-26. DOI: 10.1111/j.1749-6632.2009.05224.x
  14. 14. Boyce BF. Advances in the regulation of osteoclasts and osteoclast functions. Journal of Dental Research. 2013;92:860-867. DOI: 10.1177/0022034513500306
  15. 15. Matsuo K, Irie N. Osteoclast–osteoblast communication. Archives of Biochemistry and Biophysics. 2008;473:201-209. DOI: 10.1016/j.abb.2008.03.027
  16. 16. Komori T. Regulation of osteoblast differentiation by transcription factors. Journal of Cellular Biochemistry. 2006;99:1233-1239. DOI: 10.1002/jcb.20958
  17. 17. Capulli M, Paone R, Rucci N. Osteoblast and osteocyte: Games without frontiers. Archives of Biochemistry and Biophysics. 2014;561:3-12. DOI: 10.1016/j.abb.2014.05.003
  18. 18. Dirckx N, Van Hul M, Maes C. Osteoblast recruitment to sites of bone formation in skeletal development, homeostasis, and regeneration. Birth Defects Research Part C: Embryo Today: Reviews. 2013;99:170-191. DOI: 10.1002/bdrc.21047
  19. 19. Saran U, Gemini Piperni S, Chatterjee S. Role of angiogenesis in bone repair. Archives of Biochemistry and Biophysics. 2014;561:109-117. DOI: 10.1016/j.abb.2014.07.006
  20. 20. Katsara O, Mahaira LG, Iliopoulou EG, Moustaki A, Antsaklis A, Loutradis D, et al. Effects of donor age, gender, and In vitro cellular aging on the phenotypic, functional, and molecular characteristics of mouse bone marrow-derived mesenchymal stem cells. Stem Cells and Development. 2011;20:1549-1561. DOI: 10.1089/scd.2010.0280
  21. 21. Hass R, Kasper C, Bohm S, Jacobs R. Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC. Cell Communication and Signaling: CCS. 2011;9:12. DOI: 10.1186/1478-811X-9-12
  22. 22. Bléry P, Corre P, Malard O, Sourice S, Pilet P, Amouriq Y, et al. Evaluation of new bone formation in irradiated areas using association of mesenchymal stem cells and total fresh bone marrow mixed with calcium phosphate scaffold. Journal of Materials Science: Materials in Medicine. 2014;25:2711-2720. DOI: 10.1007/s10856-014-5282-5
  23. 23. Hao Z, Wang S, Zhang X, Lu J. Stem cell therapy: A promising biological strategy for tendon-bone healing after anterior cruciate ligament reconstruction. Cell Proliferation. 2016;49:154-162. DOI: 10.1111/cpr.12242
  24. 24. Burdon TJ, Paul A, Noiseux N, Prakash S, Shum-Tim D. Bone marrow stem cell derived paracrine factors for regenerative medicine: Current perspectives and therapeutic potential. Bone Marrow Research. 2011;2011:1-14. DOI: 10.1155/2011/207326
  25. 25. Sharma RI, Snedeker JG. Paracrine interactions between mesenchymal stem cells affect substrate driven differentiation toward tendon and bone phenotypes. PLoS One. 2012;7:e31504. DOI: 10.1371/journal.pone.0031504
  26. 26. Liu M, Sun Y, Zhang Q. Emerging role of extracellular vesicles in bone remodeling. Journal of Dental Research. 2018;97(8):859-868. DOI: 10.1177/0022034518764411
  27. 27. Keshtkar S, Azarpira N, Ghahremani MH. Mesenchymal stem cell-derived extracellular vesicles: Novel frontiers in regenerative medicine. Stem Cell Research & Therapy. 2018;9(1):63. DOI: 10.1186/s13287-018-0791-7
  28. 28. Golchin A, Hosseinzadeh S, Ardeshirylajimi A. The exosomes released from different cell types and their effects in wound healing. Journal of Cellular Biochemistry. 2018;119(7):5043-5052. DOI: 10.1002/jcb.26706
  29. 29. Harding C, Heuser J, Stahl P. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. The Journal of Cell Biology. 1983;97:329-339
  30. 30. Johnstone RM, Adam M, Hammond JR, Orr L, Turbide C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). The Journal of Biological Chemistry. 1987;262:9412-9420
  31. 31. Mathivanan S, Ji H, Simpson RJ. Exosomes: Extracellular organelles important in intercellular communication. Journal of Proteomics. 2010;73:1907-1920. DOI: 10.1016/j.jprot.2010.06.006
  32. 32. De Jong OG, Van Balkom BWM, Schiffelers RM, Bouten CVC, Verhaar MC. Extracellular vesicles: Potential roles in regenerative medicine. Frontiers in Immunology. 2014;5:608. DOI: 10.3389/fimmu.2014.00608
  33. 33. Thery C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Current Protocols in Cell Biology. 2006;Chapter 3:3.22. DOI: 10.1002/0471143030.cb0322s30
  34. 34. Raposo G, Stoorvogel W. Extracellular vesicles: Exosomes, microvesicles, and friends. The Journal of Cell Biology. 2013;200:373-383. DOI: 10.1083/jcb.201211138
  35. 35. Bjørge IM, Kim SY, Mano JF, Kalionis B, Chrzanowski W. Extracellular vesicles, exosomes and shedding vesicles in regenerative medicine—A new paradigm for tissue repair. Biomaterials Science. 2018;6:60-78. DOI: 10.1039/C7BM00479F
  36. 36. Hsu C, Morohashi Y, Yoshimura S, Manrique-Hoyos N, Jung S, Lauterbach MA, et al. Regulation of exosome secretion by Rab35 and its GTPase-activating proteins TBC1D10A–C. The Journal of Cell Biology. 2010;189:223-232. DOI: 10.1083/jcb.200911018
  37. 37. van Niel G, Porto-Carreiro I, Simoes S, Raposo G. Exosomes: A common pathway for a specialized function. The Journal of Biochemistry. 2006;140:13-21. DOI: 10.1093/jb/mvj128
  38. 38. Simons M, Raposo G. Exosomes—Vesicular carriers for intercellular communication. Current Opinion in Cell Biology. 2009;21:575-581. DOI: 10.1016/j.ceb.2009.03.007
  39. 39. Zitvogel L, Regnault A, Lozier A, Wolfers J, Flament C, Tenza D, et al. Eradication of established murine tumors using a novel cell-free vaccine: Dendritic cell-derived exosomes. Nature Medicine. 1998;4:594-600
  40. 40. Van Niel G, Raposo G, Candalh C, Boussac M, Hershberg R, Cerf Bensussan N, et al. Intestinal epithelial cells secrete exosome–like vesicles. Gastroenterology. 2001;121:337-349. DOI: 10.1053/gast.2001.26263
  41. 41. Zhang H, Xie Y, Li W, Chibbar R, Xiong S, Xiang J. CD4(+) T cell-released exosomes inhibit CD8(+) cytotoxic T-lymphocyte responses and antitumor immunity. Cellular & Molecular Immunology. 2011;8:23-30. DOI: 10.1038/cmi.2010.59
  42. 42. Goldie BJ, Dun MD, Lin M, Smith ND, Verrills NM, Dayas CV, et al. Activity-associated miRNA are packaged in Map1b-enriched exosomes released from depolarized neurons. Nucleic Acids Research. 2014;42:9195-9208. DOI: 10.1093/nar/gku594
  43. 43. Xiao H, Lasser C, Shelke GV, Wang J, Radinger M, Lunavat TR, et al. Mast cell exosomes promote lung adenocarcinoma cell proliferation—role of KIT-stem cell factor signaling. Cell Communication and Signaling: CCS. 2014;12:64. DOI: 10.1186/s12964-014-0064-8
  44. 44. Pisitkun T, Shen RF, Knepper MA. Identification and proteomic profiling of exosomes in human urine. Proceedings of the National Academy of Sciences of the United States of America. 2004;101:13368-13373. DOI: 10.1073/pnas.0403453101
  45. 45. Keller S, Ridinger J, Rupp AK, Janssen JW, Altevogt P. Body fluid derived exosomes as a novel template for clinical diagnostics. Journal of Translational Medicine. 2011;9:86. DOI: 10.1186/1479-5876-9-86
  46. 46. Muller L, Hong C, Stolz DB, Watkins SC, Whiteside TL. Isolation of biologically-active exosomes from human plasma. Journal of Immunological Methods. 2014;411:55-65. DOI: 10.1016/j.jim.2014.06.007
  47. 47. Lin J, Li J, Huang B, Liu J, Chen X, Chen X, et al. Exosomes: Novel biomarkers for clinical diagnosis. The Scientific World Journal. 2015;2015:1-8. DOI: 10.1155/2015/657086
  48. 48. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biology. 2007;9:654-659. DOI: 10.1038/ncb1596
  49. 49. Morhayim J, van de Peppel J, Dudakovic A, Chiba H, van Wijnen AJ, van Leeuwen JP. Molecular characterization of human osteoblast-derived extracellular vesicle mRNA using next-generation sequencing. Biochimica et Biophysica Acta. 2017;1864:1133-1141. DOI: 10.1016/j.bbamcr.2017.03.011
  50. 50. Quan L, Wang Y, Liang J, Qiu T, Wang H, Zhang Y, et al. Screening for genes, transcription factors and miRNAs associated with the myogenic and osteogenic differentiation of human adipose tissue-derived stem cells. International Journal of Molecular Medicine. 2016;38:1839-1849. DOI: 10.3892/ijmm.2016.2788
  51. 51. Cantaluppi V, Gatti S, Medica D, Figliolini F, Bruno S, Deregibus MC, et al. Microvesicles derived from endothelial progenitor cells protect the kidney from ischemia–reperfusion injury by microRNA-dependent reprogramming of resident renal cells. Kidney International. 2012;82:412-427. DOI: 10.1038/ki.2012.105
  52. 52. Aghajani Nargesi A, Lerman LO, Eirin A. Mesenchymal stem cell-derived extracellular vesicles for kidney repair: Current status and looming challenges. Stem Cell Research & Therapy. 2017;8(1):273. DOI: 10.1186/s13287-017-0727-7
  53. 53. Ni J, Sun Y, Liu Z. The potential of stem cells and stem cell-derived exosomes in treating cardiovascular diseases. Journal of Cardiovascular Translational Research. 2018;3(1):1-11. DOI: 10.1007/s12265-018-9799-8
  54. 54. Tsilioni I, Panagiotidou S, Theoharides TC. Exosomes in neurologic and psychiatric disorders. Clinical Therapeutics. 2014;36:882-888. DOI: 10.1016/j.clinthera.2014.05.005
  55. 55. Yamaguchi T, Izumi Y, Nakamura Y, Yamazaki T, Shiota M, Sano S, et al. Repeated remote ischemic conditioning attenuates left ventricular remodeling via exosome-mediated intercellular communication on chronic heart failure after myocardial infarction. International Journal of Cardiology. 2015;178:239-246. DOI: 10.1016/j.ijcard.2014.10.144
  56. 56. Lee C, Mitsialis SA, Aslam M, Vitali SH, Vergadi E, Konstantinou G, et al. Exosomes mediate the cytoprotective action of mesenchymal stromal cells on hypoxia-induced pulmonary hypertension. Circulation. 2012;126:2601-2611. DOI: 10.1161/CIRCULATIONAHA.112.114173
  57. 57. Burger D, Viñas JL, Akbari S, Dehak H, Knoll W, Gutsol A, et al. Human endothelial colony-forming cells protect against acute kidney injury. The American Journal of Pathology. 2015;185:2309-2323. DOI: 10.1016/j.ajpath.2015.04.010
  58. 58. Hao ZC, Lu J, Wang SZ, Wu H, Zhang YT, Xu SG. Stem cell-derived exosomes: A promising strategy for fracture healing. Cell Proliferation. 2017;50(5):1-10. DOI: 10.1111/cpr.12359
  59. 59. Qin Y, Wang L, Gao Z, Chen G, Zhang C. Bone marrow stromal/stem cell-derived extracellular vesicles regulate osteoblast activity and differentiation in vitro and promote bone regeneration in vivo. Scientific Reports. 2016;50(5):1-10. DOI: 10.1038/srep21961
  60. 60. Barba M, Cicione C, Bernardini C, Michetti F, Lattanzi W. Adipose-derived mesenchymal cells for bone regereneration: State of the art. BioMed Research International. 2013;6:21961. DOI: 10.1155/2013/416391
  61. 61. Zigdon-Giladi H, Michaeli-Geller G, Bick T, Lewinson D, Machtei EE. Human blood-derived endothelial progenitor cells augment vasculogenesis and osteogenesis. Journal of Clinical Periodontology. 2015;42:89-95. DOI: 10.1111/jcpe.12325
  62. 62. Goerke SM, Obermeyer J, Plaha J, Stark GB, Finkenzeller G. Endothelial progenitor cells from peripheral blood support bone regeneration by provoking an angiogenic response. Microvascular Research. 2015;98:40-47. DOI: 10.1016/j.mvr.2014.12.001
  63. 63. Yin P, Lv H, Li Y, Deng Y, Zhang L, Tang P. Exosome-mediated genetic information transfer, a missing piece of osteoblast–osteoclast communication puzzle. Frontiers in Endocrinology. 2017;8:336. DOI: 10.3389/fendo.2017.00336
  64. 64. Inose H, Ochi H, Kimura A, Fujita K, Xu R, Sato S, et al. A microRNA regulatory mechanism of osteoblast differentiation. Proceedings of repair in a mouse model. Arthritis and Rheumatism. 2009;60:813-823. DOI: 10.1002/art.24330
  65. 65. Martins M, Ribeiro D, Martins A, Reis RL, Neves NM. Extracellular vesicles derived from osteogenically induced human bone marrow mesenchymal stem cells can modulate lineage commitment. Stem Cell Reports. 2016;6:284-291. DOI: 10.1016/j.stemcr.2016.01.001
  66. 66. Pegtel DM, Cosmopoulos K, Thorley-Lawson DA, van Eijndhoven MAJ, Hopmans ES, Lindenberg JL, et al. Functional delivery of viral miRNAs via exosomes. Proceedings of the National Academy of Sciences. 2010;107:6328-6333. DOI: 10.1073/pnas.0914843107
  67. 67. Xu JF, Yang GH, Pan XH, Zhang SJ, Zhao C, Qiu BS, et al. Altered microRNA expression profile in exosomes during osteogenic differentiation of human bone marrow-derived mesenchymal stem cells. PLoS One. 2014;9:e114627. DOI: 10.1371/journal.pone.0114627
  68. 68. Wei J, Li H, Wang S, Li T, Fan J, Liang X, et al. Let-7 enhances osteogenesis and bone formation while repressing adipogenesis of human stromal/mesenchymal stem cells by regulating HMGA2. Stem Cells and Development. 2014;23:1452-1463. DOI: 10.1089/scd.2013.0600
  69. 69. Furuta T, Miyaki S, Ishitobi H, Ogura T, Kato Y, Kamei N, et al. Mesenchymal stem cell-derived exosomes promote fracture healing in a mouse model. Stem Cells Translational Medicine. 2016;5:1620-1630. DOI: 10.5966/sctm.2015-0285
  70. 70. Kitaori T, Ito H, Schwarz EM, Tsutsumi R, Yoshitomi H, Oishi S, et al. Stromal cell-derived factor 1/CXCR4 signaling is critical for the recruitment of mesenchymal stem cells to the fracture site during skeletal repair in a mouse model. Arthritis and Rheumatism. 2009;60:813-823. DOI: 10.1002/art.24330
  71. 71. Ishikawa M, Ito H, Kitaori T, Murata K, Shibuya H, Furu M, et al. MCP/CCR2 signaling is essential for recruitment of mesenchymal progenitor cells during the early phase of fracture healing. PLoS One. 2014;9:e104954. DOI: 10.1371/journal.pone.0104954
  72. 72. Wang X, Omar O, Vazirisani F, Thomsen P, Ekström K. Mesenchymal stem cell-derived exosomes have altered microRNA profiles and induce osteogenic differentiation depending on the stage of differentiation. PLoS One. 2018;13:e193059. DOI: 10.1371/journal.pone.0193059
  73. 73. Bakhshandeh B, Hafizi M, Ghaemi N, Soleimani M. Down-regulation of miRNA-221 triggers osteogenic differentiation in human stem cells. Biotechnology Letters. 2012;34:1579-1587. DOI: 10.1007/s10529-012-0934-3
  74. 74. Baglìo SR, Devescovi V, Granchi D, Baldini N. MicroRNA expression profiling of human bone marrow mesenchymal stem cells during osteogenic differentiation reveals Osterix regulation by miR-31. Gene. 2013;527:321-331. DOI: 10.1016/j.gene.2013.06.021
  75. 75. Sun Y, Xu L, Huang S, Hou Y, Liu Y, Chan K, et al. Mir-21 overexpressing mesenchymal stem cells accelerate fracture healing in a rat closed femur fracture model. BioMed Research International. 2015;2015:1-9. DOI: 10.1155/2015/412327
  76. 76. Huang C, Geng J, Wei X, Zhang R, Jiang S. MiR-144-3p regulates osteogenic differentiation and proliferation of murine mesenchymal stem cells by specifically targetingSmad4. FEBS Letters. 2016;590:795-807. DOI: 10.1002/1873-3468.12112
  77. 77. Cong R, Tao K, Fu P, Lou L, Zhu Y, Chen S, et al. MicroRNA218 promotes prostaglandin E2 to inhibit osteogenic differentiation in synovial mesenchymal stem cells by targeting 15hydroxyprostaglandin dehydrogenase [NAD(+)]. Molecular Medicine Reports. 2017;16:9347-9354. DOI: 10.3892/mmr.2017.7795
  78. 78. Chen C, Cheng P, Xie H, Zhou H, Wu X, Liao E, et al. MiR-503 regulates osteoclastogenesis via targeting RANK. Journal of Bone and Mineral Research. 2014;29:338-347. DOI: 10.1002/jbmr.2032
  79. 79. Xie Y, Chen Y, Zhang L, Ge W, Tang P. The roles of bone-derived exosomes and exosomal microRNAs in regulating bone remodelling. Journal of Cellular and Molecular Medicine. 2017;21:1033-1041. DOI: 10.1111/jcmm.13039
  80. 80. Xu Q, Cui Y, Luan J, Zhou X, Li H, Han J. Exosomes from C2C12 myoblasts enhance osteogenic differentiation of MC3T3-E1 pre-osteoblasts by delivering miR-27a-3p. Biochemical and Biophysical Research Communications. 2018;498:32-37. DOI: 10.1016/j.bbrc.2018.02.144
  81. 81. Li D, Liu J, Guo B, Liang C, Dang L, Lu C, et al. Osteoclast-derived exosomal miR-214-3p inhibits osteoblastic bone formation. Nature Communications. 2016;7:10872. DOI: 10.1038/ncomms10872
  82. 82. Li W, Liu Y, Zhang P, Tang Y, Zhou M, Jiang W, et al. Tissue-engineered bone immobilized with human adipose stem cells-derived exosomes promotes bone regeneration. ACS Applied Materials & Interfaces. 2018;10:5240-5254. DOI: 10.1021/acsami.7b17620
  83. 83. Wang K, Xu L, Rui Y, Huang S, Lin S, Xiong J, et al. The effects of secretion factors from umbilical cord derived mesenchymal stem cells on osteogenic differentiation of mesenchymal stem cells. PLoS One. 2015;10:e120593. DOI: 10.1371/journal.pone.0120593
  84. 84. Qi X, Zhang J, Yuan H, Xu Z, Li Q, Niu X, et al. 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. International Journal of Biological Sciences. 2016;12:836-849. DOI: 10.7150/ijbs.14809
  85. 85. Zhang Y, Xie RL, Croce CM, Stein JL, Lian JB, van Wijnen AJ, et al. A program of microRNAs controls osteogenic lineage progression by targeting transcription factor Runx2. Proceedings of the National Academy of Sciences. 2011;108:9863-9868. DOI: 10.1073/pnas.1018493108
  86. 86. Zhang Y, Xie R, Gordon J, LeBlanc K, Stein JL, Lian JB, et al. Control of mesenchymal lineage progression by MicroRNAs targeting skeletal gene regulators Trps1 and Runx2. Journal of Biological Chemistry. 2012;287:21926-21935. DOI: 10.1074/jbc.M112.340398
  87. 87. Deng Y, Zhou H, Gu P, Fan X. Repair of canine medial orbital bone defects with miR-31-modified bone marrow mesenchymal stem cells. Investigative Ophthalmology & Visual Science. 2014;55:6016-6023. DOI: 10.1167/iovs.14-14977
  88. 88. Ghosh-Choudhury N. Requirement of BMP-2-induced phosphatidylinositol 3-kinase and Akt serine/threonine kinase in osteoblast differentiation and Smad-dependent BMP-2 gene transcription. Journal of Biological Chemistry. 2002;277:33361-33368. DOI: 10.1074/jbc.M205053200
  89. 89. Suzuki E, Ochiai-Shino H, Aoki H, Onodera S, Saito A, Saito A, et al. Akt activation is required for TGF-β1-induced osteoblast differentiation of MC3T3-E1 pre-osteoblasts. PLoS One. 2014;9:e112566. DOI: 10.1371/journal.pone.0112566
  90. 90. Shabbir A, Cox A, Rodriguez-Menocal L, Salgado M, Badiavas EV. Mesenchymal stem cell exosomes induce proliferation and migration of normal and chronic wound fibroblasts, and enhance angiogenesis in vitro. Stem Cells and Development. 2015;24:1635-1647. DOI: 10.1089/scd.2014.0316
  91. 91. Zhang J, Liu X, Li H, Chen C, Hu B, Niu X, et al. Exosomes/tricalcium phosphate combination scaffolds can enhance bone regeneration by activating the PI3K/Akt signaling pathway. Stem Cell Research & Therapy. 2016;7. DOI: 10.1186/s13287-016-0391-3
  92. 92. Trohatou O, Zagoura D, Bitsika V, Pappa KI, Antsaklis A, Anagnou NP, et al. Sox2 suppression by miR-21 governs human mesenchymal stem cell properties. Stem Cells Translational Medicine. 2014;3:54-68. DOI: 10.5966/sctm.2013-0081
  93. 93. Meng YB, Li X, Li ZY, Zhao J, Yuan XB, Ren Y, et al. microRNA-21 promotes osteogenic differentiation of mesenchymal stem cells by the PI3K/beta-catenin pathway. Journal of Orthopaedic Research. 2015;33:957-964. DOI: 10.1002/jor.22884
  94. 94. Liu X, Li Q, Niu X, Hu B, Chen S, Song W, et al. Exosomes secreted from human-induced pluripotent stem cell-derived mesenchymal stem cells prevent osteonecrosis of the femoral head by promoting angiogenesis. International Journal of Biological Sciences. 2017;13:232-244. DOI: 10.7150/ijbs.16951
  95. 95. Leucht P, Lee S, Yim N. Wnt signaling and bone regeneration: Can't have one without the other. Biomaterials. 2018;3(15):S0142-9612(18)30196-0. DOI: 10.1016/j.biomaterials.2018.03.029
  96. 96. 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 Engineering Part A. 2017;23:1212-1220. DOI: 10.1089/ten.tea.2016.0548
  97. 97. Zhang B, Wu X, Zhang X, Sun Y, Yan Y, Shi H, et al. Human umbilical cord mesenchymal stem cell exosomes enhance angiogenesis through the Wnt4/beta-catenin pathway. Stem Cells Translational Medicine. 2015;4:513-522. DOI: 10.5966/sctm.2014-0267
  98. 98. McBride JD, Rodriguez-Menocal L, Guzman W, Candanedo A, Garcia-Contreras M, Badiavas EV. Bone marrow mesenchymal stem cell-derived CD63(+) exosomes transport Wnt3a exteriorly and enhance dermal fibroblast proliferation, migration, and angiogenesis In vitro. Stem Cells and Development. 2017;26:1384-1398. DOI: 10.1089/scd.2017.0087
  99. 99. Huynh N, VonMoss L, Smith D, Rahman I, Felemban MF, Zuo J, et al. Characterization of regulatory extracellular vesicles from osteoclasts. Journal of Dental Research. 2016;95:673-679. DOI: 10.1177/0022034516633189
  100. 100. Deng L, Wang Y, Peng Y, Wu Y, Ding Y, Jiang Y, et al. Osteoblast-derived microvesicles: A novel mechanism for communication between osteoblasts and osteoclasts. Bone. 2015;79:37-42. DOI: 10.1016/j.bone.2015.05.022
  101. 101. Chen X, Zhi X, Yin Z, Li X, Qin L, Qiu Z, et al. 18β-Glycyrrhetinic acid inhibits osteoclastogenesis in vivo and in vitro by blocking RANKL-mediated RANK–TRAF6 interactions and NF-κB and MAPK signaling pathways. Frontiers in Pharmacology. 2018;9:647. DOI: 10.3389/fphar.2018.00647
  102. 102. Narayanan R, Huang C, Ravindran S. Hijacking the cellular mail: Exosome mediated differentiation of mesenchymal stem cells. Stem Cells International. 2016;2016:1-11. DOI: 10.1155/2016/3808674
  103. 103. Zhao L, Jiang S, Hantash BM. Transforming growth factor beta1 induces osteogenic differentiation of murine bone marrow stromal cells. Tissue Engineering. Part A. 2010;16:725-733. DOI: 10.1089/ten.TEA.2009.0495
  104. 104. Lamplot JD, Qin J, Nan G, Wang J, Liu X, Yin L, et al. BMP9 signaling in stem cell differentiation and osteogenesis. American Journal of Stem Cells. 2013;2:1-21
  105. 105. Minardi S, Pandolfi L, Taraballi F, Wang X, De Rosa E, Mills ZD, et al. Enhancing vascularization through the controlled release of platelet-derived growth factor-BB. ACS Applied Materials & Interfaces. 2017;9:14566-14575. DOI: 10.1021/acsami.6b13760
  106. 106. Al Rifai O, Chow J, Lacombe J, Julien C, Faubert D, Susan-Resiga D, et al. Proprotein convertase furin regulates osteocalcin and bone endocrine function. Journal of Clinical Investigation. 2017;127:4104-4117. DOI: 10.1172/JCI93437
  107. 107. Feng JQ, Ward LM, Liu S, Lu Y, Xie Y, Yuan B, et al. Loss of DMP1 causes rickets and osteomalacia and identifies a role for osteocytes in mineral metabolism. Nature Genetics. 2006;38:1310-1315. DOI: 10.1038/ng1905
  108. 108. Ramachandran A, Ravindran S, Huang C, George A. TGF beta receptor II interacting protein-1, an intracellular protein has an extracellular role as a modulator of matrix mineralization. Scientific Reports. 2016;6. DOI: 10.1038/srep37885
  109. 109. Pethő A, Chen Y, George A. Exosomes in extracellular matrix bone biology. Current Osteoporosis Reports. 2018;16:58-64. DOI: 10.1007/s11914-018-0419-y
  110. 110. Li C, Sunderic K, Nicoll SB, Wang S. Downregulation of heat shock protein 70 impairs osteogenic and chondrogenic differentiation in human mesenchymal stem cells. Scientific Reports. 2018;8(1):553. DOI: 10.1038/s41598-017-18541-1
  111. 111. Flanagan M, Li C, Dietrich MA, Richard M, Yao S. Downregulation of heat shock protein B8 decreases osteogenic differentiation potential of dental pulp stem cells during in vitro proliferation. Cell Proliferation. 2018;51:e12420. DOI: 10.1111/cpr.12420
  112. 112. Lankford KL, Arroyo EJ, Nazimek K, Bryniarski K, Askenase PW, Kocsis JD. Intravenously delivered mesenchymal stem cell-derived exosomes target M2-type macrophages in the injured spinal cord. PLoS One. 2018;13:e190358. DOI: 10.1371/journal.pone.0190358
  113. 113. Zhang B, Yeo RWY, Lai RC, Sim EWK, Chin KC, Lim SK. Mesenchymal stromal cell exosome–enhanced regulatory T-cell production through an antigen-presenting cell–mediated pathway. Cytotherapy. 2018;20(5):687-696. DOI: 10.1016/j.jcyt.2018.02.372
  114. 114. Nassar W, El-Ansary M, Sabry D, Mostafa MA, Fayad T, Kotb E, et al. Umbilical cord mesenchymal stem cells derived extracellular vesicles can safely ameliorate the progression of chronic kidney diseases. Biomaterials Research. 2016;20:21. DOI: 10.1186/s40824-016-0068-0
  115. 115. Gardiner C, Vizio DD, Sahoo S, Théry C, Witwer KW, Wauben M, et al. Techniques used for the isolation and characterization of extracellular vesicles: Results of a worldwide survey. Journal of Extracellular Vesicles. 2016;5:32945. DOI: 10.3402/jev.v5.32945
  116. 116. Ahrlund-Richter L, De Luca M, Marshak DR, Munsie M, Veiga A, Rao M. Isolation and production of cells suitable for human therapy: Challenges ahead. Cell Stem Cell. 2009;4:20-26. DOI: 10.1016/j.stem.2008.11.012
  117. 117. Chen AK, Chen X, Choo ABH, Reuveny S, Oh SKW. Critical microcarrier properties affecting the expansion of undifferentiated human embryonic stem cells. Stem Cell Research. 2011;7:97-111. DOI: 10.1016/j.scr.2011.04.007
  118. 118. Li P, Kaslan M, Lee SH, Yao J, Gao Z. Progress in exosome isolation techniques. Theranostics. 2017;7:789-804. DOI: 10.7150/thno.18133
  119. 119. Beaudeux JL, Peynet J, Flourie F, Keddad K, Delattre J, Rousselet F, et al. Discrepancies between lipoprotein(a) concentrations in icteric sera measured by immunonephelometry and electroimmunodiffusion. Clinical Biochemistry. 1994;27:7-11
  120. 120. Wen Y, Chang Y, Lin L, Liao P. Collection of in vivo-like liver cell secretome with alternative sample enrichment method using a hollow fiber bioreactor culture system combined with tangential flow filtration for secretomics analysis. Analytica Chimica Acta. 2011;684:81-88. DOI: 10.1016/j.aca.2010.10.040
  121. 121. Suárez H, Gámez-Valero A, Reyes R, López-Martín S, Rodríguez MJ, Carrascosa JL, et al. A bead-assisted flow cytometry method for the semi-quantitative analysis of extracellular vesicles. Scientific Reports. 2017;7(1):11271. DOI: 10.1038/s41598-017-11249-2
  122. 122. Du T, Ju G, Wu S, Cheng Z, Cheng J, Zou X, et al. Microvesicles derived from human Wharton's jelly mesenchymal stem cells promote human renal cancer cell growth and aggressiveness through induction of hepatocyte growth factor. PLoS One. 2014;9:e96836. DOI: 10.1371/journal.pone.0096836
  123. 123. Peak TC, Praharaj PP, Panigrahi GK, Doyle M, Su Y, Schlaepfer IR, et al. Exosomes secreted by placental stem cells selectively inhibit growth of aggressive prostate cancer cells. Biochemical and Biophysical Research Communications. 2018;499(4):1004-1010. DOI: 10.1016/j.bbrc.2018.04.038
  124. 124. Qi J, Zhou Y, Jiao Z, Wang X, Zhao Y, Li Y, et al. Exosomes derived from human bone marrow mesenchymal stem cells promote tumor growth through hedgehog signaling pathway. Cellular Physiology and Biochemistry. 2017;42:2242-2254. DOI: 10.1159/000479998
  125. 125. Wang J, Hendrix A, Hernot S, Lemaire M, De Bruyne E, Van Valckenborgh E, et al. Bone marrow stromal cell-derived exosomes as communicators in drug resistance in multiple myeloma cells. Blood. 2014;124:555-566. DOI: 10.1182/blood-2014-03-562439
  126. 126. Roccaro AM, Sacco A, Maiso P, Azab AK, Tai Y, Reagan M, et al. BM mesenchymal stromal cell–derived exosomes facilitate multiple myeloma progression. Journal of Clinical Investigation. 2013;123:1542-1555. DOI: 10.1172/JCI66517
  127. 127. Umezu T, Imanishi S, Azuma K, Kobayashi C, Yoshizawa S, Ohyashiki K, et al. Replenishing exosomes from older bone marrow stromal cells with miR-340 inhibits myeloma-related angiogenesis. Blood Advances. 2017;1:812-823. DOI: 10.1182/bloodadvances.2016003251

Written By

Jiazhao Yang, Wanbo Zhu, Jinsen Lu, Kai Xie, Shiyuan Fang and Lixin Kan

Submitted: 08 June 2018 Reviewed: 22 August 2018 Published: 05 November 2018