Abstract
Mesenchymal stem cells (MSCs) are pluripotent stem cells with self-renewal. They play a critical role in cell therapy due to their powerful immunomodulatory and regenerative effects. Recent studies suggest that one of the key therapeutic mechanisms of MSCs seems to derive from their paracrine product, called extracellular vesicles (EVs). The EVs contain much DNA, messenger RNA (mRNA), microRNA, and protein components, which can exert intracellular communication to target cells. In clinical applications, the MSC-EVs have been widely used in tissue repair and immune disorder diseases. However, there are serval issues need to be considered such as how to accomplish the large-scale production of EVs and how to verify the exact mechanism of EVs. In this review, we summarize the current progress of MSC-EVs and discuss the challenges and future of MSC-EVs.
Keywords: mesenchymal stem cell, extracellular vesicles, tissue repair, immunomodulatory
Graphical Abstract.
Introduction
Stem cell therapy has hailed as one of the most promising therapy of the 21st century 1 . Among the types of stem cells, mesenchymal stem cells (MSCs) have been widely used in clinical applications due to great regenerative potential and immunomodulatory functions2,3. Also, MSCs can be isolated from umbilical cord, amniotic fluid, bone marrow, fat, and other tissues 4 . In recent years, there are more than 10 kinds of MSC drug marketing around the world. Initially, the therapeutic mechanism of MSCs was attributed to their homing into the lesion or multiple differentiative ability, but the number of cells is extremely small and quickly being cleaned by immune cells after transplantation5,6. Therefore, more and more studies suggested that MSCs play significant role in the process of therapeutic effects through their paracrine function 7 . Studies have shown that the extracellular vesicles (EVs), one of the paracrine products of MSCs, have become key role of cell-free therapy (CFT), which can escape the immune response, not tumorigenic and safer to use compared with MSCs therapy8,9.
In 1983, EVs were discovered in rat reticulocytes by Pan and Johnstone but being reproved cellular waste 10 . In 2007, researchers found that EVs contain proteins, lipids, and RNA, such as mRNAs and microRNAs9,11,12 (Fig. 1). At present, a number of in vitro and in vivo experiments have claimed that the nano-sized vesicle plays a crucial role in cell-to-cell communication, thereby promoting proliferation, simulating vascularization and regulating immune responses 13 . It is worth noting that this CFT based on EVs has functions similar to MSCs and other stem cell types. Therefore, a series of clinical practice was carried out to verity the efficacy, and the indication was extensive, including immune disorders, cardiovascular disease, COVID-19, and wound healing14–20. The purpose of this review is to sum up the current development of MSC-EVs in biomedical and discuss the current challenges and future directions.
Figure 1.

The structure of EVs. EVs consist of lipid bilayer membrane, rich in DNAs, mRNAs, microRNAs, and proteins, such as tetraspanins (CD9, CD63, CD81, etc).
Biology of MSC-EVs
EVs have a cup-shaped morphology under an electron microscope and have a diameter ranging from 30 to 120 nm 21 . These vesicles can be concentrated in the 1.10–1.21 g/ml section of a sucrose density gradient22–24. Multi-omics have shown that the contents of EVs are rich, including various microRNAs, mRNAs, and proteins. Compared with stem cells, the stem cell–derived EVs have more stable biological characteristics due to special lipid bilayer membrane structure, which can prevent the degradation of its contents and maintain the function of internal proteins and genetic materials25–27. Also, EVs can be stored long term at −80°C but stem cells should be stored at −196°C liquid nitrogen28,29. Different types of MSCs can secrete EVs under normal and pathological conditions. In addition, there are some differences between the types of MSCs. Studies have shown that EVs derived from human umbilical cord MSCs (hUC-MSCs) have higher activity than EVs derived from human bone morrow MSCs (hBM-MSCs) 30 . Compared with hBM-MSCs, the yield of EVs secreted by human amniotic fluid MSCs (hAF-MSCs) is higher30,31. However, the EVs of hUC-MSCs and hBM-MSC, sharing 60% of protein components related to cell growth and antioxidant stress, have been confirmed that can inhibit the growth and apoptosis in tumor cells32,33. Significantly, no substantial differences were found in EVs among hUC-MSCs and hBM-MSC, and generally, these EVs have similar function9,34–37.
EVs are largely originated from multivesicular bodies (MVBs). When MVB is fused with MSC membrane, the inner vesicles are secreted as EVs38,39. The formation process of EVs is as follows: first, intraluminal vesicles (ILVs), the precursors of MVBs and EVs, are formed under cellular maturation process; second, MVBs merge with the lysosomal membrane and then the inner vesicles are released into extracellular matrix as EVs 40 . As a paracrine product of MSCs, EVs have two ways to enter the target cell: one is through endocytosis by target cell, and the other is through membrane fusion between EVs and target cell 38 . And then, the various biological substances contained into target cell are released to finish signal transduction, for example, the mRNA contents can translate the corresponding protein when it binds to target cell, the microRNA contents can regulate the expression of target protein by inhibiting the process of mRNA translation or degrading mRNA directly41,42.
Clinical Significances of MSC-EVs
Currently, there are more than 150 clinical trials involving EVs as listed in www.clinicaltrails.gov. These trials not only focus on using the EVs as a diagnostic biomarker to predict the outcome of treatments, but also concentrate on using the EVs as CFT in various clinical applications, especially for tissue repair and immune disorders (Table 1).
Table 1.
Clinical Trials of MSC-EVs.
| NCT number | Indication | Intervention | Phase | Sponsor |
|---|---|---|---|---|
| NCT05475418 | Wound healing | Topical | Phase I | Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University, China |
| NCT05060107 | Knee osteoarthritis | Intra-articular injection | Phase I | Francisco Espinoza, Universidad de los Andes, Chile |
| NCT03384433 | Acute ischemic stroke | Intravenous infusion | Phase I | Isfahan University of Medical Sciences, Iran |
| NCT04388982 | Alzheimer disease | Nasal drip | Phase I | Ruijin Hospital, China |
| NCT05523011 | Psoriasis | Topical | Phase I | Paracrine Therapeutics Dermatology Pte. Ltd. |
| NCT02138331 | Diabetes mellitus Type 1 |
Intravenous infusion | Phase II/III | General Committee of Teaching Hospitals and Institutes, Egypt |
| NCT04491240 | COVID-19 | Inhalation | Phase II | State-Financed Health Facility, Russia |
| NCT05808400 | COVID-19 | Inhalation | Phase I | Huazhong University of Science and Technology, China |
MSC-EVs for Tissue Repair
Tissue repair is a very complex process that refers to the interaction and association of different repair cells, cytokines, intercellular matrix, and proteins 43 . Ideally, the tissue damage is repaired by cells similar to injured cells to rebuild the original tissue structure and function. However, human body has limited inherent proliferation ability of tissues and cells 44 . EVs, as novel biomaterials, can be absorbed by target cells and are able to migrate long distances to lesions through the circulatory system to join in various physiological and pathological processes of tissue repair 45 . When they bind to target cells, their cargo would be released. Also, intracellular signaling pathways of tissue repair, including phosphoinositide 3-kinase (PI3K)/AKT, ERK, and STAT-3 pathway, would be triggered to facilitate and accelerate this process through downstream targets such as stromal cell–derived factor, nerve growth factor (NGF), and hepatocyte growth factor (HGF)46–48.
MSCs hold a leading post as source of EVs. Many researchers have demonstrated that MSC-EVs accelerate wound healing, relieve limb ischemia, alleviate kidney damage, stimulate cartilage tissue and bone tissue regeneration, and enhance liver and neuronal regeneration49,50 (Fig. 2). Research on the therapeutic efficacy of adipose tissue EVs are currently in clinical trials (NCT05475418), where researchers have applied adipose tissue EVs mixed with sterile hydrogel to subjects’ wounds and found to promote wound healing. A clinical study (NCT05475418) conducted by Kai Liu and team is in progress, which uses the adipose tissue MSCs (AD-MSCs)–derived EVs to treat wounds and injures. Another clinical phase I study (NCT05060107) focuses on knee osteoarthritis using the allogeneic MSC-EVs (3–5 × 1011 particles) through intra-articular injection (www.clinicaltrials.com). Besides, Nassar et al. have claimed that hUC-MSCs–derived EVs can ameliorate the progression of chronic kidney disease (CKD) from a phase II/III clinical trial. In this study, two doses (1 week apart) of EVs (100 μg/kg/dose) were used to treat 20 CKD patients. And patients intervened with EVs exhibited improved eGFRs and urinary albumin creatinine ratio 14 .
Figure 2.
MSC-EVs accelerating great tissue repair functions for multiple organs and tissues by enhancing cell migration, stimulating cell proliferation, decreasing inflammation, and increasing regeneration.
MSC-EVs for Immune Disorders
More and more evidence have shown that MSC-EVs exert amazing immunomodulatory function similar to their parental MSCs 51 . On one hand, when the immune system is underactive, MSC-EVs can promote the maturation of immune cell and rebuilding of the immune system. On the other hand, when the immune system is over-activated, MSC-EVs can regulate the transformation of immune cells into anti-inflammatory phenotypes, upregulate the expression of anti-inflammatory factors (IL-10, TGF-β), and downregulate the pro-inflammatory factors (IFN-γ, IL-17) to avoid overactive immune diseases52–54 (Fig. 3). The immunomodulatory effects of MSC-EVs are mainly due to the presentation of antigen peptides, the transfer of miRNA, and diverse signaling pathways induced by surface ligands and inner contents 55 . For example, MSC-EVs carry PD-L1 that can bind to PD-1 on the surface of T cells to inhibit the proliferation of T cells; miRNA (miR-let-7b) of MSC-EVs could target TLR4 and p-p65 gene expression and suppress STAT3 and AKT phosphorylation, which regulate the macrophages plasticity 56 ; MSC-EVs carrying latency-associated peptide could induce downstream TGF-β/Smad2/3 signaling pathways in natural killer (NK) cells 57 .
Figure 3.
MSC-EVs accelerating great immunomodulatory functions for multiple immune cells by transforming working patterns, increasing the anti-inflammatory factors, and decreasing the pro-inflammatory factor.
A clinical phase I study (NCT05523011), aiming at evaluating the safety and tolerability of MSC-EVs for psoriasis, has been completed the recruitment. The subjects will apply MSC-EVs ointment (100 μg MSC-EVs/g ointment) along with Vesiderm liposome cream from day 1 to day 20. Another chronic inflammatory disease, asthma, intervened by hBM-MSC–derived EVs, has been implemented by Sun Yai-Sen Memorial Hospital of China to verify whether the EVs would upregulate the expression of PD-L1 of macrophage, promote the function of Treg, and improve airway inflammation. Besides, the phase II/III study of type 1 diabetes (NCT02138331) has been conducted by Nassar team who used the hUC-MSC–derived EVs to reduce the inflammatory state and improve the β-cell mass because of restoring the balance between T helper 1 (Th1) and T helper 2 (Th2) immunological effects in early in vivo experiments. But the clinical results have not yet been published. Especially, more than 10 clinical studies for COVID-19 have been demonstrated that MSC-EVs could reduce the level of inflammation and educate the immune system to prevent the cytokines storm.
Challenges and Opportunities
Despite the success in preclinical and clinical studies, there are several critical issues needed to be resolved such as (1) large-scale production and isolation methods, (2) quality control, (3) pharmacology and pharmacodynamics 58 . Traditional methods to acquire EVs mainly rely on a two-dimensional (2D) culture in flask by long-term MSC passaging, which is labor-intensive as well as difficult to ensure batch-to-batch reproducibility 59 . In recent years, cell bioreactors made up of hollow fibers or based on microcarrier three-dimensional (3D) culture do partly improve the yield of EVs by increasing the surface area, but the existing process is not stable and many factors, for example, cellular confluence, cell age, oxygen concentration, and media, can affect the quality and quantity of MSC-EVs60–63. Also, there are many new methods and technologies such as tangential flow filtration (TFF), molecular exclusion method to isolate large-scale EVs, and using a combination of TFF and size exclusion chromatography. Visan et al. 64 have demonstrated that TFF is a reliable and robust EVs isolation approach that surpasses ultracentrifugation in yield, reproducibility, time, costs, and scalability. Hua et al. 65 developed a double TFF-based microfluidic device for exosome isolation from cell supernatants and human serum. Although these technologies can achieve a certain volume increase, it is still difficult to meet the purification and extraction of tens or even hundreds of liters in industrial production, and more attempts and research are needed in the future. And good manufacturing practice (GMP) for isolation, as well as quality control, needs to be established urgently. Generally, ultracentrifugation has been recognized as the standard method to separate EVs 66 (Table 2). Although studies have shown that MSC-EVs share functions similar to MSCs, the therapeutic mechanism of MSC-EVs is still unclear. Also, MSC-EVs are rapidly eliminated from blood after systemic injection. It is reported that the plasma half-life of MSC-EVs is only 2–4 min 67 .
Table 2.
Typical Isolation Methods of EVs.
| Method | Principle | Advantage | Disadvantage |
|---|---|---|---|
| Ultracentrifugation67,68 | Density, size, and shape | High quality, low cost | Time-consuming, expensive equipment |
| Immunoaffinity69,70 | Antigen capture | High purity | Time-consuming, high cost |
| Chromatography 71 | Molecular size | High purity, fast | Low quality, low yield |
| Microfluidics 72 | Physical and biochemical properties | Low cost, automated | No standardization, no clinical testing |
With the rapid development of technology, more and more large-scale bioreactors will be presented in future, which satisfy the GMP production and isolation processes. And more and more full-grown methods would be built up. Nowadays, there are many experiments using bio-imaging technology to trace the biodistribution and targeting of MSC-EVs to target tissues or cells, for instance, a study that used PKH26 or 5-(and-6)-carboxyfluorescein diacetate succinimidyl ester (CFSE) to label MSC-EVs has found that these nano-sized particles migrated to the infarcted brain in a rat model of stroke73,74. Smart biomaterial scaffolds are becoming a hot research area in tissue engineering due to great efficient and continuous release of MSC-EVs 75 . Several scaffold materials such as hydrogel, collagen, and fibroin have been used to load MSC-EVs aiming at sustaining release of EVs as well as extending the plasma half-life of EVs76–78. A dual-cargo smart bilayer hydrogel, loaded with the anti-inflammatory medication sodium diclofenac (DC) and EVs derived from MSCs of the bone marrow, has been engineered to alleviate early-stage inflammation and to stimulate late-stage stem cell attraction and cartilage-forming differentiation. This innovative hydrogel has been validated through both laboratory (in vitro) and living organism (in vivo) tests, demonstrating its effectiveness in reducing inflammation following injury and in boosting cartilage repair by efficiently neutralizing reactive oxygen species (ROS) and reprogramming macrophages to a pro-healing state 79 . A versatile collagen-based wound dressing (CTM) was developed by incorporating highly potent EVs-mimetic nanovesicles (l-Meseomes) into a porous collagen matrix crosslinked with transglutaminase (TGase). The CTM features a 3D, porous architecture that is biocompatible, has favorable swelling characteristics, and is biodegradable, allowing for a sustained release of l-Meseomes 80 .
Conclusion
EVs, as paracrine product of MSCs, contain abundant mRNA, microRNA, and protein components, which act as active signal molecular to communicate with the target cell. They have powerful tissue repair and immunomodulatory functions same as parental MSC. Compared with MSC therapy, MSC-EVs have advantages that can avoid long-term abnormal differentiation and tumor formation after cell infusion. So, MSC-EVs can also be called CFT. In past decades, MSC-EVs have shown great clinical efficacy, especially in regenerative medicine, such as wound healing and knee osteoarthritis, and immune disorder diseases, such as psoriasis and COVID-19. However, for MSC-EVs, the exact mechanism of how to stimulate proliferation, promote angiogenesis, and inhibit inflammatory infiltration is not fully understood. Therefore, understanding the biological process will help researchers better design and explore the pharmacology and pharmacodynamics of MSC-EVs. In future, there is no doubt that large-scale production equipment for EVs will be invented to increase the yield and fulfilment of the GMP process. Also, more and more biomaterials or 3D bioprinting materials will be discovered to improve the half-life and efficacy of EVs significantly.
In summary, MSC-EVs technology is still in the initial stage of clinical application. With the discoveries and new technologies coming forth, they have immense potential and prospect.
Footnotes
Author Contributions: Conceptualization—C-MT; writing—MG, BZ, XW; original draft preparation—BZ and XW; funding acquisition—C-MT. All authors have read and agreed to the published version of the manuscript.
Ethical Approval: This study was approved by our institutional review board.
Statement of Human and Animal Rights: This article does not contain any studies with human or animal subjects.
Statement of Informed Consent: There are no human subjects in this article and informed consent is not applicable.
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by the National Key R&D Program of China (2020YFA0112900), the Science and Technology Grant of Xiamen and Xiamen Emerging NPC project (no. X2106101), Natural Science Foundation of Fujian Province (2020J05302 and 2021J011358), Natural Science Basic Research Program of Shaanxi Province (2021JQ-780), National Natural Science Foundation of China (81802332), and Science Foundation of the Fujian provincial Commission of Health and Family Planning, China (2021GGB026), respectively.
ORCID iD: Chi-Meng Tzeng
https://orcid.org/0000-0003-3490-2574
References
- 1. Gage FH, Verma IM. Stem cells at the dawn of the 21st century. Proc Natl Acad Sci. 2003;100(suppl 1):11817–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Kolios G, Moodley Y. Introduction to stem cells and regenerative medicine. Respiration. 2013;85(1):3–10. [DOI] [PubMed] [Google Scholar]
- 3. Song N, Scholtemeijer M, Shah K. Mesenchymal stem cell immunomodulation: mechanisms and therapeutic potential. Trends Pharmacol Sci. 2020;41(9):653–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Mushahary D, Spittler A, Kasper C, Weber V, Charwat V. Isolation, cultivation, and characterization of human mesenchymal stem cells. Cytometry A. 2018;93(1):19–31. [DOI] [PubMed] [Google Scholar]
- 5. van Koppen A, Joles JA, van Balkom BW, Lim SK, de Kleijn D, Giles RH, Verhaar MC. Human embryonic mesenchymal stem cell-derived conditioned medium rescues kidney function in rats with established chronic kidney disease. PLoS ONE. 2012;7(6):e38746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Kourembanas S. Exosomes: vehicles of intercellular signaling, biomarkers, and vectors of cell therapy. Annu Rev Physiol. 2015;77:13–27. [DOI] [PubMed] [Google Scholar]
- 7. Spees JL, Lee RH, Gregory CA. Mechanisms of mesenchymal stem/stromal cell function. Stem Cell Res Ther. 2016;7(1):125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Phinney DG, Pittenger MF. Concise review: MSC-derived exosomes for cell-free therapy. Stem Cells. 2017;35(4):851–58. [DOI] [PubMed] [Google Scholar]
- 9. Tang Y, Zhou Y, Li HJ. Advances in mesenchymal stem cell exosomes: a review. Stem Cell Res Ther. 2021;12(1):71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Harding C, Heuser J, Stahl P. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J Cell Biol. 1983;97(2):329–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Lee BR, Kim JH, Choi ES, Cho JH, Kim E. Effect of young exosomes injected in aged mice. Int J Nanomedicine. 2018;13:5335–45. doi: 10.2147/IJN.S170680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Zhang L, Hao C, Yao S, Tang R, Guo W, Cong H, Li J, Bao L, Wang D, Li Y, Yu X, et al. Exosomal miRNA profiling to identify nanoparticle phagocytic mechanisms. Small. 2018;14(15):e1704008. doi: 10.1002/smll.201704008. Erratum in: Small. 2021;17(31):e2103783. doi:10.1002/smll.202103783. [DOI] [PubMed] [Google Scholar]
- 13. Yu B, Zhang X, Li X. Exosomes derived from mesenchymal stem cells. Int J Mol Sci. 2014;15(3):4142–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Mendt M, Rezvani K, Shpall E. Mesenchymal stem cell-derived exosomes for clinical use. Bone Marrow Transplant. 2019;54(suppl 2):789–92. [DOI] [PubMed] [Google Scholar]
- 15. Suzuki E, Fujita D, Takahashi M, Oba S, Nishimatsu H. Therapeutic effects of mesenchymal stem cell-derived exosomes in cardiovascular disease. Adv Exp Med Biol. 2017;998:179–85. [DOI] [PubMed] [Google Scholar]
- 16. Rezakhani L, Kelishadrokhi AF, Soleimanizadeh A, Rahmati S. Mesenchymal stem cell (MSC)-derived exosomes as a cell-free therapy for patients Infected with COVID-19: real opportunities and range of promises. Chem Phys Lipids. 2021;234:105009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Casado-Díaz A, Quesada-Gómez JM, Dorado G. Extracellular vesicles derived from mesenchymal stem cells (MSC) in regenerative medicine: applications in skin wound healing. Front Bioeng Biotechnol. 2020;8:146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Hu N, Cai Z, Jiang X, Wang C, Tang T, Xu T, Chen H, Li X, Du X, Cui W. Hypoxia-pretreated ADSC-derived exosome-embedded hydrogels promote angiogenesis and accelerate diabetic wound healing. Acta Biomater. 2023;157:175–86. doi: 10.1016/j.actbio.2022.11.057. [DOI] [PubMed] [Google Scholar]
- 19. Pan Y, Wu W, Jiang X, Liu Y. Mesenchymal stem cell-derived exosomes in cardiovascular and cerebrovascular diseases: from mechanisms to therapy. Biomed Pharmacother. 2023;163:114817. doi: 10.1016/j.biopha.2023.114817. [DOI] [PubMed] [Google Scholar]
- 20. Ivosevic Z, Ljujic B, Pavlovic D, Matovic V, Gazdic Jankovic M. Mesenchymal stem cell–derived extracellular vesicles: new soldiers in the war on immune-mediated diseases. Cell Transplant. 2023;32:9636897231207194. doi: 10.1177/09636897231207194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Simpson RJ, Kalra H, Mathivanan S. ExoCarta as a resource for exosomal research. J Extracell Vesicles. Epub 2012 Apr 16. doi: 10.3402/jev.v1i0.18374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Simons M, Raposo G. Exosomes: vesicular carriers for intercellular communication. Curr Opin Cell Biol. 2009;21:575–81. [DOI] [PubMed] [Google Scholar]
- 23. Mathivanan S, Ji H, Simpson RJ. Exosomes: extracellular organelles important in intercellular communication. J Proteomics. 2010;73:1907–20. [DOI] [PubMed] [Google Scholar]
- 24. Gross JC, Chaudhary V, Bartscherer K, Boutros M. Active Wnt proteins are secreted on exosomes. Nat Cell Biol. 2012;14(10):1036–45. [DOI] [PubMed] [Google Scholar]
- 25. van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19(4):213–28. [DOI] [PubMed] [Google Scholar]
- 26. Matsuzaka Y, Yashiro R. Therapeutic strategy of mesenchymal-stem-cell-derived extracellular vesicles as regenerative medicine. Int J Mol Sci. 2022;23(12):6480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Skotland T, Hessvik NP, Sandvig K, Llorente A. Exosomal lipid composition and the role of ether lipids and phosphoinositides in exosome biology. J Lipid Res. 2019;60(1):9–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Maroto R, Zhao Y, Jamaluddin M, Popov VL, Wang H, Kalubowilage M, Zhang Y, Luisi J, Sun H, Culbertson CT, Bossmann SH, et al. Effects of storage temperature on airway exosome integrity for diagnostic and functional analyses. J Extracell Vesicles. 2017;6(1):1359478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Duan W, Lopez MJ, Hicok K. Adult multipotent stromal cell cryopreservation: pluses and pitfalls. Vet Surg. 2018;47(1):19–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Katsuda T, Tsuchiya R, Kosaka N, Yoshioka Y, Takagaki K, Oki K, Takeshita F, Sakai Y, Kuroda M, Ochiya T. Human adipose tissue-derived mesenchymal stem cells secrete functional neprilysin-bound exosomes. Sci Rep. 2013;3:1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Tracy SA, Ahmed A, Tigges JC, Ericsson M, Pal AK, Zurakowski D, Fauza DO. A comparison of clinically relevant sources of mesenchymal stem cell-derived exosomes: bone marrow and amniotic fluid. J Pediatr Surg. 2019;54(1):86–90. [DOI] [PubMed] [Google Scholar]
- 32. Angulski AB, Capriglione LG, Batista M, Marcon BH, Senegaglia AC, Stimamiglio MA, Correa A. The protein content of extracellular vesicles derived from expanded human umbilical cord blood-derived CD133(+) and human bone marrow-derived mesenchymal stem cells partially explains why both sources are advantageous for regenerative medicine. Stem Cell Rev Rep. 2017;13(2):244–57. [DOI] [PubMed] [Google Scholar]
- 33. van Balkom BWM, Gremmels H, Giebel B, Lim SK. Proteomic signature of mesenchymal stromal cell-derived small extracellular vesicles. Proteomics. 2019;19(1–2):e1800163. [DOI] [PubMed] [Google Scholar]
- 34. Song A, Wang J, Tong Y, Fang J, Zhang Y, Zhang H, Ruan H, Wang K, Liu Y. BKCa channels regulate the immunomodulatory properties of WJ-MSCs by affecting the exosome protein profiles during the inflammatory response. Stem Cell Res Ther. 2020;11(1):440. doi: 10.1186/s13287-020-01952-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Wei H, Rongchun C, Fangjun Z, et al. Study on bone regeneration mechanism of exosomes derived from bone marrow mesenchymal stem cells. Chinese J Osteoporosis. 2019;025(4):477–83. [Google Scholar]
- 36. 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. doi: 10.3389/fbioe.2020.564731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Zhao Y, Sun X, Cao W, Ma J, Sun L, Qian H, Zhu W, Xu W. Exosomes derived from human umbilical cord mesenchymal stem cells relieve acute myocardial ischemic injury. Stem Cells Int. 2015;2015:761643. doi: 10.1155/2015/761643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Gurung S, Perocheau D, Touramanidou L, Baruteau J. The exosome journey: from biogenesis to uptake and intracellular signalling. Cell Commun Signal. 2021;19(1):47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Abels ER, Breakefield XO. Introduction to extracellular vesicles: biogenesis, RNA cargo selection, content, release, and uptake. Cell Mol Neurobiol. 2016;36(3):301–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255–89. [DOI] [PubMed] [Google Scholar]
- 41. Bu T, Li Z, Hou Y, Sun W, Zhang R, Zhao L, Wei M, Yang G, Yuan L. Exosome-mediated delivery of inflammation-responsive Il-10 mRNA for controlled atherosclerosis treatment. Theranostics. 2021;11(20):9988–10000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Zhang J, Li S, Li L, Li M, Guo C, Yao J, Mi S. Exosome and exosomal microRNA: trafficking, sorting, and function. Genomics Proteomics Bioinformatics. 2015;13(1):17–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Galliot B, Crescenzi M, Jacinto A, Tajbakhsh S. Trends in tissue repair and regeneration. Development. 2017;144(3):357–64. [DOI] [PubMed] [Google Scholar]
- 44. Huang J, Xiong J, Yang L, Zhang J, Sun S, Liang Y. Cell-free exosome-laden scaffolds for tissue repair. Nanoscale. 2021;13(19):8740–50. [DOI] [PubMed] [Google Scholar]
- 45. Kim YG, Choi J, Kim K. Mesenchymal stem cell-derived exosomes for effective cartilage tissue repair and treatment of osteoarthritis. Biotechnol J. 2020;15(12):e2000082. [DOI] [PubMed] [Google Scholar]
- 46. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Ren S, Chen J, Duscher D, Liu Y, Guo G, Kang Y, Xiong H, Zhan P, Wang Y, Wang C, Machens HG, et al. Microvesicles from human adipose stem cells promote wound healing by optimizing cellular functions via AKT and ERK signaling pathways. Stem Cell Res Ther. 2019;10(1):47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Zhao G, Ge Y, Zhang C, Zhang L, Xu J, Qi L, Li W. Progress of mesenchymal stem cell-derived exosomes in tissue repair. Curr Pharm Des. 2020;26(17):2022–37. [DOI] [PubMed] [Google Scholar]
- 49. Yaghoubi Y, Movassaghpour A, Zamani M, Zamani M, Talebi M, Mehdizadeh A, Yousefi M. Human umbilical cord mesenchymal stem cells derived-exosomes in diseases treatment. Life Sci. 2019;233:116733. [DOI] [PubMed] [Google Scholar]
- 50. An T, Chen Y, Tu Y, Lin P. Mesenchymal stromal cell-derived extracellular vesicles in the treatment of diabetic foot ulcers: application and challenges. Stem Cell Rev Rep. 2021;17(2):369–78. [DOI] [PubMed] [Google Scholar]
- 51. Harrell CR, Jovicic N, Djonov V, Arsenijevic N, Volarevic V. Mesenchymal stem cell-derived exosomes and other extracellular vesicles as new remedies in the therapy of inflammatory diseases. Cells. 2019;8(12):1605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Qian X, An N, Ren Y, Yang C, Zhang X, Li L. Immunosuppressive effects of mesenchymal stem cells-derived exosomes. Stem Cell Rev Rep. 2021;17(2):411–27. [DOI] [PubMed] [Google Scholar]
- 53. Ribeiro A, Laranjeira P, Mendes S, Velada I, Leite C, Andrade P, Santos F, Henriques A, Grãos M, Cardoso CM, Martinho A, et al. Mesenchymal stem cells from umbilical cord matrix, adipose tissue and bone marrow exhibit different capability to suppress peripheral blood B, natural killer and T cells. Stem Cell Res Ther. 2013;4(5):125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Castro-Manrreza ME, Montesinos JJ. Immunoregulation by mesenchymal stem cells: biological aspects and clinical applications. J Immunol Res. 2015;2015:394917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Shin H, Ryu HH, Kwon O, Park BS, Jo SJ. Clinical use of conditioned media of adipose tissue-derived stem cells in female pattern hair loss: a retrospective case series study. Int J Dermatol. 2015;54(6):730–35. [DOI] [PubMed] [Google Scholar]
- 56. TI D, Hao H, Tong C, Liu J, Dong L, Zheng J, Zhao Y, Liu H, Fu X, Han W. LPS-preconditioned mesenchymal stromal cells modify macrophage polarization for resolution of chronic inflammation via exosome-shuttled let-7b. J Transl Med. 2015;13:308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Fan Y, Herr F, Vernochet A, Mennesson B, Oberlin E, Durrbach A. Human fetal liver mesenchymal stem cell-derived exosomes impair natural killer cell function. Stem Cells Dev. 2019;28(1):44–55. [DOI] [PubMed] [Google Scholar]
- 58. Gowen A, Shahjin F, Chand S, Odegaard KE, Yelamanchili SV. Mesenchymal stem cell-derived extracellular vesicles: challenges in clinical applications. Front Cell Dev Biol. 2020;8:149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Nekanti U, Mohanty L, Venugopal P, Balasubramanian S, Totey S, Ta M. Optimization and scale-up of Wharton’s jelly-derived mesenchymal stem cells for clinical applications. Stem Cell Res. 2010;5(3):244–54. [DOI] [PubMed] [Google Scholar]
- 60. Lee SY, Lee JW. 3D spheroid cultures of stem cells and exosome applications for cartilage repair. Life. 2022;12(7):939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Gobin J, Muradia G, Mehic J, Westwood C, Couvrette L, Stalker A, Bigelow S, Luebbert CC, Bissonnette FS, Johnston MJW, Sauvé S, et al. Hollow-fiber bioreactor production of extracellular vesicles from human bone marrow mesenchymal stromal cells yields nanovesicles that mirrors the immuno-modulatory antigenic signature of the producer cell. Stem Cell Res Ther. 2021;12(1):127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Mennan C, Garcia J, Roberts S, Hulme C, Wright K. A comprehensive characterisation of large-scale expanded human bone marrow and umbilical cord mesenchymal stem cells. Stem Cell Res Ther. 2019;10(1):99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Lener T, Gimona M, Aigner L, Börger V, Buzas E, Camussi G, Chaput N, Chatterjee D, Court FA, Del Portillo HA, O’Driscoll L, et al. Applying extracellular vesicles based therapeutics in clinical trials—an ISEV position paper. J Extracell Vesicles. 2015;4:30087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Visan KS, Lobb RJ, Ham S, Lima LG, Palma C, Edna CPZ, Wu LY, Gowda H, Datta KK, Hartel G, Salomon C, et al. Comparative analysis of tangential flow filtration and ultracentrifugation, both combined with subsequent size exclusion chromatography, for the isolation of small extracellular vesicles. J Extracell Vesicles. 2022;11(9):e12266. doi: 10.1002/jev2.12266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Hua X, Zhu Q, Liu Y, Zhou S, Huang P, Li Q, Liu S. A double tangential flow filtration-based microfluidic device for highly efficient separation and enrichment of exosomes. Analytica Chimica Acta. 2023:1258. [DOI] [PubMed] [Google Scholar]
- 66. Chen BY, Sung CW, Chen C, Cheng CM, Lin DP, Huang CT, Hsu MY. Advances in exosomes technology. Clin Chim Acta. 2019;493:14–19. [DOI] [PubMed] [Google Scholar]
- 67. Saunderson SC, Dunn AC, Crocker PR, McLellan AD. CD169 mediates the capture of exosomes in spleen and lymph node. Blood. 2014;123(2):208–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Momen-Heravi F. Isolation of extracellular vesicles by ultracentrifugation. Methods Mol Biol. 2017;1660:25–32. [DOI] [PubMed] [Google Scholar]
- 69. Lane RE, Korbie D, Trau M, Hill MM. Purification protocols for extracellular vesicles. Methods Mol Biol. 2017;1660:111–30. [DOI] [PubMed] [Google Scholar]
- 70. Mizutani K, Terazawa R, Kameyama K, Kato T, Horie K, Tsuchiya T, Seike K, Ehara H, Fujita Y, Kawakami K, Ito M, Deguchi T. Isolation of prostate cancer-related exosomes. Anticancer Res. 2014;34(7):3419–23. [PubMed] [Google Scholar]
- 71. Cho S, Jo W, Heo Y, Kang JY, Kwak R, Park J. Isolation of extracellular vesicle from blood plasma using electrophoretic migration through porous membrane. Sensors Actuators B Chem. 2016;233:289–97. [Google Scholar]
- 72. Li P, Kaslan M, Lee SH, Yao J, Gao Z. Progress in exosome isolation techniques. Theranostics. 2017;7(3):789–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Moon GJ, Sung JH, Kim DH, Kim EH, Cho YH, Son JP, Cha JM, Bang OY. Application of mesenchymal stem cell-derived extracellular vesicles for stroke: biodistribution and MicroRNA study. Transl Stroke Res. 2019;10(5):509–21. [DOI] [PubMed] [Google Scholar]
- 74. Salunkhe S, Dheeraj, Basak M, Chitkara D, Mittal A. Surface functionalization of exosomes for target-specific delivery and in vivo imaging & tracking: strategies and significance. J Control Release. 2020;326:599–614. [DOI] [PubMed] [Google Scholar]
- 75. Gu C, Feng J, Waqas A, Deng Y, Zhang Y, Chen W, Long J, Huang S, Chen L. Technological advances of 3D scaffold-based stem cell/exosome therapy in tissues and organs. Front Cell Dev Biol. 2021;9:709204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Lin J, Wang Z, Huang J, Tang S, Saiding Q, Zhu Q, Cui W. Microenvironment-protected exosome-hydrogel for facilitating endometrial regeneration, fertility restoration, and live birth of offspring. Small. 2021;17(11):e2007235. [DOI] [PubMed] [Google Scholar]
- 77. Liu B, Lee BW, Nakanishi K, Villasante A, Williamson R, Metz J, Kim J, Kanai M, Bi L, Brown K, Di Paolo G, et al. Cardiac recovery via extended cell-free delivery of extracellular vesicles secreted by cardiomyocytes derived from induced pluripotent stem cells. Nat Biomed Eng. 2018;2(5):293–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Poongodi R, Chen YL, Yang TH, Huang YH, Yang KD, Lin HC, Cheng JK. Bio-scaffolds as cell or exosome carriers for nerve injury repair. Int J Mol Sci. 2021;22(24):13347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Lu X, Dai S, Huang B, Li S, Wang P, Zhao Z, Li X, Li N, Wen J, Sun Y, Man Z, et al. Exosomes loaded a smart bilayer-hydrogel scaffold with ROS-scavenging and macrophage-reprogramming properties for repairing cartilage defect. Bioact Mater. 2024;38:137–53. doi: 10.1016/j.bioactmat.2024.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Liu X, Liu Y, Zhou J, Yu X, Wan J, Wang J, Lei S, Zhang Z, Zhang L, Wang S. Porous collagen sponge loaded with large efficacy-potentiated exosome-mimicking nanovesicles for diabetic skin wound healing. ACS Biomater Sci Eng. 2024;10(2):975–86. doi: 10.1021/acsbiomaterials.3c01282. [DOI] [PubMed] [Google Scholar]



