260
Views
12
CrossRef citations to date
0
Altmetric
Review

Potential of Exosomes as Cell-Free Therapy in Articular Cartilage Regeneration: A Review

ORCID Icon, , ORCID Icon, , ORCID Icon, ORCID Icon & ORCID Icon show all
Pages 6749-6781 | Published online: 01 Oct 2021

References

  • Cui A, Li H, Wang D, Zhong J, Chen Y, Lu H. Global, regional prevalence, incidence and risk factors of knee osteoarthritis in population-based studies. EClinicalMedicine. 2020;29–30. doi:10.1016/j.eclinm.2020.100587
  • Mobasheri A, Batt M. An update on the pathophysiology of osteoarthritis. Ann Phys Rehabil Med. 2016;59(5–6):333–339. doi:10.1016/j.rehab.2016.07.004
  • Abramoff B, Caldera FE. Osteoarthritis: pathology, diagnosis, and treatment options. Med Clin North Am. 2020;104(2):293–311. doi:10.1016/j.mcna.2019.10.007
  • Buckwalter JA, Mankin HJ. Articular cartilage: degeneration and osteoarthritis, repair, regeneration, and transplantation. Instr Course Lect. 1998;47:487–504.
  • Re’Em T, Witte F, Willbold E, Ruvinov E, Cohen S. Simultaneous regeneration of articular cartilage and subchondral bone induced by spatially presented TGF-beta and BMP-4 in a bilayer affinity binding system. Acta Biomater. 2012;8(9):3283–3293. doi:10.1016/j.actbio.2012.05.014
  • van Dijk CN, Reilingh ML, Zengerink M, van Bergen CJA. Osteochondral defects in the ankle: why painful? Knee Surg Sports Traumatol Arthrosc. 2010;18(5):570–580. doi:10.1007/s00167-010-1064-x
  • Anandacoomarasamy A, Leibman S, Smith G, et al. Weight loss in obese people has structure-modifying effects on medial but not on lateral knee articular cartilage. Ann Rheum Dis. 2012;71(1):26–32. doi:10.1136/ard.2010.144725
  • Lund H, Weile U, Christensen R, et al. A randomized controlled trial of aquatic and land-based exercise in patients with knee osteoarthritis. J Rehabil Med. 2008;40(2):137–144. doi:10.2340/16501977-0134
  • Uthman OA, Van Der Windt DA, Jordan JL, et al. Exercise for lower limb osteoarthritis: systematic review incorporating trial sequential analysis and network meta-analysis. BMJ (Online). 2013;347(7928). doi:10.1136/bmj.f5555
  • Towheed T, Maxwell L, Judd M, Catton M, Hochberg MC, Wells GA. Acetaminophen for osteoarthritis. Cochrane Database Syst Rev. 2006;2010(1). doi:10.1002/14651858.cd004257.pub2
  • Richette P, Latourte A, Frazier A. Safety and efficacy of paracetamol and NSAIDs in osteoarthritis: which drug to recommend? Expert Opin Drug Saf. 2015;14(8):1259–1268. doi:10.1517/14740338.2015.1056776
  • SooHoo NF, Lieberman JR, Ko CY, Zingmond DS. Factors predicting complication rates following total knee replacement. J Bone Joint Surg Am. 2006;88(3):480–485. doi:10.2106/JBJS.E.00629
  • Atkinson HDE. The negatives of knee replacement surgery: complications and the dissatisfied patient. Orthop Trauma. 2017;31(1):25–33. doi:10.1016/j.mporth.2016.09.011
  • Badekas T, Takvorian M, Souras N. Treatment principles for osteochondral lesions in foot and ankle. Int Orthop. 2013;37(9):1697–1706. doi:10.1007/s00264-013-2076-1
  • Block TJ, Garza JR. Regenerative cells for the management of osteoarthritis and joint disorders: a concise literature review. Aesthet Surg J. 2017;37(suppl_3):S9–S15. doi:10.1093/asj/sjx015
  • Murphy JM, Fink DJ, Hunziker EB, Barry FP. Stem cell therapy in a caprine model of osteoarthritis. Arthritis Rheum. 2003;48(12):3464–3474. doi:10.1002/art.11365
  • Desando G, Cavallo C, Sartoni F, et al. Intra-articular delivery of adipose derived stromal cells attenuates osteoarthritis progression in an experimental rabbit model. Arthritis Res Ther. 2013;15(1):R22. doi:10.1186/ar4156
  • Murata D, Kunitomi Y, Harada K, Tokunaga S, Takao S, Nakayama K. Osteochondral regeneration using scaffold-free constructs of adipose tissue-derived mesenchymal stem cells made by a bio three-dimensional printer with a needle-array in rabbits. Regen Ther. 2020;15:77–89. doi:10.1016/j.reth.2020.05.004
  • Yamasaki A, Kunitomi Y, Murata D, et al. Osteochondral regeneration using constructs of mesenchymal stem cells made by bio three-dimensional printing in mini-pigs. J Orthop Res. 2019;37(6):1398–1408. doi:10.1002/jor.24206
  • Lee WS, Kim HJ, Kim KI, Kim GB, Jin W. Intra-articular injection of autologous adipose tissue-derived mesenchymal stem cells for the treatment of knee osteoarthritis: a phase IIb, randomized, placebo-controlled clinical trial. Stem Cells Transl Med. 2019;8(6):504–511. doi:10.1002/sctm.18-0122
  • Freitag J, Bates D, Wickham J, et al. Adipose-derived mesenchymal stem cell therapy in the treatment of knee osteoarthritis: a randomized controlled trial. Regen Med. 2019;14(3):213–230. doi:10.2217/rme-2018-0161
  • Freitag J, Wickham J, Shah K, Tenen A. Effect of autologous adipose-derived mesenchymal stem cell therapy in the treatment of an osteochondral lesion of the ankle. BMJ Case Rep. 2020;13(7):e234595. doi:10.1136/bcr-2020-234595
  • Wei ZJ, Wang QQ, Cui ZG, Inadera H, Jiang X, Wu CA. Which is the most effective one in knee osteoarthritis treatment from mesenchymal stem cells obtained from different sources? - A systematic review with conventional and network meta-analyses of randomized controlled trials. Ann Transl Med. 2021;9(6):452. doi:10.21037/atm-20-5116
  • Song Y, Zhang J, Xu H, et al. Mesenchymal stem cells in knee osteoarthritis treatment: a systematic review and meta-analysis. J Orthop Transl. 2020;24:121–130. doi:10.1016/j.jot.2020.03.015
  • Lukomska B, Stanaszek L, Zuba-Surma E, Legosz P, Sarzynska S, Drela K. Challenges and controversies in human mesenchymal stem cell therapy. Stem Cells Int. 2019;2019:e9628536. doi:10.1155/2019/9628536
  • Bonzo Di LV, Ferrero I, Cravanzola C, et al. Human mesenchymal stem cells as a two-edged sword in hepatic regenerative medicine: engraftment and hepatocyte differentiation versus profibrogenic potential. Gut. 2008;57(2):223–231. doi:10.1136/gut.2006.111617
  • Jeong JO, Han JW, Kim JM, et al. Malignant tumor formation after transplantation of short-term cultured bone marrow mesenchymal stem cells in experimental myocardial infarction and diabetic neuropathy. Circ Res. 2011;108(11):1340–1347. doi:10.1161/CIRCRESAHA.110.239848
  • De Boeck A, Pauwels P, Hensen K, et al. Bone marrow-derived mesenchymal stem cells promote colorectal cancer progression through paracrine neuregulin 1/HER3 signalling. Gut. 2013;62(4):550–560. doi:10.1136/gutjnl-2011-301393
  • Spaeth E, Dembinski J, Sasser A, et al. Mesenchymal stem cell transition to tumor-associated fibroblasts contributes to fibrovascular network expansion and tumor progression. PLoS One. 2009;4(4):e4992. doi:10.1371/journal.pone.0004992
  • Karnoub AE, Dash AB, Vo AP, et al. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 2007;449(7162):557–563. doi:10.1038/nature06188
  • Kim JS, Lee JH, Kwon O, et al. Rapid deterioration of preexisting renal insufficiency after autologous mesenchymal stem cell therapy. Kidney Res Clin Pract. 2017;36(2):200–204. doi:10.23876/j.krcp.2017.36.2.200
  • Broekema M, Harmsen MC, van Luyn MJA, et al. Bone marrow-derived myofibroblasts contribute to the renal interstitial myofibroblast population and produce procollagen I after ischemia/reperfusion in rats. J Am Soc Nephrol. 2007;18(1):165–175. doi:10.1681/ASN.2005070730
  • Aslam M, Baveja R, Liang OD, et al. Bone marrow stromal cells attenuate lung injury in a murine model of neonatal chronic lung disease. Am J Respir Crit Care Med. 2009;180(11):1122–1130. doi:10.1164/rccm.200902-0242OC
  • Timmers L, Lim SK, Arslan F, et al. Reduction of myocardial infarct size by human mesenchymal stem cell conditioned medium. Stem Cell Res. 2007;1(2):129–137. doi:10.1016/j.scr.2008.02.002
  • Goolaerts A, Pellan-Randrianarison N, Larghero J, et al. Conditioned media from mesenchymal stromal cells restore sodium transport and preserve epithelial permeability in an in vitro model of acute alveolar injury. Am J Physiol Lung Cell Mol Physiol. 2014;306(11):L975–L985. doi:10.1152/ajplung.00242.2013
  • Liang X, Ding Y, Zhang Y, Tse HF, Lian Q. Paracrine mechanisms of mesenchymal stem cell-based therapy: current status and perspectives. Cell Transplant. 2014;23(9):1045–1059. doi:10.3727/096368913X667709
  • 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. Biomater Sci. 2017;6(1):60–78. doi:10.1039/c7bm00479f
  • Zhao AG, Shah K, Cromer B, Sumer H. Mesenchymal stem cell-derived extracellular vesicles and their therapeutic potential. Stem Cells Int. 2020;2020:e8825771. doi:10.1155/2020/8825771
  • Maas SLN, Breakefield XO, Weaver AM. Extracellular vesicles: unique intercellular delivery vehicles. Trends Cell Biol. 2017;27(3):172–188. doi:10.1016/j.tcb.2016.11.003
  • Lai RC, Arslan F, Lee MM, et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010;4(3):214–222. doi:10.1016/j.scr.2009.12.003
  • Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30(1):255–289. doi:10.1146/annurev-cellbio-101512-122326
  • Lo Cicero A, Stahl PD, Raposo G. Extracellular vesicles shuffling intercellular messages: for good or for bad. Curr Opin Cell Biol. 2015;35:69–77. doi:10.1016/j.ceb.2015.04.013
  • Teng X, Chen L, Chen W, Yang J, Yang Z, Shen Z. Mesenchymal stem cell-derived exosomes improve the microenvironment of infarcted myocardium contributing to angiogenesis and anti-inflammation. Cell Physiol Biochem. 2015;37(6):2415–2424. doi:10.1159/000438594
  • Li T, Yan Y, Wang B, et al. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate liver fibrosis. Stem Cells Dev. 2013;22(6):845–854. doi:10.1089/scd.2012.0395
  • Zhang J, Guan J, Niu X, et al. Exosomes released from human induced pluripotent stem cells-derived MSCs facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis. J Transl Med. 2015;13(1):49. doi:10.1186/s12967-015-0417-0
  • Ni Z, Zhou S, Li S, et al. Exosomes: roles and therapeutic potential in osteoarthritis. Bone Res. 2020;8:25. doi:10.1038/s41413-020-0100-9
  • Toh WS, Lai RC, Hui JHP, Lim SK. MSC exosome as a cell-free MSC therapy for cartilage regeneration: implications for osteoarthritis treatment. Semin Cell Dev Biol. 2017;67:56–64. doi:10.1016/j.semcdb.2016.11.008
  • Kim YG, Park U, Park BJ, Kim K. Exosome-mediated bidirectional signaling between mesenchymal stem cells and chondrocytes for enhanced chondrogenesis. Biotechnol Bioprocess Eng. 2019;24(5):734–744. doi:10.1007/s12257-019-0332-y
  • Théry C, Witwer KW, Aikawa E, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the international society for extracellular vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7(1):1535750. doi:10.1080/20013078.2018.1535750
  • Yáñez-Mó M, Siljander PRM, Andreu Z, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles. 2015;4(1):27066. doi:10.3402/jev.v4.27066
  • Willms E, Cabañas C, Mäger I, Wood MJA, Vader P. Extracellular vesicle heterogeneity: subpopulations, isolation techniques, and diverse functions in cancer progression. Front Immunol. 2018;9:738. doi:10.3389/fimmu.2018.00738
  • Zhang B, Tian X, Qu Z, Liu J, Yang L, Zhang W. Efficacy of extracellular vesicles from mesenchymal stem cells on osteoarthritis in animal models: a systematic review and meta-analysis. Nanomedicine (Lond). 2021;16(15):1297–1310. doi:10.2217/nnm-2021-0047
  • Tan SSH, Tjio CKE, Wong JRY, et al. Mesenchymal stem cell exosomes for cartilage regeneration: a systematic review of preclinical in vivo studies. Tissue Eng Part B Rev. 2021;27(1):1–13. doi:10.1089/ten.TEB.2019.0326
  • To K, Romain K, Mak C, Kamaraj A, Henson F, Khan W. The treatment of cartilage damage using human mesenchymal stem cell-derived extracellular vesicles: a systematic review of in vivo studies. Front Bioeng Biotechnol. 2020;8:580. doi:10.3389/fbioe.2020.00580
  • Hooijmans CR, Rovers MM, de Vries RBM, Leenaars M, Ritskes-Hoitinga M, Langendam MW. SYRCLE’s risk of bias tool for animal studies. BMC Med Res Methodol. 2014;14(1):43. doi:10.1186/1471-2288-14-43
  • Chen X, Shi Y, Xue P, Ma X, Li J, Zhang J. Mesenchymal stem cell-derived exosomal microRNA-136-5p inhibits chondrocyte degeneration in traumatic osteoarthritis by targeting ELF3. Arthritis Res Ther. 2020;22(1):256. doi:10.1186/s13075-020-02325-6
  • Jin Z, Ren J, Qi S. Human bone mesenchymal stem cells-derived exosomes overexpressing microRNA-26a-5p alleviate osteoarthritis via down-regulation of PTGS2. Int Immunopharmacol. 2020;78:105946. doi:10.1016/j.intimp.2019.105946
  • Liu X, Yang Y, Li Y, et al. Integration of stem cell-derived exosomes with in situ hydrogel glue as a promising tissue patch for articular cartilage regeneration. Nanoscale. 2017;9(13):4430–4438. doi:10.1039/c7nr00352h
  • Mao G, Zhang Z, Hu S, et al. Exosomes derived from miR-92a-3p-overexpressing human mesenchymal stem cells enhance chondrogenesis and suppress cartilage degradation via targeting WNT5A. Stem Cell Res Ther. 2018;9(1):247. doi:10.1186/s13287-018-1004-0
  • Tao SC, Yuan T, Zhang YL, Yin WJ, Guo SC, Zhang CQ. Exosomes derived from miR-140-5p-overexpressing human synovial mesenchymal stem cells enhance cartilage tissue regeneration and prevent osteoarthritis of the knee in a rat model. Theranostics. 2017;7(1):180–195. doi:10.7150/thno.17133
  • Wang Y, Yu D, Liu Z, et al. Exosomes from embryonic mesenchymal stem cells alleviate osteoarthritis through balancing synthesis and degradation of cartilage extracellular matrix. Stem Cell Res Ther. 2017;8(1):189. doi:10.1186/s13287-017-0632-0
  • Wang Z, Yan K, Ge G, et al. Exosomes derived from miR-155-5p–overexpressing synovial mesenchymal stem cells prevent osteoarthritis via enhancing proliferation and migration, attenuating apoptosis, and modulating extracellular matrix secretion in chondrocytes. Cell Biol Toxicol. 2021;37(1):85–96. doi:10.1007/s10565-020-09559-9
  • Wong KL, Zhang S, Wang M, et al. Intra-articular injections of mesenchymal stem cell exosomes and hyaluronic acid improve structural and mechanical properties of repaired cartilage in a rabbit model. Arthroscopy. 2020;36(8):2215–2228.e2. doi:10.1016/j.arthro.2020.03.031
  • Wu J, Kuang L, Chen C, et al. MiR-100-5p-abundant exosomes derived from infrapatellar fat pad MSCs protect articular cartilage and ameliorate gait abnormalities via inhibition of mTOR in osteoarthritis. Biomaterials. 2019;206:87–100. doi:10.1016/j.biomaterials.2019.03.022
  • Yan L, Liu G, Wu X. Exosomes derived from umbilical cord mesenchymal stem cells in mechanical environment show improved osteochondral activity via upregulation of LncRNA H19. J Orthop Transl. 2021;26:111–120. doi:10.1016/j.jot.2020.03.005
  • Yan L, Wu X. Exosomes produced from 3D cultures of umbilical cord mesenchymal stem cells in a hollow-fiber bioreactor show improved osteochondral regeneration activity. Cell Biol Toxicol. 2020;36(2):165–178. doi:10.1007/s10565-019-09504-5
  • Zavatti M, Beretti F, Casciaro F, Bertucci E, Maraldi T. Comparison of the therapeutic effect of amniotic fluid stem cells and their exosomes on monoiodoacetate-induced animal model of osteoarthritis. BioFactors. 2020;46(1):106–117. doi:10.1002/biof.1576
  • Zhang S, Chu WC, Lai RC, Lim SK, Hui JHP, Toh WS. Exosomes derived from human embryonic mesenchymal stem cells promote osteochondral regeneration. Osteoarthritis Cartilage. 2016;24(12):2135–2140. doi:10.1016/j.joca.2016.06.022
  • Zhang S, Chuah SJ, Lai RC, Hui JHP, Lim SK, Toh WS. MSC exosomes mediate cartilage repair by enhancing proliferation, attenuating apoptosis and modulating immune reactivity. Biomaterials. 2018;156:16–27. doi:10.1016/j.biomaterials.2017.11.028
  • Zhang S, Teo KYW, Chuah SJ, Lai RC, Lim SK, Toh WS. MSC exosomes alleviate temporomandibular joint osteoarthritis by attenuating inflammation and restoring matrix homeostasis. Biomaterials. 2019;200:35–47. doi:10.1016/j.biomaterials.2019.02.006
  • Zhou X, Liang H, Hu X, et al. BMSC-derived exosomes from congenital polydactyly tissue alleviate osteoarthritis by promoting chondrocyte proliferation. Cell Death Discov. 2020;6(1):142. doi:10.1038/s41420-020-00374-z
  • Zhu Y, Wang Y, Zhao B, et al. Comparison of exosomes secreted by induced pluripotent stem cell-derived mesenchymal stem cells and synovial membrane-derived mesenchymal stem cells for the treatment of osteoarthritis. Stem Cell Res Ther. 2017;8(1):64. doi:10.1186/s13287-017-0510-9
  • Chen P, Zheng L, Wang Y, et al. Desktop-stereolithography 3D printing of a radially oriented extracellular matrix/mesenchymal stem cell exosome bioink for osteochondral defect regeneration. Theranostics. 2019;9(9):2439–2459. doi:10.7150/thno.31017
  • Cosenza S, Ruiz M, Toupet K, Jorgensen C, Noël D. Mesenchymal stem cells derived exosomes and microparticles protect cartilage and bone from degradation in osteoarthritis. Sci Rep. 2017;7(1):16214. doi:10.1038/s41598-017-15376-8
  • He L, He T, Xing J, et al. Bone marrow mesenchymal stem cell-derived exosomes protect cartilage damage and relieve knee osteoarthritis pain in a rat model of osteoarthritis. Stem Cell Res Ther. 2020;11(1):276. doi:10.1186/s13287-020-01781-w
  • Jin Z, Ren J, Qi S. Exosomal miR-9-5p secreted by bone marrow-derived mesenchymal stem cells alleviates osteoarthritis by inhibiting syndecan-1. Cell Tissue Res. 2020;381(1):99–114. doi:10.1007/s00441-020-03193-x
  • Liang Y, Xu X, Li X, et al. Chondrocyte-targeted microRNA delivery by engineered exosomes toward a cell-free osteoarthritis therapy. ACS Appl Mater Interfaces. 2020;12(33):36938–36947. doi:10.1021/acsami.0c10458
  • Liu C, Li Y, Yang Z, Zhou Z, Lou Z, Zhang Q. Kartogenin enhances the therapeutic effect of bone marrow mesenchymal stem cells derived exosomes in cartilage repair. Nanomedicine (Lond). 2020;15(3):273–288. doi:10.2217/nnm-2019-0208
  • Wang R, Xu B, Xu H. TGF-β1 promoted chondrocyte proliferation by regulating Sp1 through MSC-exosomes derived miR-135b. Cell Cycle. 2018;17(24):2756–2765. doi:10.1080/15384101.2018.1556063
  • Wang Y, He SH, Liang X, Zhang XX, Li S-S, Li TF. ATF4-modified serum exosomes derived from osteoarthritic mice inhibit osteoarthritis by inducing autophagy. IUBMB Life. 2021;73(1):146–158. doi:10.1002/iub.2414
  • Zheng L, Wang Y, Qiu P, et al. Primary chondrocyte exosomes mediate osteoarthritis progression by regulating mitochondrion and immune reactivity. Nanomedicine (Lond). 2019;14(24):3193–3212. doi:10.2217/nnm-2018-0498
  • Liu X, Wang L, Ma C, Wang G, Zhang Y, Sun S. Exosomes derived from platelet-rich plasma present a novel potential in alleviating knee osteoarthritis by promoting proliferation and inhibiting apoptosis of chondrocyte via Wnt/β-catenin signaling pathway. J Orthop Surg Res. 2019;14(1):470. doi:10.1186/s13018-019-1529-7
  • Qiu B, Xu X, Yi P, Hao Y. Curcumin reinforces MSC-derived exosomes in attenuating osteoarthritis via modulating the miR-124/NF-kB and miR-143/ROCK1/TLR9 signalling pathways. J Cell Mol Med. 2020;24(18):10855–10865. doi:10.1111/jcmm.15714
  • Zhang J, Rong Y, Luo C, Cui W. Bone marrow mesenchymal stem cell-derived exosomes prevent osteoarthritis by regulating synovial macrophage polarization. Aging (Albany NY). 2020;12(24):25138–25152. doi:10.18632/aging.104110
  • Cai J, Wu J, Wang J, et al. Extracellular vesicles derived from different sources of mesenchymal stem cells: therapeutic effects and translational potential. Cell Biosci. 2020;10(1):69. doi:10.1186/s13578-020-00427-x
  • Liau LL, Al-Masawa ME, Koh B, et al. The potential of mesenchymal stromal cell as therapy in neonatal diseases. Front Pediatr. 2020;8:591693. doi:10.3389/fped.2020.591693
  • Looi QH, Eng SP, Liau LL, et al. Mesenchymal stem cell therapy for sports injuries-from research to clinical practice. Sains Malays. 2020;49(4):825–838. doi:10.17576/jsm-2020-4904-12
  • Gao F, Chiu SM, Motan DAL, et al. Mesenchymal stem cells and immunomodulation: current status and future prospects. Cell Death Dis. 2016;7(1):e2062. doi:10.1038/cddis.2015.327
  • Pittenger MF, Discher DE, Péault BM, Phinney DG, Hare JM, Caplan AI. Mesenchymal stem cell perspective: cell biology to clinical progress. NPJ Regen Med. 2019;4(1):22. doi:10.1038/s41536-019-0083-6
  • Lian J, Lv S, Liu C, et al. Effects of serial passage on the characteristics and cardiac and neural differentiation of human umbilical cord Wharton’s jelly-derived mesenchymal stem cells. Stem Cells Int. 2016;2016:9291013. doi:10.1155/2016/9291013
  • Liau LL, Looi QH, Chia WC, Subramaniam T, Ng MH, Law JX. Treatment of spinal cord injury with mesenchymal stem cells. Cell Biosci. 2020;10:112. doi:10.1186/s13578-020-00475-3
  • Lopez-Verrilli MA, Caviedes A, Cabrera A, Sandoval S, Wyneken U, Khoury M. Mesenchymal stem cell-derived exosomes from different sources selectively promote neuritic outgrowth. Neuroscience. 2016;320:129–139. doi:10.1016/j.neuroscience.2016.01.061
  • Del Fattore A, Luciano R, Saracino R, et al. Differential effects of extracellular vesicles secreted by mesenchymal stem cells from different sources on glioblastoma cells. Expert Opin Biol Ther. 2015;15(4):495–504. doi:10.1517/14712598.2015.997706
  • Katsuda T, Tsuchiya R, Kosaka N, et al. Human adipose tissue-derived mesenchymal stem cells secrete functional neprilysin-bound exosomes. Sci Rep. 2013;3(1):1197. doi:10.1038/srep01197
  • Álvarez-viejo M. Mesenchymal stem cells from different sources and their derived exosomes: a pre-clinical perspective. World J Stem Cells. 2020;12(2):100. doi:10.4252/wjsc.v12.i2.100
  • Nikfarjam S, Rezaie J, Zolbanin NM, Jafari R. Mesenchymal stem cell derived-exosomes: a modern approach in translational medicine. J Transl Med. 2020;18(1):449. doi:10.1186/s12967-020-02622-3
  • Yin K, Wang S, Zhao RC. Exosomes from mesenchymal stem/stromal cells: a new therapeutic paradigm. Biomark Res. 2019;7(1):8. doi:10.1186/s40364-019-0159-x
  • Tang Y, Zhou Y, Li H-J. Advances in mesenchymal stem cell exosomes: a review. Stem Cell Res Ther. 2021;12(1):71. doi:10.1186/s13287-021-02138-7
  • Kourembanas S. Exosomes: vehicles of intercellular signaling, biomarkers, and vectors of cell therapy. Annu Rev Physiol. 2015;77(1):13–27. doi:10.1146/annurev-physiol-021014-071641
  • Fu S, Wang Y, Xia X, Zheng JC. Exosome engineering: current progress in cargo loading and targeted delivery. NanoImpact. 2020;20:100261. doi:10.1016/j.impact.2020.100261
  • Gholami L, Nooshabadi VT, Shahabi S, et al. Extracellular vesicles in bone and periodontal regeneration: current and potential therapeutic applications. Cell Biosci. 2021;11(1):16. doi:10.1186/s13578-020-00527-8
  • 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. Nat Cell Biol. 2007;9(6):654–659. doi:10.1038/ncb1596
  • Wu C, Tian B, Qu X, et al. MicroRNAs play a role in chondrogenesis and osteoarthritis (review). Int J Mol Med. 2014;34(1):13–23. doi:10.3892/ijmm.2014.1743
  • Mihanfar A, Shakouri SK, Khadem-Ansari MH, et al. Exosomal miRNAs in osteoarthritis. Mol Biol Rep. 2020;47(6):4737–4748. doi:10.1007/s11033-020-05443-1
  • Peng B, Chen Y, Leong KW. MicroRNA delivery for regenerative medicine. Adv Drug Deliv Rev. 2015;88:108–122. doi:10.1016/j.addr.2015.05.014
  • Schwarzenbach H, Gahan PB. MicroRNA shuttle from cell-to-cell by exosomes and its impact in cancer. Noncoding RNA. 2019;5(1). doi:10.3390/ncrna5010028
  • Conigliaro A, Costa V, Lo Dico A, et al. CD90+ liver cancer cells modulate endothelial cell phenotype through the release of exosomes containing H19 lncRNA. Mol Cancer. 2015;14(1):155. doi:10.1186/s12943-015-0426-x
  • Zhang C, Wang P, Jiang P, et al. Upregulation of lncRNA HOTAIR contributes to IL-1β-induced MMP overexpression and chondrocytes apoptosis in temporomandibular joint osteoarthritis. Gene. 2016;586(2):248–253. doi:10.1016/j.gene.2016.04.016
  • Yang L, Zhai Y, Hao Y, Zhu Z, Cheng G. The regulatory functionality of exosomes derived from hUMSCs in 3D culture for Alzheimer’s disease therapy. Small. 2019;16:3. doi:10.1002/smll.201906273
  • Cao J, Wang B, Tang T, et al. Three-dimensional culture of MSCs produces exosomes with improved yield and enhanced therapeutic efficacy for cisplatin-induced acute kidney injury. Stem Cell Res Ther. 2020;11(1):206. doi:10.1186/s13287-020-01719-2
  • Liau LL, Hassan MN, Tang YL, Ng MH, Law JX. Feasibility of human platelet lysate as an alternative to foetal bovine serum for in vitro expansion of chondrocytes. Int J Mol Sci. 2021;22(3):1269. doi:10.3390/ijms22031269
  • Evans CH, Kraus VB, Setton LA. Progress in intra-articular therapy. Nat Rev Rheumatol. 2014;10(1):11–22. doi:10.1038/nrrheum.2013.159
  • Pinheiro A, Silva AM, Teixeira JH, et al. Extracellular vesicles: intelligent delivery strategies for therapeutic applications. J Control Release. 2018;289:56–69. doi:10.1016/j.jconrel.2018.09.019
  • Patil M, Henderson J, Luong H, Annamalai D, Sreejit G, Krishnamurthy P. The art of intercellular wireless communications: exosomes in heart disease and therapy. Front Cell Dev Biol. 2019;7:315. doi:10.3389/fcell.2019.00315
  • Quesenberry PJ, Aliotta J, Deregibus MC, Camussi G. Role of extracellular RNA-carrying vesicles in cell differentiation and reprogramming. Stem Cell Res Ther. 2015;6(1):153. doi:10.1186/s13287-015-0150-x
  • Qi H, Liu DP, Xiao DW, Tian DC, Su YW, Jin SF. Exosomes derived from mesenchymal stem cells inhibit mitochondrial dysfunction-induced apoptosis of chondrocytes via p38, ERK, and Akt pathways. In Vitro Cell Dev Biol Anim. 2019;55(3):203–210. doi:10.1007/s11626-019-00330-x
  • De Jong OG, Van Balkom BWM, Schiffelers RM, Bouten CVC, Verhaar MC. Extracellular vesicles: potential roles in regenerative medicine. Front Immunol. 2014;5:608. doi:10.3389/fimmu.2014.00608
  • Wang Y, Yao J, Cai L, et al. Bone-targeted extracellular vesicles from mesenchymal stem cells for osteoporosis therapy. Int J Nanomedicine. 2020;15:7967–7977. doi:10.2147/IJN.S263756
  • Glasson SS, Chambers MG, Berg WBVD, Little CB. The OARSI histopathology initiative – recommendations for histological assessments of osteoarthritis in the mouse. Osteoarthritis Cartilage. 2010;18:S17–S23. doi:10.1016/j.joca.2010.05.025
  • Paramitha D, Fakhrul Ulum M, Purnama A, Wicaksono D, Noviana D, Hermawan H. Monitoring degradation products and metal ions in vivo. In: Narayan R, editor. Monitoring and Evaluation of Biomaterials and Their Performance in vivo. Sawston: Woodhead Publishing; 2016:19–43. doi:10.1016/B978-0-08-100603-0.00002-X
  • Bergholt NL, Lysdahl H, Lind M, Foldager CB. A standardized method of applying toluidine blue metachromatic staining for assessment of chondrogenesis. Cartilage. 2019;10(3):370–374. doi:10.1177/1947603518764262
  • Henrotin Y, Sanchez C, Bay-Jensen AC, Mobasheri A. Osteoarthritis biomarkers derived from cartilage extracellular matrix: current status and future perspectives. Ann Phys Rehabil Med. 2016;59(3):145–148. doi:10.1016/j.rehab.2016.03.004
  • Roughley PJ, Mort JS. The role of aggrecan in normal and osteoarthritic cartilage. J Exp Orthop. 2014;1(1):8. doi:10.1186/s40634-014-0008-7
  • Hoshi H, Akagi R, Yamaguchi S, et al. Effect of inhibiting MMP13 and ADAMTS5 by intra-articular injection of small interfering RNA in a surgically induced osteoarthritis model of mice. Cell Tissue Res. 2017;368(2):379–387. doi:10.1007/s00441-016-2563-y
  • Hwang HS, Kim HA. Chondrocyte apoptosis in the pathogenesis of osteoarthritis. Int J Mol Sci. 2015;16(11):26035. doi:10.3390/ijms161125943
  • Liu H, Liang Z, Wang F, et al. Exosomes from mesenchymal stromal cells reduce murine colonic inflammation via a macrophage-dependent mechanism. JCI Insight. 2019;4(24):131273. doi:10.1172/jci.insight.131273
  • Li X, Liu L, Yang J, et al. Exosome derived from human umbilical cord mesenchymal stem cell mediates miR-181c attenuating burn-induced excessive inflammation. EBioMedicine. 2016;8:72–82. doi:10.1016/j.ebiom.2016.04.030
  • Zhang B, Yin Y, Lai RC, Tan SS, Choo ABH, Lim SK. Mesenchymal stem cells secrete immunologically active exosomes. Stem Cells Dev. 2014;23(11):1233–1244. doi:10.1089/scd.2013.0479
  • Lee C, Mitsialis SA, Aslam M, et al. Exosomes mediate the cytoprotective action of mesenchymal stromal cells on hypoxia-induced pulmonary hypertension. Circulation. 2012;126(22):2601–2611. doi:10.1161/CIRCULATIONAHA.112.114173
  • Li G, Yin J, Gao J, et al. Subchondral bone in osteoarthritis: insight into risk factors and microstructural changes. Arthritis Res Ther. 2013;15(6):223. doi:10.1186/ar4405
  • Hu W, Chen Y, Dou C, Dong S. Microenvironment in subchondral bone: predominant regulator for the treatment of osteoarthritis. Ann Rheum Dis. 2020;80(4):413–422. doi:10.1136/annrheumdis-2020-218089
  • Donell S. Subchondral bone remodelling in osteoarthritis. EFORT Open Rev. 2019;4(6):221–229. doi:10.1302/2058-5241.4.180102
  • Bellido M, Lugo L, Roman-Blas JA, et al. Improving subchondral bone integrity reduces progression of cartilage damage in experimental osteoarthritis preceded by osteoporosis. Osteoarthritis Cartilage. 2011;19(10):1228–1236. doi:10.1016/j.joca.2011.07.003
  • Hayami T, Pickarski M, Zhuo Y, Wesolowski GA, Rodan GA, Duong LT. Characterization of articular cartilage and subchondral bone changes in the rat anterior cruciate ligament transection and meniscectomized models of osteoarthritis. Bone. 2006;38(2):234–243. doi:10.1016/j.bone.2005.08.007
  • Hayami T, Pickarski M, Wesolowski GA, et al. The role of subchondral bone remodeling in osteoarthritis: reduction of cartilage degeneration and prevention of osteophyte formation by alendronate in the rat anterior cruciate ligament transection model. Arthritis Rheum. 2004;50(4):1193–1206. doi:10.1002/art.20124
  • Burr DB, Schaffler MB. The involvement of subchondral mineralized tissues in osteoarthrosis: quantitative microscopic evidence. Microsc Res Tech. 1997;37(4):343–357. doi:10.1002/(SICI)1097-0029(19970515)37:4<343::AID-JEMT9>3.0.CO;2-L
  • Lories RJ, Luyten FP. The bone-cartilage unit in osteoarthritis. Nat Rev Rheumatol. 2011;7(1):43–49. doi:10.1038/nrrheum.2010.197
  • Yuan XL, Meng HY, Wang YC, et al. Bone-cartilage interface crosstalk in osteoarthritis: potential pathways and future therapeutic strategies. Osteoarthritis Cartilage. 2014;22(8):1077–1089. doi:10.1016/j.joca.2014.05.023
  • Zhang W, Lian Q, Li D, et al. Cartilage repair and subchondral bone migration using 3d printing osteochondral composites: a one-year-period study in rabbit trochlea. Biomed Res Int. 2014;2014:e746138. doi:10.1155/2014/746138
  • Zhu S, Zhu J, Zhen G, et al. Subchondral bone osteoclasts induce sensory innervation and osteoarthritis pain. J Clin Invest. 2019;129(3):1076–1093. doi:10.1172/JCI121561
  • Zhu J, Zhen G, An S, et al. Aberrant subchondral osteoblastic metabolism modifies NaV1.8 for osteoarthritis. Elife. 2020;9. doi:10.7554/eLife.57656
  • Sousa M, Ferraresi C, Carolina A, Yoshimura E, Hamblin M. Building, testing and validating a set of home-made von Frey filaments: a precise, accurate and cost effective alternative for nociception assessment. J Neurosci Methods. 2014;232:1–5. doi:10.1016/j.jneumeth.2014.04.017
  • Deuis JR, Dvorakova LS, Vetter I. Methods used to evaluate pain behaviors in rodents. Front Mol Neurosci. 2017;10:284. doi:10.3389/fnmol.2017.00284
  • Hawker GA, Stewart L, French MR, et al. Understanding the pain experience in hip and knee osteoarthritis–an OARSI/OMERACT initiative. Osteoarthritis Cartilage. 2008;16(4):415–422. doi:10.1016/j.joca.2007.12.017
  • Duarte RV, Raphael JH, Dimitroulas T, et al. Osteoarthritis pain has a significant neuropathic component: an exploratory in vivo patient model. Rheumatol Int. 2014;34(3):315–320. doi:10.1007/s00296-013-2893-y
  • Valdes AM, Suokas AK, Doherty SA, Jenkins W, Doherty M. History of knee surgery is associated with higher prevalence of neuropathic pain-like symptoms in patients with severe osteoarthritis of the knee. Semin Arthritis Rheum. 2014;43(5):588–592. doi:10.1016/j.semarthrit.2013.10.001
  • Lakes EH, Allen KD. Gait analysis methods for rodent models of arthritic disorders: reviews and recommendations. Osteoarthritis Cartilage. 2016;24(11):1837–1849. doi:10.1016/j.joca.2016.03.008
  • Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478):eaau6977. doi:10.1126/science.aau6977
  • Gomzikova MO, James V, Rizvanov AA. Therapeutic application of mesenchymal stem cells derived extracellular vesicles for immunomodulation. Front Immunol. 2019;10:2663. doi:10.3389/fimmu.2019.02663
  • Elahi FM, Farwell DG, Nolta JA, Anderson JD. Preclinical translation of exosomes derived from mesenchymal stem/stromal cells. Stem Cells. 2020;38(1):15–21. doi:10.1002/stem.3061
  • Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat Rev Immunol. 2014;14(3):195–208. doi:10.1038/nri3622
  • Toma C, Wagner WR, Bowry S, Schwartz A, Villanueva F. Fate of culture-expanded mesenchymal stem cells in the microvasculature: in vivo observations of cell kinetics. Circ Res. 2009;104(3):398–402. doi:10.1161/CIRCRESAHA.108.187724
  • Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators. J Cell Biochem. 2006;98(5):1076–1084. doi:10.1002/jcb.20886
  • Beltrami AP, Cesselli D, Beltrami CA. Stem cell senescence and regenerative paradigms. Clin Pharmacol Ther. 2012;91(1):21–29. doi:10.1038/clpt.2011.262
  • Sverdlov ED, Mineev K. Mutation rate in stem cells: an underestimated barrier on the way to therapy. Trends Mol Med. 2013;19(5):273–280. doi:10.1016/j.molmed.2013.01.004
  • Lee AS, Tang C, Rao MS, Weissman IL, Wu JC. Tumorigenicity as a clinical hurdle for pluripotent stem cell therapies. Nat Med. 2013;19(8):998–1004. doi:10.1038/nm.3267
  • Vizoso FJ, Eiro N, Cid S, Schneider J, Perez-Fernandez R. mesenchymal stem cell secretome: toward cell-free therapeutic strategies in regenerative medicine. Int J Mol Sci. 2017;18(9):1852. doi:10.3390/ijms18091852
  • Naseri Z, Oskuee RK, Jaafari MR, Forouzandeh Moghadam M. Exosome-mediated delivery of functionally active miRNA-142-3p inhibitor reduces tumorigenicity of breast cancer in vitro and in vivo. Int J Nanomedicine. 2018;13:7727–7747. doi:10.2147/IJN.S182384
  • Huyan T, Li H, Peng H, et al. Extracellular vesicles - advanced nanocarriers in cancer therapy: progress and achievements. Int J Nanomedicine. 2020;15:6485–6502. doi:10.2147/IJN.S238099
  • Imai T, Takahashi Y, Nishikawa M, et al. Macrophage-dependent clearance of systemically administered B16BL6-derived exosomes from the blood circulation in mice. J Extracell Vesicles. 2015;4(1):26238. doi:10.3402/jev.v4.26238
  • Lai CP, Mardini O, Ericsson M, et al. Dynamic biodistribution of extracellular vesicles in vivo using a multimodal imaging reporter. ACS Nano. 2014;8(1):483–494. doi:10.1021/nn404945r
  • Cheng J, Abdi S. Complications of joint, tendon, and muscle injections. Tech Reg Anesth Pain Manag. 2007;11(3):141–147. doi:10.1053/j.trap.2007.05.006
  • Witwer KW, Théry C. Extracellular vesicles or exosomes? On primacy, precision, and popularity influencing a choice of nomenclature. J Extracell Vesicles. 2019;8(1):1648167. doi:10.1080/20013078.2019.1648167
  • Haraszti RA, Miller R, Stoppato M, et al. Exosomes produced from 3D cultures of MSCs by tangential flow filtration show higher yield and improved activity. Mol Ther. 2018;26(12):2838–2847. doi:10.1016/j.ymthe.2018.09.015
  • Pachler K, Lener T, Streif D, et al. A good manufacturing practice-grade standard protocol for exclusively human mesenchymal stromal cell-derived extracellular vesicles. Cytotherapy. 2017;19(4):458–472. doi:10.1016/j.jcyt.2017.01.001
  • Chen T, Arslan F, Yin Y, et al. Enabling a robust scalable manufacturing process for therapeutic exosomes through oncogenic immortalization of human ESC-derived MSCs. J Transl Med. 2011;9(1):47. doi:10.1186/1479-5876-9-47
  • Lian Q, Lye E, Suan Yeo K, et al. Derivation of clinically compliant MSCs from CD105+, CD24- differentiated human ESCs. Stem Cells. 2007;25(2):425–436. doi:10.1634/stemcells.2006-0420
  • Herrmann M, Diederichs S, Melnik S, et al. Extracellular vesicles in musculoskeletal pathologies and regeneration. Front Bioeng Biotechnol. 2021;8:624096. doi:10.3389/fbioe.2020.624096
  • Sabapathy V, Kumar S. hiPSC‐derived iMSCs: NextGen MSCs as an advanced therapeutically active cell resource for regenerative medicine. J Cell Mol Med. 2016;20(8):1571–1588. doi:10.1111/jcmm.12839