Mesenchymal Stem Cell-Derived Extracellular Vesicles in Tendon and Ligament Repair—A Systematic Review of In Vivo Studies
Abstract
:1. Introduction
2. Methodology
- Isolation and characterisation of MSCs, including source of MSCs, cellular origin, cell treatment to extract MSCs, and procedures to verify MSCs (e.g., flow cytometry, western blotting).
- Characterisation and purification of EVs, including MSC purification to extract EVs, EV dimensions, EV biomarkers, imaging used to visualise EVs, and EV active component.
- In vivo model, including method of EV delivery, type of in vivo model, how tendon/ligament injury was induced, animal age, animal weight, animal gender, total number of animals used per experimental group, and follow-up time.
- In vivo findings, including macroscopic appearance, imaging results, histopathological results, biochemical findings, and biomechanical findings.
3. Results
3.1. Characterisation of MSCs
3.2. Characterisation of EVs
3.3. Animal Models
3.4. In Vivo Findings
3.5. Risk of Bias
4. Discussion
4.1. MSC Isolation, Differentiation, and Culture Media
4.2. EV Isolation and Administration
4.3. Modifying EVs to Enhance their Biological Function
Role of MMP-14 and miR-21 in Tendon/Ligament Repair
4.4. EV Educated Macrophages
4.5. Animal Models of Tendon/Ligament Injury
4.6. In Vivo Findings
4.7. Limitations
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Wu, F.; Nerlich, M.; Docheva, D. Tendon injuries: Basic science and new repair proposals. EFORT Open Rev. 2017, 2, 332–342. [Google Scholar] [CrossRef]
- Parmar, K. Tendon and ligament: Basic science, injury and repair. Orthop. Trauma 2018, 32, 241–244. [Google Scholar] [CrossRef]
- Andarawis-Puri, N.; Flatow, E.L.; Soslowsky, L.J. Tendon basic science: Development, repair, regeneration, and healing. J. Orthop. Res. 2015, 33, 780–784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yuan, T.; Zhang, C.Q.; Wang, J.H. Augmenting tendon and ligament repair with platelet-rich plasma (PRP). Muscles Ligaments Tendons J. 2013, 3, 139–149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arner, J.W.; Johannsen, A.M.; Ruzbarsky, J.J.; Godin, J.A. Open Popliteal Tendon Repair. Arthrosc. Tech. 2021, 10, e499–e505. [Google Scholar] [CrossRef]
- Kainth, G.; Goel, A. A useful technique of using anterior cruciate ligament reconstruction jig for preparing patellar tunnel in surgical repair of extensor tendon ruptures. Ann. R. Coll. Surg. Engl. 2021, 103, 142–143. [Google Scholar] [CrossRef]
- Villalba, J.; Molina-Corbacho, M.; García, R.; Martínez-Carreres, L. Home-Based Intravenous Analgesia with an Elastomeric Pump after Medial Patellofemoral Ligament Repair: A Case Series. J. PeriAnesthesia Nurs. 2021. [Google Scholar] [CrossRef]
- Linderman, S.W.; Gelberman, R.H.; Thomopoulos, S.; Shen, H. Cell and Biologic-Based Treatment of Flexor Tendon Injuries. Oper. Tech. Orthop. 2016, 26, 206–215. [Google Scholar] [CrossRef] [Green Version]
- Thomopoulos, S.; Parks, W.C.; Rifkin, D.B.; Derwin, K.A. Mechanisms of tendon injury and repair. J. Orthop. Res. 2015, 33, 832–839. [Google Scholar] [CrossRef] [Green Version]
- Carlson, J.; Fox, O.; Kilby, P. Massive Chondrolysis and Joint Destruction after Artificial Anterior Cruciate Ligament Repair. Case Rep. Orthop. 2021, 2021, 1–5. [Google Scholar] [CrossRef]
- Lim, W.L.; Liau, L.L.; Ng, M.H.; Chowdhury, S.R.; Law, J.X. Current Progress in Tendon and Ligament Tissue Engineering. Tissue Eng. Regen. Med. 2019, 16, 549–571. [Google Scholar] [CrossRef]
- Ramdass, B. Ligament and Tendon Repair through Regeneration Using Mesenchymal Stem Cells. Curr. Stem Cell Res. Ther. 2014, 10, 84–88. [Google Scholar] [CrossRef] [Green Version]
- Blum, B.; Bar-Nur, O.; Golan-Lev, T.; Benvenisty, N. The anti-apoptotic gene survivin contributes to teratoma formation by human embryonic stem cells. Nat. Biotechnol. 2009, 27, 281–287. [Google Scholar] [CrossRef] [PubMed]
- Harris, M.T.; Butler, D.L.; Boivin, G.P.; Florer, J.B.; Schantz, E.J.; Wenstrup, R.J. Mesenchymal stem cells used for rabbit tendon repair can form ectopic bone and express alkaline phosphatase activity in constructs. J. Orthop. Res. 2004, 22, 998–1003. [Google Scholar] [CrossRef] [PubMed]
- Law, J.X.; Liau, L.L.; Bin Saim, A.; Yang, Y.; Idrus, R. Electrospun Collagen Nanofibers and Their Applications in Skin Tissue Engineering. Tissue Eng. Regen. Med. 2017, 14, 699–718. [Google Scholar] [CrossRef] [PubMed]
- Bobis-Wozowicz, S.; Kmiotek, K.; Sekula, M.; Kedracka-Krok, S.; Kamycka, E.; Adamiak, M.; Jankowska, U.; Madetko-Talowska, A.; Sarna, M.; Bik-Multanowski, M.; et al. Human Induced Pluripotent Stem Cell-Derived Microvesicles Transmit RNAs and Proteins to Recipient Mature Heart Cells Modulating Cell Fate and Behavior. Stem Cells 2015, 33, 2748–2761. [Google Scholar] [CrossRef] [PubMed]
- Bernardo, M.E.; Fibbe, W.E. Mesenchymal Stromal Cells: Sensors and Switchers of Inflammation. Cell Stem Cell 2013, 13, 392–402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jia, Y.-J.; Zhou, Y.; Wen, L.-L.; Li, Y.-F.; Wu, K.-M.; Duan, R.-R.; Yao, Y.-B.; Jing, L.-J.; Gong, Z.; Teng, J.-F. Exosomes derived from bone marrow mesenchymal stem cells protect the injured spinal cord by inhibiting pericyte pyroptosis. Neural Regen. Res. 2022, 17, 194–202, in press. [Google Scholar] [CrossRef]
- Liao, Q.; Li, B.J.; Li, Y.; Xiao, Y.; Zeng, H.; Liu, J.M.; Yuan, L.X.; Liu, G. Low-intensity pulsed ultrasound promotes osteoarthritic cartilage regeneration by BMSC-derived exosomes via modulating the NF-κB signaling pathway. Int. Immunopharmacol. 2021, 97, 107824. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Tang, Y.; Liu, Z.; Yin, Y.; Li, Q.; Liu, G.; Yan, B. The Application Potential and Advance of Mesenchymal Stem Cell-Derived Exosomes in Myocardial Infarction. Stem Cells Int. 2021, 2021, 1–15. [Google Scholar] [CrossRef]
- Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. J. Clin. Epidemiol. 2021, 134, 178–189. [Google Scholar] [CrossRef]
- Witwer, K.W.; Van Balkom, B.W.M.; Bruno, S.; Choo, A.; Dominici, M.; Gimona, M.; Hill, A.F.; De Kleijn, D.; Koh, M.; Lai, R.C.; et al. Defining mesenchymal stromal cell (MSC)-derived small extracellular vesicles for therapeutic applications. J. Extracell. Vesicles 2019, 8, 1609206. [Google Scholar] [CrossRef] [Green Version]
- Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; 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, 1535750. [Google Scholar] [CrossRef] [Green Version]
- Hooijmans, C.R.; Rovers, M.M.; de Vries, R.B.; Leenaars, M.; Ritskes-Hoitinga, M.; Langendam, M.W. SYRCLE’s risk of bias tool for animal studies. BMC Med. Res. Methodol. 2014, 14, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shi, Z.; Wang, Q.; Jiang, D. Extracellular vesicles from bone marrow-derived multipotent mesenchymal stromal cells regulate inflammation and enhance tendon healing. J. Transl. Med. 2019, 17, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shen, H.; Yoneda, S.; Abu-Amer, Y.; Guilak, F.; Gelberman, R.H. Stem cell-derived extracellular vesicles attenuate the early inflammatory response after tendon injury and repair. J. Orthop. Res. 2020, 38, 117–127. [Google Scholar] [CrossRef]
- Yu, H.; Cheng, J.; Shi, W.; Ren, B.; Zhao, F.; Shi, Y.; Yang, P.; Duan, X.; Zhang, J.; Fu, X.; et al. Bone marrow mesenchymal stem cell-derived exosomes promote tendon regeneration by facilitating the proliferation and migration of endogenous tendon stem/progenitor cells. Acta Biomater. 2020, 106, 328–341. [Google Scholar] [CrossRef]
- Shi, Y.; Kang, X.; Wang, Y.; Bian, X.; He, G.; Zhou, M.; Tang, K. Exosomes Derived from Bone Marrow Stromal Cells (BMSCs) Enhance Tendon-Bone Healing by Regulating Macrophage Polarization. Med. Sci. Monit. 2020, 26. [Google Scholar] [CrossRef]
- Gissi, C.; Radeghieri, A.; Passeri, C.A.L.; Gallorini, M.; Calciano, L.; Oliva, F.; Veronesi, F.; Zendrini, A.; Cataldi, A.; Bergese, P.; et al. Extracellular vesicles from rat-bone-marrow mesenchymal stromal/stem cells improve tendon repair in rat Achilles tendon injury model in dose-dependent manner: A pilot study. PLoS ONE 2020, 15, e0229914. [Google Scholar] [CrossRef]
- Yao, Z.; Li, J.; Wang, X.; Peng, S.; Ning, J.; Qian, Y.; Fan, C. MicroRNA-21-3p Engineered Umbilical Cord Stem Cell-Derived Exosomes Inhibit Tendon Adhesion. J. Inflamm. Res. 2020, 13, 303–316. [Google Scholar] [CrossRef]
- Chamberlain, C.S.; Kink, J.A.; Wildenauer, L.A.; McCaughey, M.; Henry, K.; Spiker, A.M.; Halanski, M.A.; Hematti, P.; Vanderby, R. Exosome-educated macrophages and exosomes differentially improve ligament healing. Stem Cells 2020, 39, 55–61. [Google Scholar]
- Huang, Y.; He, B.; Wang, L.; Yuan, B.; Shu, H.; Zhang, F.; Sun, L. Bone marrow mesenchymal stem cell-derived exosomes promote rotator cuff tendon-bone healing by promoting angiogenesis and regulating M1 macrophages in rats. Stem Cell Res. Ther. 2020, 11, 1–16. [Google Scholar] [CrossRef]
- Li, J.; Yao, Z.; Xiong, H.; Cui, H.; Wang, X.; Zheng, W.; Qian, Y.; Fan, C. Extracellular vesicles from hydroxycamptothecin primed umbilical cord stem cells enhance anti-adhesion potential for treatment of tendon injury. Stem Cell Res. Ther. 2020, 11, 1–14. [Google Scholar] [CrossRef]
- Wang, Y.; He, G.; Guo, Y.; Tang, H.; Shi, Y.; Bian, X.; Zhu, M.; Kang, X.; Zhou, M.; Lyu, J.; et al. Exosomes from tendon stem cells promote injury tendon healing through balancing synthesis and degradation of the tendon extracellular matrix. J. Cell. Mol. Med. 2019, 23, 5475–5485. [Google Scholar] [CrossRef] [Green Version]
- Wang, C.; Hu, Q.; Song, W.; Yu, W.; He, Y. Adipose Stem Cell–Derived Exosomes Decrease Fatty Infiltration and Enhance Rotator Cuff Healing in a Rabbit Model of Chronic Tears. Am. J. Sports Med. 2020, 48, 1456–1464. [Google Scholar] [CrossRef]
- Song, Y.; Dou, H.; Li, X.; Zhao, X.; Li, Y.; Liu, D.; Ji, J.; Liu, F.; Ding, L.; Ni, Y.; et al. Exosomal miR-146a Contributes to the Enhanced Therapeutic Efficacy of Interleukin-1β-Primed Mesenchymal Stem Cells against Sepsis. Stem Cells 2017, 35, 1208–1221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Markides, H.; Foster, N.; McLaren, J.; Hopkins, T.; Black, C.; Oreffo, R.; Scammell, B.; Echevarria, I.; White, L.; El Haj, A. Short-Term Evaluation of Cellular Fate in an Ovine Bone Formation Model. Cells 2021, 10, 1776. [Google Scholar] [CrossRef] [PubMed]
- Futrega, K.; Music, E.; Robey, P.; Gronthos, S.; Crawford, R.; Saifzadeh, S.; Klein, T.J.; Doran, M.R. Characterisation of ovine bone marrow-derived stromal cells (oBMSC) and evaluation of chondrogenically induced micro-pellets for cartilage tissue repair in vivo. Stem Cell Res. Ther. 2021, 12, 1–19. [Google Scholar] [CrossRef] [PubMed]
- Nordin, J.Z.; Lee, Y.; Vader, P.; Mäger, I.; Johansson, H.J.; Heusermann, W.; Wiklander, O.P.; Hällbrink, M.; Seow, Y.; Bultema, J.J.; et al. Ultrafiltration with size-exclusion liquid chromatography for high yield isolation of extracellular vesicles preserving intact biophysical and functional properties. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 879–883. [Google Scholar] [CrossRef] [Green Version]
- Benedikter, B.J.; Bouwman, F.G.; Vajen, T.; Heinzmann, A.C.A.; Grauls, G.; Mariman, E.C.; Wouters, E.F.M.; Savelkoul, P.H.; Lopez-Iglesias, C.; Koenen, R.R.; et al. Ultrafiltration combined with size exclusion chromatography efficiently isolates extracellular vesicles from cell culture media for compositional and functional studies. Sci. Rep. 2017, 7, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Mol, E.A.; Goumans, M.-J.; Doevendans, P.A.; Sluijter, J.P.G.; Vader, P. Higher functionality of extracellular vesicles isolated using size-exclusion chromatography compared to ultracentrifugation. Nanomedicine 2017, 13, 2061–2065. [Google Scholar] [CrossRef] [PubMed]
- Monguió-Tortajada, M.; Gálvez-Montón, C.; Bayes-Genis, A.; Roura, S.; Borràs, F.E. Extracellular vesicle isolation methods: Rising impact of size-exclusion chromatography. Cell. Mol. Life Sci. 2019, 76, 2369–2382. [Google Scholar] [CrossRef]
- Spohn, G.; Witte, A.-S.; Kretschmer, A.; Seifried, E.; Schäfer, R. More Human BM-MSC With Similar Subpopulation Composition and Functional Characteristics Can Be Produced With a GMP-Compatible Fabric Filter System Compared to Density Gradient Technique. Front. Cell Dev. Biol. 2021, 9. [Google Scholar] [CrossRef]
- Hagmann, S.; Moradi, B.; Frank, S.; Dreher, T.; Kämmerer, P.W.; Richter, W.; Gotterbarm, T. Different culture media affect growth characteristics, surface marker distribution and chondrogenic differentiation of human bone marrow-derived mesenchymal stromal cells. BMC Musculoskelet. Disord. 2013, 14, 223. [Google Scholar] [CrossRef] [Green Version]
- Sotiropoulou, P.A.; Perez, S.A.; Salagianni, M.; Baxevanis, C.N.; Papamichail, M. Characterization of the Optimal Culture Conditions for Clinical Scale Production of Human Mesenchymal Stem Cells. Stem Cells 2006, 24, 462–471. [Google Scholar] [CrossRef] [Green Version]
- Dexheimer, V.; Mueller, S.; Braatz, F.; Richter, W. Reduced Reactivation from Dormancy but Maintained Lineage Choice of Human Mesenchymal Stem Cells with Donor Age. PLoS ONE 2011, 6, e22980. [Google Scholar] [CrossRef]
- Schop, D.; Janssen, F.W.; Van Rijn, L.D.S.; Fernandes, H.; Bloem, R.M.; De Bruijn, J.D.; Van Dijkhuizen-Radersma, R. Growth, Metabolism, and Growth Inhibitors of Mesenchymal Stem Cells. Tissue Eng. Part A 2009, 15, 1877–1886. [Google Scholar] [CrossRef] [PubMed]
- Scuteri, A.; Donzelli, E.; Rodriguez-Menendez, V.; Ravasi, M.; Monfrini, M.; Bonandrini, B.; Figliuzzi, M.; Remuzzi, A.; Tredici, G. A Double Mechanism for the Mesenchymal Stem Cells’ Positive Effect on Pancreatic Islets. PLoS ONE 2014, 9, e84309. [Google Scholar] [CrossRef] [Green Version]
- Martínez-Lorenzo, M.J.; Cañas, M.R.; Alegre-Aguarón, E.; Desportes, P.; Castiella, T.; García-Álvarez, F.; Larrad, L. Phenotype and chondrogenic differentiation of mesenchymal cells from adipose tissue of different species. J. Orthop. Res. 2009, 27, 1499–1507. [Google Scholar] [CrossRef] [PubMed]
- Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef] [PubMed]
- Nagano, M.; Kimura, K.; Yamashita, T.; Ohneda, K.; Nozawa, D.; Hamada, H.; Yoshikawa, H.; Ochiai, N.; Ohneda, O. Hypoxia Responsive Mesenchymal Stem Cells Derived from Human Umbilical Cord Blood Are Effective for Bone Repair. Stem Cells Dev. 2010, 19, 1195–1210. [Google Scholar] [CrossRef]
- Coipeau, P.; Rosset, P.; Langonné, A.; Gaillard, J.; Delorme, B.; Rico, A.; Domenech, J.; Charbord, P.; Sensebé, L. Impaired differentiation potential of human trabecular bone mesenchymal stromal cells from elderly patients. Cytotherapy 2009, 11, 584–594. [Google Scholar] [CrossRef] [PubMed]
- Smith, R.P.; Eckalbar, W.L.; Morrissey, K.M.; Luizon, M.; Hoffmann, T.J.; Sun, X.; Jones, S.L.; Aldred, S.F.; Ramamoorthy, A.; Desta, Z.; et al. Genome-Wide Discovery of Drug-Dependent Human Liver Regulatory Elements. PLoS Genet. 2014, 10, e1004648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ryu, A.H.; Eckalbar, W.L.; Kreimer, A.; Yosef, N.; Ahituv, N. Use antibiotics in cell culture with caution: Genome-wide identification of antibiotic-induced changes in gene expression and regulation. Sci. Rep. 2017, 7, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Dessels, C.; Potgieter, M.; Pepper, M.S. Making the Switch: Alternatives to Fetal Bovine Serum for Adipose-Derived Stromal Cell Expansion. Front. Cell Dev. Biol. 2016, 4, 115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mesalam, A.; Lee, K.-L.; Khan, I.; Chowdhury, M.M.R.; Zhang, S.; Song, S.-H.; Joo, M.-D.; Lee, J.-H.; Jin, J.-I.; Kong, I.-K. A combination of bovine serum albumin with insulin–transferrin–sodium selenite and/or epidermal growth factor as alternatives to fetal bovine serum in culture medium improves bovine embryo quality and trophoblast invasion by induction of matrix metalloproteinases. Reprod. Fertil. Dev. 2019, 31, 333–346. [Google Scholar] [CrossRef] [PubMed]
- Lindroos, B.; Boucher, S.; Chase, L.; Kuokkanen, H.; Huhtala, H.; Haataja, R.; Vemuri, M.; Suuronen, R.; Miettinen, S. Serum-free, xeno-free culture media maintain the proliferation rate and multipotentiality of adipose stem cells in vitro. Cytotherapy 2009, 11, 958–972. [Google Scholar] [CrossRef]
- Shahdadfar, A.; Frønsdal, K.; Haug, T.; Reinholt, F.P.; Brinchmann, J.E. In Vitro Expansion of Human Mesenchymal Stem Cells: Choice of Serum Is a Determinant of Cell Proliferation, Differentiation, Gene Expression, and Transcriptome Stability. Stem Cells 2005, 23, 1357–1366. [Google Scholar] [CrossRef]
- Brennan, K.; Martin, K.; Fitzgerald, S.P.; O’Sullivan, J.; Wu, Y.; Blanco, A.; Richardson, C.; Mc Gee, M.M. A comparison of methods for the isolation and separation of extracellular vesicles from protein and lipid particles in human serum. Sci. Rep. 2020, 10, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Takov, K.; Yellon, D.M.; Davidson, S.M. Comparison of small extracellular vesicles isolated from plasma by ultracentrifugation or size-exclusion chromatography: Yield, purity and functional potential. J. Extracell. Vesicles 2019, 8, 1560809. [Google Scholar] [CrossRef]
- Nanoparticle Tracking Analysis vs Tunable Resistive Pulse Sensing. Available online: https://www.izon.com/trps/compare-nta-and-trps (accessed on 10 July 2021).
- Otahal, A.; Kuten-Pella, O.; Kramer, K.; Neubauer, M.; Lacza, Z.; Nehrer, S.; De Luna, A. Functional repertoire of EV-associated miRNA profiles after lipoprotein depletion via ultracentrifugation and size exclusion chromatography from autologous blood products. Sci. Rep. 2021, 11, 1–16. [Google Scholar] [CrossRef]
- Hashimoto, K.; Akagi, M. The role of oxidation of low-density lipids in pathogenesis of osteoarthritis: A narrative review. J. Int. Med. Res. 2020, 48. [Google Scholar] [CrossRef] [PubMed]
- Vickers, K.C.; Palmisano, B.T.; Shoucri, B.M.; Shamburek, R.D.; Remaley, A.T. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat. Cell Biol. 2011, 13, 423–433. [Google Scholar] [CrossRef] [Green Version]
- Phinney, D.G.; Pittenger, M.F. Concise Review: MSC-Derived Exosomes for Cell-Free Therapy. Stem Cells 2017, 35, 851–858. [Google Scholar] [CrossRef] [Green Version]
- Spicer, P.P.; Mikos, A.G. Fibrin glue as a drug delivery system. J. Control. Release 2010, 148, 49–55. [Google Scholar] [CrossRef] [Green Version]
- Lamichhane, T.N.; Jay, S.M. Production of Extracellular Vesicles Loaded with Therapeutic Cargo. Target. Drug Deliv. 2018, 1831, 37–47. [Google Scholar] [CrossRef]
- Chen, S.; Tang, Y.; Liu, Y.; Zhang, P.; Lv, L.; Zhang, X.; Jia, L.; Zhou, Y. Exosomes derived from miR-375-overexpressing human adipose mesenchymal stem cells promote bone regeneration. Cell Prolif. 2019, 52, e12669. [Google Scholar] [CrossRef] [Green Version]
- Domenis, R.; Cifù, A.; Quaglia, S.; Pistis, C.; Moretti, M.; Vicario, A.; Parodi, P.C.; Fabris, M.; Niazi, K.R.; Soon-Shiong, P.; et al. Pro inflammatory stimuli enhance the immunosuppressive functions of adipose mesenchymal stem cells-derived exosomes. Sci. Rep. 2018, 8, 1–11. [Google Scholar] [CrossRef]
- Harting, M.T.; Srivastava, A.; Zhaorigetu, S.; Bair, H.; Prabhakara, K.S.; Furman, N.E.T.; Vykoukal, J.V.; Ruppert, K.A.; Cox, C.S.; Olson, S.D. Inflammation-Stimulated Mesenchymal Stromal Cell-Derived Extracellular Vesicles Attenuate Inflammation. Stem Cells 2018, 36, 79–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zayed, M.; Adair, S.; Dhar, M. Effects of Normal Synovial Fluid and Interferon Gamma on Chondrogenic Capability and Immunomodulatory Potential Respectively on Equine Mesenchymal Stem Cells. Int. J. Mol. Sci. 2021, 22, 6391. [Google Scholar] [CrossRef] [PubMed]
- Gulotta, L.V.; Kovacevic, D.; Montgomery, S.; Ehteshami, J.R.; Packer, J.D.; Rodeo, S.A. Stem Cells Genetically Modified With the Developmental Gene MT1-MMP Improve Regeneration of the Supraspinatus Tendon-to-Bone Insertion Site. Am. J. Sports Med. 2010, 38, 1429–1437. [Google Scholar] [CrossRef]
- Oshiro, W.; Lou, J.; Xing, X.; Tu, Y.; Manske, P.R. Flexor tendon healing in the rat: A histologic and gene expression study. J. Hand Surg. 2003, 28, 814–823. [Google Scholar] [CrossRef]
- Apte, S.S.; Fukai, N.; Beier, D.R.; Olsen, B.R. The Matrix Metalloproteinase-14 (MMP-14) Gene Is Structurally Distinct from Other MMP Genes and Is Co-expressed with the TIMP-2 Gene during Mouse Embryogenesis. J. Biol. Chem. 1997, 272, 25511–25517. [Google Scholar] [CrossRef] [Green Version]
- Kinoh, H.; Sato, H.; Tsunezuka, Y.; Takino, T.; Kawashima, A.; Okada, Y.; Seiki, M. MT-MMP, the cell surface activator of proMMP-2 (pro-gelatinase A), is expressed with its substrate in mouse tissue during embryogenesis. J. Cell Sci. 1996, 109, 953–959. [Google Scholar] [CrossRef]
- Mias, C.; Lairez, O.; Trouche, E.; Roncalli, J.; Calise, D.; Seguelas, M.H.; Ordener, C.; Piercecchi-Marti, M.D.; Auge, N.; Salvayre, A.N.; et al. Mesenchymal stem cells promote matrix metalloproteinase secretion by cardiac fibroblasts and reduce cardiac ventricular fibrosis after myocardial infarction. Stem Cells 2009, 27, 2734–2743. [Google Scholar] [CrossRef]
- Zigrino, P.; Brinckmann, J.; Niehoff, A.; Lu, Y.; Giebeler, N.; Eckes, B.; Kadler, K.E.; Mauch, C. Fibroblast-Derived MMP-14 Regulates Collagen Homeostasis in Adult Skin. J. Investig. Dermatol. 2016, 136, 1575–1583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Annabi, B.; Laflamme, C.; Sina, A.; Lachambre, M.-P.; Béliveau, R. A MT1-MMP/NF-κB signaling axis as a checkpoint controller of COX-2 expression in CD133(+) U87 glioblastoma cells. J. Neuroinflamm. 2009, 6, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cohen, D.B.; Kawamura, S.; Ehteshami, J.R.; Rodeo, S.A. Indomethacin and Celecoxib Impair Rotator Cuff Tendon-to-Bone Healing. Am. J. Sports Med. 2006, 34, 362–369. [Google Scholar] [CrossRef] [PubMed]
- Mori, M.A.; Ludwig, R.G.; Garcia-Martin, R.; Brandão, B.B.; Kahn, C.R. Extracellular miRNAs: From Biomarkers to Mediators of Physiology and Disease. Cell Metab. 2019, 30, 656–673. [Google Scholar] [CrossRef] [PubMed]
- Wright, R.W.; Allen, T.; El-Zawawy, H.B.; Brodt, M.D.; Silva, M.J.; Gill, C.S.; Sandell, L.J. Medial collateral ligament healing in macrophage metalloelastase (MMP-12)-deficient mice. J. Orthop. Res. 2006, 24, 2106–2113. [Google Scholar] [CrossRef]
- Chamberlain, C.S.; Leiferman, E.M.; Frisch, K.E.; Wang, S.; Yang, X.; Van Rooijen, N.; Baer, G.S.; Brickson, S.L.; Vanderby, R. The influence of macrophage depletion on ligament healing. Connect. Tissue Res. 2010, 52, 203–211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de la Durantaye, M.; Piette, A.B.; van Rooijen, N.; Frenette, J. Macrophage depletion reduces cell proliferation and extracellular matrix accumulation but increases the ultimate tensile strength of injured Achilles tendons. J. Orthop. Res. 2014, 32, 279–285. [Google Scholar] [CrossRef] [PubMed]
- Schlundt, C.; El Khassawna, T.; Serra, A.; Dienelt, A.; Wendler, S.; Schell, H.; van Rooijen, N.; Radbruch, A.; Lucius, R.; Hartmann, S.; et al. Macrophages in bone fracture healing: Their essential role in endochondral ossification. Bone 2018, 106, 78–89. [Google Scholar] [CrossRef]
- Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage Activation and Polarization: Nomenclature and Experimental Guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef] [Green Version]
- Zhang, H.; Lin, C.; Zeng, C.; Wang, Z.; Wang, H.; Lu, J.; Liu, X.; Shao, Y.; Zhao, C.; Pan, J.; et al. Synovial macrophage M1 polarisation exacerbates experimental osteoarthritis partially through R-spondin-2. Ann. Rheum. Dis. 2018, 77, 1524–1534. [Google Scholar] [CrossRef]
- Chen, S.; Deng, G.; Li, K.; Zheng, H.; Wang, G.; Yu, B.; Zhang, K. Interleukin-6 Promotes Proliferation but Inhibits Tenogenic Differentiation via the Janus Kinase/Signal Transducers and Activators of Transcription 3 (JAK/STAT3) Pathway in Tendon-Derived Stem Cells. Med. Sci. Monit. 2018, 24, 1567–1573. [Google Scholar] [CrossRef] [Green Version]
- Manning, C.N.; Havlioglu, N.; Knutsen, E.; Sakiyama-Elbert, S.E.; Silva, M.; Thomopoulos, S.; Gelberman, R.H. The early inflammatory response after flexor tendon healing: A gene expression and histological analysis. J. Orthop. Res. 2014, 32, 645–652. [Google Scholar] [CrossRef]
- Dakin, S.G.; Werling, D.; Hibbert, A.; Abayasekara, D.R.E.; Young, N.J.; Smith, R.K.W.; Dudhia, J. Macrophage Sub-Populations and the Lipoxin A4 Receptor Implicate Active Inflammation during Equine Tendon Repair. PLoS ONE 2012, 7, e32333. [Google Scholar] [CrossRef] [Green Version]
- Xu, T.; Xu, M.; Bai, J.; Lin, J.; Yu, B.; Liu, Y.; Guo, X.; Shen, J.; Sun, H.; Hao, Y.; et al. Tenocyte-derived exosomes induce the tenogenic differentiation of mesenchymal stem cells through TGF-β. Cytotechnology 2019, 71, 57–65. [Google Scholar] [CrossRef] [PubMed]
- Wall, R.; Shani, M. Are animal models as good as we think? Theriogenology 2008, 69, 2–9. [Google Scholar] [CrossRef] [Green Version]
- Laudanski, K.; Stentz, M.; DiMeglio, M.; Furey, W.; Steinberg, T.; Patel, A. Potential Pitfalls of the Humanized Mice in Modeling Sepsis. Int. J. Inflamm. 2018, 2018, 1–9. [Google Scholar] [CrossRef]
- Soslowsky, L.J.; Carpenter, J.E.; DeBano, C.M.; Banerji, I.; Moalli, M.R. Development and use of an animal model for investigations on rotator cuff disease. J. Shoulder Elb. Surg. 1996, 5, 383–392. [Google Scholar] [CrossRef]
- Derwin, K.A.; Baker, A.R.; Iannotti, J.P.; McCarron, J.A. Preclinical Models for Translating Regenerative Medicine Therapies for Rotator Cuff Repair. Tissue Eng. Part B Rev. 2010, 16, 21–30. [Google Scholar] [CrossRef] [Green Version]
- Edelstein, L.; Thomas, S.J.; Soslowsky, L.J. Rotator cuff tears: What have we learned from animal models? J. Musculoskelet. Neuronal Interact. 2011, 11, 150–162. [Google Scholar]
- Poppen, N.K.; Walker, P.S. Forces at the Glenohumeral Joint in Abduction. Clin. Orthop. Relat. Res. 1978, 135, 165–170. [Google Scholar] [CrossRef]
- Perry, S.M.; Getz, C.L.; Soslowsky, L.J. Alterations in function after rotator cuff tears in an animal model. J. Shoulder Elb. Surg. 2009, 18, 296–304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sarver, J.J.; Dishowitz, M.I.; Kim, S.-Y.; Soslowsky, L.J. Transient decreases in forelimb gait and ground reaction forces following rotator cuff injury and repair in a rat model. J. Biomech. 2010, 43, 778–782. [Google Scholar] [CrossRef] [Green Version]
- Collin, P.; Matsumura, N.; Lädermann, A.; Denard, P.J.; Walch, G. Relationship between massive chronic rotator cuff tear pattern and loss of active shoulder range of motion. J. Shoulder Elb. Surg. 2014, 23, 1195–1202. [Google Scholar] [CrossRef] [PubMed]
- Galatz, L.M.; Charlton, N.; Das, R.; Kim, H.; Havlioglu, N.; Thomopoulos, S. Complete removal of load is detrimental to rotator cuff healing. J. Shoulder Elb. Surg. 2009, 18, 669–675. [Google Scholar] [CrossRef]
- Dines, J.S.; Weber, L.; Razzano, P.; Prajapati, R.; Timmer, M.; Bowman, S.; Bonasser, L.; Dines, D.M.; Grande, D.P. The effect of growth differentiation factor-5–coated sutures on tendon repair in a rat model. J. Shoulder Elb. Surg. 2007, 16, S215–S221. [Google Scholar] [CrossRef]
- Murray, D.H.; Kubiak, E.N.; Jazrawi, L.M.; Araghi, A.; Kummer, F.; Loebenberg, M.I.; Zuckerman, J.D. The effect of cartilage-derived morphogenetic protein 2 on initial healing of a rotator cuff defect in a rat model. J. Shoulder Elb. Surg. 2007, 16, 251–254. [Google Scholar] [CrossRef] [PubMed]
- Willbold, E.; Wellmann, M.; Welke, B.; Angrisani, N.; Gniesmer, S.; Kampmann, A.; Hoffmann, A.; De Cassan, D.; Menzel, H.; Hoheisel, A.L.; et al. Possibilities and limitations of electrospun chitosan-coated polycaprolactone grafts for rotator cuff tear repair. J. Tissue Eng. Regen. Med. 2019, 14, 186–197. [Google Scholar] [CrossRef] [PubMed]
- Ide, J.; Kikukawa, K.; Hirose, J.; Iyama, K.-I.; Sakamoto, H.; Mizuta, H. Reconstruction of large rotator-cuff tears with acellular dermal matrix grafts in rats. J. Shoulder Elb. Surg. 2009, 18, 288–295. [Google Scholar] [CrossRef]
- Turner, A.S. Experiences with sheep as an animal model for shoulder surgery: Strengths and shortcomings. J. Shoulder Elb. Surg. 2007, 16, S158–S163. [Google Scholar] [CrossRef] [PubMed]
- Gerber, C.; Schneeberger, A.G.; Perren, S.M.; Nyffeler, R.W. Experimental Rotator Cuff Repair. A Preliminary Study*. J. Bone Jt. Surg.-Am. Vol. 1999, 81, 1281–1290. [Google Scholar] [CrossRef]
- Derwin, K.A.; Codsi, M.J.; Milks, R.A.; Baker, A.R.; McCarron, J.A.; Iannotti, J.P. Rotator Cuff Repair Augmentation in a Canine Model with Use of a Woven Poly-L-Lactide Device. J. Bone Jt. Surg.-Am. Vol. 2009, 91, 1159–1171. [Google Scholar] [CrossRef] [Green Version]
- Huri, G.; Biçer, Ö.S.; Özgözen, L.; Uçar, Y.; Garbis, N.G.; Hyun, Y.S. A novel repair method for the treatment of acute Achilles tendon rupture with minimally invasive approach using button implant: A biomechanical study. Foot Ankle Surg. 2013, 19, 261–266. [Google Scholar] [CrossRef]
- Baums, M.H.; Buchhorn, G.H.; Spahn, G.; Poppendieck, B.; Schultz, W.; Klinger, H.-M. Biomechanical characteristics of single-row repair in comparison to double-row repair with consideration of the suture configuration and suture material. Knee Surg. Sports Traumatol. Arthrosc. 2008, 16, 1052–1060. [Google Scholar] [CrossRef] [Green Version]
- Nelson, C.O.; Sileo, M.J.; Grossman, M.G.; Serra-Hsu, F. Single-Row Modified Mason-Allen versus Double-Row Arthroscopic Rotator Cuff Repair: A Biomechanical and Surface Area Comparison. Arthrosc. J. Arthrosc. Relat. Surg. 2008, 24, 941–948. [Google Scholar] [CrossRef]
- Ozbaydar, M.; Elhassan, B.; Esenyel, C.; Atalar, A.; Bozdag, E.; Sunbuloglu, E.; Kopuz, N.; Demirhan, M. A comparison of single-versus double-row suture anchor techniques in a simulated repair of the rotator cuff. J. Bone Jt. Surg. Br. Vol. 2008, 90-B, 1386–1391. [Google Scholar] [CrossRef]
- Schlegel, T.F.; Hawkins, R.J.; Lewis, C.W.; Turner, A.S. An in vivo comparison of the modified Mason-Allen suture technique versus an inclined horizontal mattress suture technique with regard to tendon-to-bone healing: A biomechanical and histologic study in sheep. J. Shoulder Elb. Surg. 2007, 16, 115–121. [Google Scholar] [CrossRef]
- Coleman, S.H.; Fealy, S.; Ehteshami, J.R.; MacGillivray, J.D.; Altchek, D.W.; Warren, R.F.; Turner, A.S. Chronic rotator cuff injury and repair model in sheep. J. Bone Jt. Surg.-Am. Vol. 2003, 85, 2391–2402. [Google Scholar] [CrossRef]
- Frey, E.; Regenfelder, F.; Sussmann, P.; Zumstein, M.; Gerber, C.; Born, W.; Fuchs, B. Adipogenic and myogenic gene expression in rotator cuff muscle of the sheep after tendon tear. J. Orthop. Res. 2009, 27, 504–509. [Google Scholar] [CrossRef] [PubMed]
- Rowshan, K.; Hadley, S.; Pham, K.; Caiozzo, V.; Lee, T.Q.; Gupta, R. Development of Fatty Atrophy after Neurologic and Rotator Cuff Injuries in an Animal Model of Rotator Cuff Pathology. J. Bone Jt. Surg.-Am. Vol. 2010, 92, 2270–2278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Warner, J.J.; Higgins, L.; Parsons, I.; Dowdy, P. Diagnosis and treatment of anterosuperior rotator cuff tears. J. Shoulder Elb. Surg. 2001, 10, 37–46. [Google Scholar] [CrossRef] [PubMed]
- Czernik, M.; Fidanza, A.; Sardi, M.; Galli, C.; Brunetti, D.; Malatesta, D.; Della Salda, L.; Matsukawa, K.; Ptak, G.E.; Loi, P. Differentiation potential and GFP labeling of sheep bone marrow-derived mesenchymal stem cells. J. Cell. Biochem. 2013, 114, 134–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Andersson, G.; Backman, L.J.; Scott, A.; Lorentzon, R.; Forsgren, S.; Danielson, P. Substance P accelerates hypercellularity and angiogenesis in tendon tissue and enhances paratendinitis in response to Achilles tendon overuse in a tendinopathy model. Br. J. Sports Med. 2011, 45, 1017–1022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ng, G.Y.-F.; Chung, P.Y.-M.; Wang, J.S.; Cheung, R.T.-H. Enforced Bipedal Downhill Running Induces Achilles Tendinosis in Rats. Connect. Tissue Res. 2011, 52, 466–471. [Google Scholar] [CrossRef]
- Cho, N.S.; Hwang, J.H.; Lee, Y.T.; Chae, S.W. Tendinosis-like Histologic and Molecular Changes of the Achilles Tendon to Repetitive Stress: A Pilot Study in Rats. Clin. Orthop. Relat. Res. 2011, 469, 3172–3180. [Google Scholar] [CrossRef] [Green Version]
- Thorpe, C.; Clegg, P.; Birch, H. A review of tendon injury: Why is the equine superficial digital flexor tendon most at risk? Equine Vet. J. 2010, 42, 174–180. [Google Scholar] [CrossRef]
- Alexander, R.M. Tendon elasticity and muscle function. Comp. Biochem. Physiol. Part A Mol. Integr. Physiol. 2002, 133, 1001–1011. [Google Scholar] [CrossRef]
- Dudhia, J.; Scott, C.M.; Draper, E.; Heinegård, D.; Pitsillides, A.A.; Smith, R.K. Aging enhances a mechanically-induced reduction in tendon strength by an active process involving matrix metalloproteinase activity. Aging Cell 2007, 6, 547–556. [Google Scholar] [CrossRef] [PubMed]
- Ribitsch, I.; Baptista, P.M.; Lange-Consiglio, A.; Melotti, L.; Patruno, M.; Jenner, F.; Schnabl-Feichter, E.; Dutton, L.C.; Connolly, D.J.; Van Steenbeek, F.G.; et al. Large Animal Models in Regenerative Medicine and Tissue Engineering: To Do or Not to Do. Front. Bioeng. Biotechnol. 2020, 8, 972. [Google Scholar] [CrossRef]
- Hurtig, M.B.; Buschmann, M.; Fortier, L.A.; Hoemann, C.; Hunziker, E.B.; Jurvelin, J.S.; Mainil-Varlet, P.; McIlwraith, C.W.; Sah, R.L.; Whiteside, R.A. Preclinical Studies for Cartilage Repair: Recommendations from the International Cartilage Repair Society. Cartilage 2011, 2, 137–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oreff, G.L.; Fenu, M.; Vogl, C.; Ribitsch, I.; Jenner, F. Species variations in tenocytes’ response to inflammation require careful selection of animal models for tendon research. Sci. Rep. 2021, 11, 1–14. [Google Scholar] [CrossRef]
- Svensson, M.; Kartus, J.; Christensen, L.R.; Movin, T.; Papadogiannakis, N.; Karlsson, J. A long-term serial histological evaluation of the patellar tendon in humans after harvesting its central third. Knee Surg. Sports Traumatol. Arthrosc. 2005, 13, 398–404. [Google Scholar] [CrossRef] [PubMed]
- Cook, J.L.; Feller, J.A.; Bonar, S.F.; Khan, K.M. Abnormal tenocyte morphology is more prevalent than collagen disruption in asymptomatic athletes’ patellar tendons. J. Orthop. Res. 2004, 22, 334–338. [Google Scholar] [CrossRef]
- Jiang, D.; Gao, P.; Lin, H.; Geng, H. Curcumin improves tendon healing in rats: A histological, biochemical, and functional evaluation. Connect. Tissue Res. 2016, 57, 20–27. [Google Scholar] [CrossRef] [PubMed]
- Shepherd, J.H.; Screen, H. Fatigue loading of tendon. Int. J. Exp. Pathol. 2013, 94, 260–270. [Google Scholar] [CrossRef]
- Neyton, L.; Godenèche, A.; Nové-Josserand, L.; Carrillon, Y.; Cléchet, J.; Hardy, M.B. Arthroscopic Suture-Bridge Repair for Small to Medium Size Supraspinatus Tear: Healing Rate and Retear Pattern. Arthrosc. J. Arthrosc. Relat. Surg. 2013, 29, 10–17. [Google Scholar] [CrossRef]
- Kurtaliaj, I.; Golman, M.; Abraham, A.C.; Thomopoulos, S. Biomechanical Testing of Murine Tendons. J. Vis. Exp. 2019, e60280. [Google Scholar] [CrossRef] [PubMed]
- Meeremans, M.; Van de Walle, G.R.; Van Vlierberghe, S.; De Schauwer, C. The Lack of a Representative Tendinopathy Model Hampers Fundamental Mesenchymal Stem Cell Research. Front. Cell Dev. Biol. 2021, 9. [Google Scholar] [CrossRef] [PubMed]
Article | Source | Cell Origin | Cell Treatment | MSC Verification |
---|---|---|---|---|
Shi et al. [25] | Sprague–Dawley rats | Bone marrow cells from the femur and tibia | Cultured in α-MEM containing 10% FBS until third to fifth passage. MSCs were then cultured in Mesen Gro MSC medium. | Flow cytometry: CD44, CD90 +ve; CD11b, CD34 −ve |
Shen et al. [26] | NGL transgenic reporter mice and Scleraxis–GFP tendon reporter mice | Adipose tissue (subcutaneous fat) | Cultured in 10% FBS, 100 unit/mL penicillin, and 100 μg/mL streptomycin in α-MEM | Flow cytometry: CD29, CD44, CD90 +ve |
Yu et al. [27] | Sprague–Dawley rats | Bone marrow cells from the femur and tibia | Cultured in α-MEM containing 10% FBS, 100 U/mL penicillin and 100 mg/mL streptomycin until second passage. Cells were then cultured in an exosome-depleted medium. | Trilineage differentiation (adipocytes, osteoblasts, and chondrocytes); Flow cytometry: CD44, CD90 +ve |
Shi et al. [28] | C57BL/6 male mice | Bone marrow cells from the femur | Cultured in α-MEM supplemented with 20% FBS, 1% penicillin, and streptomycin until third to fifth passage. | Flow cytometry: CD44, CD90, Sca-1 +ve; CD34, CD45 −ve |
Gissi et al. [29] | Lewis rats | Bone marrow cells from the femur and tibia | Cultured in MesenCult Basal Medium, supplemented with penicillin/streptomycin (100 U/mL–100 μg/mL) and 10% FBS until second passage. | Trilineage differentiation (adipocytes, osteoblasts, and chondrocytes); Flow cytometry: CD29, CD44, CD90 +ve; CD45, CD34 −ve |
Yao et al. [30] | Human | Umbilical cord | Cultured in α-MEM mixed with 10% FBS until 3rd to 5th passage. | Not done. |
Chamberlain et al. [31] | Human | Bone marrow cells | Cultured in α-MEM mixed with 10% FBS, 1× nonessential amino acids, and 4 mM l-glutamine until 4th to 6th passage. | Not done. |
Huang et al. [32] | Sprague–Dawley rats | Bone marrow cells from the femur and tibia | Cultured in standard media comprising DMEM supplemented with 10% FBS and 1% double antibiotics (streptomycin + penicillin). | Trilineage differentiation (adipocytes, osteoblasts, and chondrocytes); Flow cytometry: CD44, CD73, CD90, CD105 +ve; CD34 −ve |
Li et al. [33] | Human | Umbilical cord | Cultured α-MEM supplemented with 10% FBS and 1% penicillin/streptomycin | Not done. |
Wang et al. [34] | Sprague–Dawley rats | Tendon stem cells from the Achilles tendon | Cultured in DMEM containing 10% FBS, 100 U/mL penicillin and 100 mg/mL streptomycin until day 7. Cells were then trypsinised with EDTA. | Trilineage differentiation (adipocytes, osteoblasts, and chondrocytes). Immunostaining; CD44, CD90 +ve; CD3, CD34 −ve, staining by Sirius red, Oil Red O and Alizarin red respectively. |
Wang et al. [35] | Human | Adipose tissue (subcutaneous fat) | Cultured in serum-free medium (OriCell). The second and third passages were used for ASCs-Exos isolation. | Not done. |
Article | EV Purification | EV Dimensions | EV Biomarkers | Imaging | Active Component |
---|---|---|---|---|---|
Shi et al. [25] | Conditioned media concentrated by several centrifugation and ultracentrifugation, then passed through a 0.22 μm filter. | qNano Gold: 70–600 nm in diameter. | CD9, CD63, HSP70 | TEM | Not assessed |
Shen et al. [26] | “Conditioned media concentrated by several centrifugation and ultracentrifugation, then passed through a 0.22 μm filter. | qNano Gold: Mode diameter of iEVs and EVs were 108 ± 2 nm and 113 ± 3 nm, respectively. | CD9, CD63 | TEM | Not assessed |
Yu et al. [27] | Conditioned media concentrated by several centrifugation and ultracentrifugation. | NTA with ZetaView: 101.1 ± 50.6 nm in diameter | CD9, ALIX, TSG101 | TEM | Not assessed |
Shi et al. [28] | Conditioned media concentrated by several ultracentrifugation steps. | NTA with ZetaView: average diameter 120.3 nm; peak of size distribution 127.1 nm | CD81, TSG101, CD9 | TEM | Not assessed |
Gissi et al. [29] | Conditioned media concentrated by several centrifugation and ultracentrifugation. | AFM: 40–280 mm in diameter, with a peak at 80 mm. | Annexin XI, Annexin V and TSG-101 +ve; GM-130 −ve | AFM | Pro-collagen1A2 and MMP14 |
Yao et al. [30] | Conditioned media concentrated by several ultracentrifugation steps. | NTA with ZetaView: 30–200 nm, average of 131 nm. | CD9, CD63, ALIX, TSG101 | 80 kV electron microscope | MicroRNA-21-3p |
Chamberlain et al. [31] | Conditioned media concentrated by differential centrifugation and ultracentrifugation steps. | qNano Gold: 61–121 nm | CD146, CD29, CD44, CD63, CD81, and CD105. | TEM | Not assessed |
Huang et al. [32] | Conditioned media concentrated by differential centrifugation and ultracentrifugation steps. | TEM: 30–150 nm | CD9, CD63, and CD81 | TEM | Not assessed |
Li et al. [33] | Conditioned media concentrated by differential centrifugation and ultracentrifugation steps. | TEM: 30–150 nm | CD9, CD63, ALIX, TSG101 | TEM | Not assessed |
Wang et al. [34] | Conditioned media concentrated by differential centrifugation and ultracentrifugation steps. | TEM: 40–200 nm | CD63 and CD81 | TEM | Not assessed |
Wang et al. [35] | Conditioned media concentrated by several centrifugation and ultracentrifugation. | qNano Gold: 50–150 nm | CD9, CD63, TSG-101 +ve, GM130 −ve | TEM | Not assessed |
Article | Method of Delivery | Animal Model | Number Used | Animal Age | Animal Weight | Animal Gender | Follow-up | Number per Experimental Group |
---|---|---|---|---|---|---|---|---|
Shi et al. [25] | 10 µL of fibrin containing 25 µg BMSC-EVs was applied around the injury site. | Sprague–Dawley rats Central 1/3 of patellar tendon was removed from the distal apex of the patella to the insertion of the tibial tuberosity. | 48 | N/A | N/A | Male | 2 weeks (immunohistochemistry analysis) 4 weeks (histological analysis) | (1) BMSC-EVs group (n = 16) (2) Fibrin group (n = 16) (3) Control group, left untreated (n = 16) |
Shen et al. [26] | ASC-EVs were loaded to the surface of a collagen sheet, that was cut into strips, each containing 5–6 × 109 EVs. Applied around the defect site. | NGL transgenic reporter mice. Right Achilles tendon 2/3 transection at midpoint level between calcaneal insertion and musculotendinous junction. | 32 | 3–4 months | 27 ± 5 g | Male and female | 7 days | (1) Collagen sheet loaded with EVs from naïve ASCs (n = 11) (2) Collagen sheet loaded with EVs from IFNγ-primed ASCs (n = 10) (3) Collagen sheet only (n = 11) |
Yu et al. [27] | 5 µL of BMSCs-exos (4 µg/µL) was mixed with 1 µL thrombin (500 IU/mL) and 4 µL fibrinogen (50 mg/mL), injected into the defect site. | Sprague–Dawley rats Central 1/3 of the patellar tendon (0.8 mm in width) was removed from the distal apex of the patella to the insertion of the tibial tuberosity. | 52 | Adult | 200 g | Male | 1 week, 2 weeks, and 4 weeks (macroscopic and histological examination) 4 weeks (mechanical test) | (1) Fibrin-exos (fibrinogen, thrombin, and exosomes injected) (n = 26) (2) Fibrin-vehicle (fibrinogen, thrombin, and PBS injected) (n = 26) |
Shi et al. [28] | Exosomes were mixed with hydrogel before implantation into the cut Achilles tendon. | C57BL/6 mice. The Achilles tendon was cut off above the calcaneus. | 90 | 8 weeks | 20–25 g | Male | 7 days | (1) Control group (n = 30) (2) Hydrogel group (n = 30) (3) Hydrogel + exosome group (n = 30) |
Gissi et al. [29] | 50 μL of PBS was injected locally with either EVL (2.8 × 1012) or EVH (8.4 × 1012). | Lewis mice. Bilateral Achilles tendon defect 2 mm in diameter was made in each animal. | 16 | Adult | 180–200 g | Male | 30 days | (1) PBS alone (control group) (n = 4) (2) rBMSCgroup: 4 × 106 cells (n = 4) (3) EVL group: 2.8 × 1012 EVs (n = 4) (4) EVH group: 8.4 × 1012 EVs. (n = 4) |
Yao et al. [30] | Injected subcutaneously around injury site with HUMSC-Exos (200 μg) dissolved in PBS and an equal volume of PBS (50 μL). | Sprague-Dawley rats. The Achilles tendon was cut in the middle. | 60 | Adult | 200–250 g | Male | 3 weeks | (1) Sham group (n = 20) (2) HUMSC-Exos group (n = 20) (3) PBS group (n = 20) |
Chamberlain et al. [31] | Injected 1 × 109 exosomes to MCL transection site. | Wistar rats Bilateral MCL transection at its midpoint. | 10 | Adult | 300–350 g | Male | 14 days | (1) Exosomes (n = 5) (2) PBS (control) (n = 5) |
Huang et al. [32] | 200 μg of BMSC-Exos precipitated in 200 μL of PBS was injected into the tail vein. | Sprague-Dawley rats. 2 mm of the distal tendon of the supraspinatus was cut off. | 54 | 4 weeks | 70–100 g | Male | 4 weeks | (1) BMSC-Exos group (n = 27) (2) PBS (control) (n = 27) |
Li et al. [33] | HCPT-EVs were both subcutaneously injected at the injury site at a dose of 200 μg. | 33 Sprague-Dawley rats The Achilles tendon was transected in the middle and repaired using the 6-0 polypropylene suture. | 33 | Adult | 250–300 g | Male | 3 weeks | (1) PBS (n = 11) (2) Unprimed EV injection (n = 11) (3) HCPT-EV injection (n = 11) |
Wang et al. [34] | 20 μL of exosomes (486.3 μg/mL) was injected into the Achilles tendon injury site twice a week. | 18 male Sprague-Dawley rats Rats were injected with 30 µL type I collagenase solution (10 mg/mL) into both Achilles tendons. | 18 | 8 weeks | 200–250 g | Male | 4 weeks | (1) PBS (control) (n = 6) (2) Injury group with TSCs (n = 6) (3) Injury group with exosomes (n = 6) |
Wang et al. [35] | 1011 ASC-Exos suspended in 20 µL of saline were injected at the injury site of the supraspinatus muscle. | Rabbits: Bilateral rotator cuff tear model. The supraspinatus tendon was detached at the insertion on the humerus. The torn tendon was wrapped with a silicon Penrose drain to prevent adhesion. | 35 | 4 months | 3.3 ± 0.3 kg | Male | 18 weeks | (1) Repair + saline (n = 7) (2) Repair + ASC-Exos (n = 7) (3) Sham surgery (n = 14) (4) Fatty infiltration assay (n = 7) |
Article | Macroscopic Appearance | Imaging and Histology | Biochemical Analysis | Biomechanical Analysis |
---|---|---|---|---|
Shi et al. [25] | Not undertaken. | Regularly aligned and compact collagen fibres. Fibre alignment score of 2 (50% to 75% parallel fibre alignment) Increased tendon cell proliferation, especially after treatment with BMSC-EVs at 20 μg/mL. | Elevated number of cells expressing CD163, IL-4, and IL-10 in the BMSC-EVs group. Reduced number of cells expressing IFNγ, IL-1B, IL-6, and CCR7 in the BMSC-EVs group. Increased expression of SCX, TNMD, COL1a1, and COL3a1 in the BMSC-EVs group. Increased gene expression of collagen type I in the BMSC-EVs group. Reduced cleaved caspase 3 signals in the BMSC-EVs group. | Not done |
Shen et al. [26] | Not undertaken. | Significant reductions of NF-κB activity in iEV-treated tendons compared to untreated tendons, but little reduction after EV treatment. Lower gap-rupture rate. iEV-treated tendons exhibited more collagen staining at the site of tendon injury than did untreated and EV-treated tendons. | Expression levels of inflammatory genes Ifng, Nos2, Tnf, Il6, Mmp1, Col1a1 and Col3a1 increased after injury. Treatment with iEV but not EVs significantly reduced Il1b and Ifng expression. Treatment with both iEVs and EVs significantly attenuated the Mmp1 expression, increased Col2a1 and Sox9 expression. iEV but not EV treatment further increased both Col1a1 and Col3a1 expression. | Not done |
Yu et al. [27] | The exosome-treated group showed improved integration of the healing tissue with the host tendon at week 2, and showed a more approximate appearance (colour and transparency) to the native tendon at week 4. | More deposition of extracellular matrix type I collagen at week 2. At week 4, cell density and alignment in the defect region of the exosome-treated group were much closer to the native tendon. Lower histological score in the exosome-treated group at week 4 (suggesting better tendon regeneration). | The exosome-treated group showed much higher expression of Col I and Tnmd. The ratio of proliferating CD146+ TSPC to total CD146+ cells was 1.73–fold higher in the exosome-treated group 1–3 days post-injury, but not afterwards. | Method: The tendon tissue was put on a universal tensile testing machine (AGS-X, SHIMADZU), cyclically elongated for 20 cycles, and stress at failure was calculated as ultimate load divided by cross sectional area. Results: The stress at failure of the healing tendons and modulus were 1.84–fold and 1.86–fold higher in the exosome-treated group compared to the control. |
Shi et al. [28] | Initially, there was less scar hyperplasia in the hydrogel + exosome group than in the control and hydrogel groups. | In the hydrogel+exosome group, a transition structure similar to tendon–bone interface was seen, chondrocyte numbers increased and were tightly arranged, collagen tissues were arranged orderly. | M2 macrophages (Arg1+) increased and M1 macrophages (iNOS+) decreased in the hydrogel+exosome group. The hydrogel+exosome group showed decreased IL-1β and IL-6 and increased IL-10 and TGF-β1. Reduced TUNEL-positive apoptotic cells, increased CD146-positive stem cells in the hydrogel+exosome group. Increased gene expression level of collagen II in hydrogel+exosome group. | Method: The tissue was loaded into a universal testing machine, preloaded with small tension, then stretched to failure at a constant speed. Results: Maximum force, elastic modulus, and strength in the hydrogel+exosome group were higher than hydrogel and control groups, but no significant difference with the normal group. No significant difference in stiffness between groups. |
Gissi et al. [29] | Not undertaken. | Lower overall histomorphometric score in EVH group than rBMSC and EVL groups. Only the cellularity sub-score in the EVH group was higher than the control group. Overall suggests a better restoration of tendon architecture, optimal alignment of tendon fibres and blood vessels in the EVH group. | The EVH group had a more favourable collagen ratio: higher collagen type I and lower collagen type III than rBMSC, EVL and control groups. | Not done. |
Yao et al. [30] | The degree of adhesion of tendon tissue with HUMSC-Exos application was lower than in the PBS and sham groups. Lower adhesion grade score in the HUMSC-Exos group. | Hyperproliferative adhesion tissue, and degree of inflammatory infiltration were lower in the HUMSC-Exos group compared to the PBS and sham groups. The HUMSC-Exos group had the lowest histological adhesion score. The histological healing score was not statistically different among the three groups. The HUMSC-Exos group had the least collagen deposition. | HUMSC-Exos significantly decreased COL III, α-SMA, p-p65, and COX2 expression. | Method: Tendon tissue fixed to a biomechanical analyser (Instron 8841 DynaMight axial servo hydraulic test system), stretched at constant speed until the tendon broke. Results: No significant difference in maximum tensile strength between the three groups. |
Chamberlain et al. [31] | Treatment with exosomes significantly reduced scar formation 14 days post-injury compared to the control. | Exosome treatment increased type I and type III collagen production within the granulation tissue. Exosome treatment improved collagen organisation. | IHC analysis CD68, CD163, CD31, and α-smooth muscle actin levels, to identify M1 and M2 macrophages; no changes elicited in M1 and M2 macrophages. | Method: Mounted in the mechanical testing machine, preconditioned with cyclic preloading, then pulled to failure at constant strain rate, with parameters recorded. Results: Treatment with exosomes did not significantly improve mechanical function. |
Huang et al. [32] | Not done. | Angiography showed that BMSC-Exos promoted angiogenesis around the rotator cuff endpoint. | BMSC-Exos promoted the expression of CD31 and endomucin. BMSC-Exos significantly reduced the serum levels of TNF-α, IL-1β, IL-6, and IL-8. BMSC-Exos promoted Col I and Col II expression, expression of Sharpey’s fibres and proteoglycan at the tendon-bone interface. | Method: Freshly excised tissue was immediately placed in paraformaldehyde solution, then loaded onto biomechanical tester, with a constant displacement distance being applied until failure. Results: BMSC-Exos increased the maximum breaking load and stiffness. |
Li et al. [33] | Macroscopic observation showed that both HCPT-EVs and unprimed EVs effectively attenuated tendon adhesion to peri-tendinous tissues. | Histological adhesion scores based on histological findings. The results showed that both unprimed EVs treatment and HCPT-EVs treatment dramatically lowered the adhesion grade of the tendon. Comparing the scores achieved by HCPT-EVs with unprimed EVs showed a tendency toward decreasing, although it was not significant. The histological healing score was significantly lower in the group treated with HCPT-EVs than with unprimed EVs. | HCPT-EVs more effectively decreased myofibroblast activation induced by TGFβ after tendon injury, as demonstrated by weaker WB staining of both COL III and α-SMA in unprimed EVs or HCPT-EVs. qRT-PCR analysis suggested that unprimed EVs and HCPT-EVs suppressed COL III and α-SMA at the transcription level and displayed a larger decreasing trend after HCPT-EVs administration than with unprimed EVs. | Method: Both ends of the tissue are clamped in a tensile testing system, and stretched at a constant speed of 10mm/minute until rupture. The maximum tensile force was recorded. Five samples from each group were used for analysis. Results: The maximal tensile strength of the regenerated tendon remained the same among the three groups. |
Wang et al. [34] | Not done. | The arrangement of collagens in the exosomes group was more uniform than that of the injury group. | TSCs injection and exosomes injection significantly decreased matrix metalloproteinases (MMP)-3 expression, increased expression of tissue inhibitor of metalloproteinase-3 (TIMP-3) and Col-1a1 | Method: The two bony ends of the tendon were fixed on a custom-made testing jig with two clamps. No further details are available. Results: Ultimate stress and maximum loading were significantly increased in the exosome treated group compared with injury. No TSC vs exosome comparison was made. |
Wang et al. [35] | Fatty infiltration was significantly higher in rabbits with rotator cuff tear than those receiving sham surgery (confirming the establishment of a rotator cuff tear model). | Few inflammatory cells were present in the ASC-Exos group than in the saline group Cellularity and vessel numbers at tendon-bone interface of the ASC-Exos group were significantly lower than those in the saline group. The fibrocartilage area in the ASC-Exos group was significantly greater than in the saline group. More abundant collagen II and tenascin-C appeared in the ASC-Exos group than in the saline group. | Lower expression of CD31 in the ASC-Exos group than the saline group, attributed to the maturation of tiny capillaries into proper blood vessels. | Method: Tendons harvested and loaded into clamping device. Specimens were preloaded to 5 N for 5 min, with 10 cycles of preconditioning (5 N to 30 N at a rate of 15 N/s). Then, each specimen was loaded to failure using a 0.5-mm/min uniaxial tension. Ultimate load to failure, stiffness, and stress were calculated according to the load-elongation curve. Results: Mean ultimate load to failure of the ASC-Exos group (132.7 ± 10.3 N) significantly greater than that in the saline group (96.0 ± 9.8 N) though lower than in the sham surgery group (162.2 ± 12.1 N). |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Lu, V.; Tennyson, M.; Zhang, J.; Khan, W. Mesenchymal Stem Cell-Derived Extracellular Vesicles in Tendon and Ligament Repair—A Systematic Review of In Vivo Studies. Cells 2021, 10, 2553. https://doi.org/10.3390/cells10102553
Lu V, Tennyson M, Zhang J, Khan W. Mesenchymal Stem Cell-Derived Extracellular Vesicles in Tendon and Ligament Repair—A Systematic Review of In Vivo Studies. Cells. 2021; 10(10):2553. https://doi.org/10.3390/cells10102553
Chicago/Turabian StyleLu, Victor, Maria Tennyson, James Zhang, and Wasim Khan. 2021. "Mesenchymal Stem Cell-Derived Extracellular Vesicles in Tendon and Ligament Repair—A Systematic Review of In Vivo Studies" Cells 10, no. 10: 2553. https://doi.org/10.3390/cells10102553
APA StyleLu, V., Tennyson, M., Zhang, J., & Khan, W. (2021). Mesenchymal Stem Cell-Derived Extracellular Vesicles in Tendon and Ligament Repair—A Systematic Review of In Vivo Studies. Cells, 10(10), 2553. https://doi.org/10.3390/cells10102553