Mesenchymal Stem Cell-Derived Exosomes and MicroRNAs in Cartilage Regeneration: Biogenesis, Efficacy, miRNA Enrichment and Delivery
Abstract
:1. Introduction
2. Cartilage Damage and Osteoarthritis
3. Management of Osteoarthritis
4. Exosomes
4.1. Categories
4.2. Biogenesis
4.3. Therapeutic Cargos of Exosomes
5. Promoting Cartilage Repair Using Exosomes
6. Exosomal miRNAs
6.1. Biogenesis of miRNA
6.2. Mechanism of miRNA-Mediated Gene Regulation
7. Modulating the miRNA Content of Exosomes
7.1. Cell Modification
7.1.1. Transfection
7.1.2. RNA Binding Proteins
7.1.3. Cell Priming
7.2. Direct Introduction of miRNA into Extracellular Vesicles
8. Delivery of Exosomes for Cartilage Repair
8.1. Direct Needle Injection: Exosome Gel and Formulation
8.2. Direct Needle Injection: Exosome Suspension
9. Conclusions and Perspective
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Shane Anderson, A.; Loeser, R.F. Why is osteoarthritis an age-related disease? Best Pract. Res. Clin. Rheumatol. 2010, 24, 15–26. [Google Scholar] [CrossRef] [Green Version]
- Pang, K.-L.; Chow, Y.Y.; Leong, L.M.; Law, J.X.; Ghafar, N.A.; Soelaiman, I.N.; Chin, K.-Y. Establishing SW1353 Chondrocytes as a Cellular Model of Chondrolysis. Life 2021, 11, 272. [Google Scholar] [CrossRef] [PubMed]
- Liau, L.L.; Hassan, M.N.F.B.; Tang, Y.L.; Ng, M.H.; Law, J.X. Feasibility of Human Platelet Lysate as an Alternative to Foetal Bovine Serum for In Vitro Expansion of Chondrocytes. Int. J. Mol. Sci. 2021, 22, 1269. [Google Scholar] [CrossRef]
- Armiento, A.R.; Alini, M.; Stoddart, M.J. Articular fibrocartilage—Why does hyaline cartilage fail to repair? Adv. Drug Deliv. Rev. 2019, 146, 289–305. [Google Scholar] [CrossRef]
- Liu, Y.; Zhou, G.; Cao, Y. Recent Progress in Cartilage Tissue Engineering—Our Experience and Future Directions. Engineering 2017, 3, 28–35. [Google Scholar] [CrossRef]
- Mora, J.C.; Przkora, R.; Cruz-Almeida, Y. Knee osteoarthritis: Pathophysiology and current treatment modalities. J. Pain Res. 2018, 11, 2189–2196. [Google Scholar] [CrossRef] [Green Version]
- Liau, L.L.; Al-Masawa, M.E.; Koh, B.; Looi, Q.H.; Foo, J.B.; Lee, S.H.; Cheah, F.C.; Law, J.X. The Potential of Mesenchymal Stromal Cell as Therapy in Neonatal Diseases. Front. Pediatr. 2020, 8, 591693. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Q.; Cai, Y.; Jiang, Y.; Lin, X. Exosomes in osteoarthritis and cartilage injury: Advanced development and potential therapeutic strategies. Int. J. Biol. Sci. 2020, 16, 1811. [Google Scholar] [CrossRef] [Green Version]
- Duan, L.; Liang, Y.; Xu, X.; Xiao, Y.; Wang, D. Recent progress on the role of miR-140 in cartilage matrix remodelling and its implications for osteoarthritis treatment. Arthrit. Res. Ther. 2020, 22, 194. [Google Scholar] [CrossRef] [PubMed]
- Baek, D.; Lee, K.-M.; Park, K.W.; Suh, J.W.; Choi, S.M.; Park, K.H.; Lee, J.W.; Kim, S.-H. Inhibition of miR-449a Promotes Cartilage Regeneration and Prevents Progression of Osteoarthritis in In Vivo Rat Models. Mol. Ther. Nucleic Acids 2018, 13, 322–333. [Google Scholar] [CrossRef] [Green Version]
- Lolli, A.; Sivasubramaniyan, K.; Vainieri, M.L.; Oieni, J.; Kops, N.; Yayon, A.; van Osch, G.J.V.M. Hydrogel-based delivery of antimiR-221 enhances cartilage regeneration by endogenous cells. J. Control. Release 2019, 309, 220–230. [Google Scholar] [CrossRef]
- O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sophia Fox, A.J.; Bedi, A.; Rodeo, S.A. The basic science of articular cartilage: Structure, composition, and function. Sports Health 2009, 1, 461–468. [Google Scholar] [CrossRef]
- Bhosale, A.M.; Richardson, J.B. Articular cartilage: Structure, injuries and review of management. Br. Med. Bull. 2008, 87, 77–95. [Google Scholar] [CrossRef] [PubMed]
- Zingler, C.; Carl, H.-D.; Swoboda, B.; Krinner, S.; Hennig, F.; Gelse, K. Limited evidence of chondrocyte outgrowth from adult human articular cartilage. Osteoarthr. Cartil. 2016, 24, 124–128. [Google Scholar] [CrossRef] [Green Version]
- Mobasheri, A.; Matta, C.; Uzielienè, I.; Budd, E.; Martín-Vasallo, P.; Bernotiene, E. The chondrocyte channelome: A narrative review. Jt. Bone Spine 2019, 86, 29–35. [Google Scholar] [CrossRef]
- Archer, C.W.; Francis-West, P. The chondrocyte. Int. J. Biochem. Cell Biol. 2003, 35, 401–404. [Google Scholar] [CrossRef]
- Alberton, P.; Farkas, Z.; Prein, C.; Schwarz, J.; Li, P.; Clausen-Schaumann, H.; Oohashi, T.; Aszodi, A. Aggrecan is critical in maintaining the cartilage matrix biomechanics which in turn influences the correct development of the growth plate. Osteoarthr. Cartil. 2019, 27, S178. [Google Scholar] [CrossRef] [Green Version]
- Carballo, C.B.; Nakagawa, Y.; Sekiya, I.; Rodeo, S.A. Basic science of articular cartilage. Clin. Sports Med. 2017, 36, 413–425. [Google Scholar] [CrossRef] [PubMed]
- Las Heras, F.; Gahunia, H.K.; Pritzker, K.P. Articular cartilage development: A molecular perspective. Orthop. Clin. 2012, 43, 155–171. [Google Scholar] [CrossRef] [PubMed]
- Goldring, M.B.; Marcu, K.B. Cartilage homeostasis in health and rheumatic diseases. Arthrit. Res. Ther. 2009, 11, 224. [Google Scholar] [CrossRef] [Green Version]
- Keller, B.; Yang, T.; Chen, Y.; Munivez, E.; Bertin, T.; Zabel, B.; Lee, B. Interaction of TGFβ and BMP signaling pathways during chondrogenesis. PLoS ONE 2011, 6, e16421. [Google Scholar] [CrossRef] [Green Version]
- Van der Kraan, P.M.; Buma, P.; van Kuppevelt, T.; van Den Berg, W.B. Interaction of chondrocytes, extracellular matrix and growth factors: Relevance for articular cartilage tissue engineering. Osteoarthr. Cartil. 2002, 10, 631–637. [Google Scholar] [CrossRef] [Green Version]
- Pap, T.; Korb-Pap, A. Cartilage damage in osteoarthritis and rheumatoid arthritis—Two unequal siblings. Nat. Rev. Rheumatol. 2015, 11, 606–615. [Google Scholar] [CrossRef]
- James, S.L.; Abate, D.; Abate, K.H.; Abay, S.M.; Abbafati, C.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdela, J.; Abdelalim, A. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2018, 392, 1789–1858. [Google Scholar] [CrossRef] [Green Version]
- Wang, H.; Bai, J.; He, B.; Hu, X.; Liu, D. Osteoarthritis and the risk of cardiovascular disease: A meta-analysis of observational studies. Sci. Rep. 2016, 6, 1–7. [Google Scholar]
- Kye, S.-Y.; Park, K. Suicidal ideation and suicidal attempts among adults with chronic diseases: A cross-sectional study. Compr. Psychiatry 2017, 73, 160–167. [Google Scholar] [CrossRef] [PubMed]
- Jang, S.; Lee, K.; Ju, J.H. Recent Updates of Diagnosis, Pathophysiology, and Treatment on Osteoarthritis of the Knee. Int. J. Mol. Sci. 2021, 22, 2619. [Google Scholar] [CrossRef] [PubMed]
- Cicuttini, F.M.; Wluka, A.E. Is OA a mechanical or systemic disease? Nat. Rev. Rheumatol. 2014, 10, 515–516. [Google Scholar] [CrossRef]
- Deveza, L.A.; Loeser, R.F. Is osteoarthritis one disease or a collection of many? Rheumatology 2018, 57, iv34–iv42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Trenkmann, M. Decoding osteoarthritis. Nat. Rev. Rheumatol. 2019, 15, 3. [Google Scholar] [CrossRef] [PubMed]
- Loeser, R.F.; Goldring, S.R.; Scanzello, C.R.; Goldring, M.B. Osteoarthritis: A disease of the joint as an organ. Arthritis Rheumatol. 2012, 64, 1697. [Google Scholar] [CrossRef] [Green Version]
- He, Y.; Li, Z.; Alexander, P.G.; Ocasio-Nieves, B.D.; Yocum, L.; Lin, H.; Tuan, R.S. Pathogenesis of Osteoarthritis: Risk Factors, Regulatory Pathways in Chondrocytes, and Experimental Models. Biology 2020, 9, 194. [Google Scholar] [CrossRef] [PubMed]
- Maruotti, N.; Corrado, A.; Cantatore, F.P. Osteoblast role in osteoarthritis pathogenesis. J. Cell Physiol. 2017, 232, 2957–2963. [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]
- Croft, A.P.; Campos, J.; Jansen, K.; Turner, J.D.; Marshall, J.; Attar, M.; Savary, L.; Wehmeyer, C.; Naylor, A.J.; Kemble, S.; et al. Distinct fibroblast subsets drive inflammation and damage in arthritis. Nature 2019, 570, 246–251. [Google Scholar] [CrossRef] [PubMed]
- Zapata-Linares, N.; Eymard, F.; Berenbaum, F.; Houard, X. Role of adipose tissues in osteoarthritis. Curr. Opin. Rheumatol. 2021, 33, 84–93. [Google Scholar] [CrossRef]
- McCulloch, K.; Litherland, G.J.; Rai, T.S. Cellular senescence in osteoarthritis pathology. Aging Cell 2017, 16, 210–218. [Google Scholar] [CrossRef] [PubMed]
- Rim, Y.A.; Nam, Y.; Ju, J.H. The Role of Chondrocyte Hypertrophy and Senescence in Osteoarthritis Initiation and Progression. Int. J. Mol. Sci. 2020, 21, 2358. [Google Scholar] [CrossRef] [Green Version]
- Tsang, K.Y.; Chan, D.; Cheah, K.S. Fate of growth plate hypertrophic chondrocytes: Death or lineage extension? Dev. Growth Differ. 2015, 57, 179–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lotz, M.K.; Otsuki, S.; Grogan, S.P.; Sah, R.; Terkeltaub, R.; D’Lima, D. Cartilage cell clusters. Arthritis Rheumatol. 2010, 62, 2206–2218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prasadam, I.; Crawford, R.; Xiao, Y. Aggravation of ADAMTS and Matrix Metalloproteinase Production and Role of ERK1/2 Pathway in the Interaction of Osteoarthritic Subchondral Bone Osteoblasts and Articular Cartilage Chondrocytes—Possible Pathogenic Role in Osteoarthritis. J. Rheumatol. 2012, 39, 621–634. [Google Scholar] [CrossRef] [PubMed]
- Yang, C.Y.; Chanalaris, A.; Troeberg, L. ADAMTS and ADAM metalloproteinases in osteoarthritis—Looking beyond the ‘usual suspects’. Osteoarthr. Cartil. 2017, 25, 1000–1009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hwang, H.S.; Kim, H.A. Chondrocyte Apoptosis in the Pathogenesis of Osteoarthritis. Int. J. Mol. Sci. 2015, 16, 26035–26054. [Google Scholar] [CrossRef]
- Musumeci, G.; Loreto, C.; Carnazza, M.L.; Martinez, G. Characterization of apoptosis in articular cartilage derived from the knee joints of patients with osteoarthritis. Knee Surg. Sports Traumatol. Arthrosc. 2011, 19, 307–313. [Google Scholar] [CrossRef]
- Diekman, B.O.; Sessions, G.A.; Collins, J.A.; Knecht, A.K.; Strum, S.L.; Mitin, N.K.; Carlson, C.S.; Loeser, R.F.; Sharpless, N.E. Expression of p16(INK) (4a) is a biomarker of chondrocyte aging but does not cause osteoarthritis. Aging Cell 2018, 17, e12771. [Google Scholar] [CrossRef]
- Del Rey, M.J.; Valín, Á.; Usategui, A.; Ergueta, S.; Martín, E.; Municio, C.; Cañete, J.D.; Blanco, F.J.; Criado, G.; Pablos, J.L. Senescent synovial fibroblasts accumulate prematurely in rheumatoid arthritis tissues and display an enhanced inflammatory phenotype. Immun. Ageing 2019, 16, 29. [Google Scholar] [CrossRef] [Green Version]
- Coryell, P.R.; Diekman, B.O.; Loeser, R.F. Mechanisms and therapeutic implications of cellular senescence in osteoarthritis. Nat. Rev. Rheumatol. 2021, 17, 47–57. [Google Scholar] [CrossRef]
- Martin, J.A.; Brown, T.; Heiner, A.; Buckwalter, J.A. Post-traumatic osteoarthritis: The role of accelerated chondrocyte senescence. Biorheology 2004, 41, 479–491. [Google Scholar]
- Jeon, O.H.; Kim, C.; Laberge, R.M.; Demaria, M.; Rathod, S.; Vasserot, A.P.; Chung, J.W.; Kim, D.H.; Poon, Y.; David, N.; et al. Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment. Nat. Med. 2017, 23, 775–781. [Google Scholar] [CrossRef]
- Lu, Y.; Liu, L.; Pan, J.; Luo, B.; Zeng, H.; Shao, Y.; Zhang, H.; Guan, H.; Guo, D.; Zeng, C. MFG-E8 regulated by miR-99b-5p protects against osteoarthritis by targeting chondrocyte senescence and macrophage reprogramming via the NF-κB pathway. Cell Death Dis. 2021, 12, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Lin, Z.; Chen, D.; He, Y. CY-09 attenuates the progression of osteoarthritis via inhibiting NLRP3 inflammasome-mediated pyroptosis. Biochem. Biophys. Res. Commun. 2021, 553, 119–125. [Google Scholar] [CrossRef]
- Chen, Y.; Zhao, B.; Zhu, Y.; Zhao, H.; Ma, C. HIF-1-VEGF-Notch mediates angiogenesis in temporomandibular joint osteoarthritis. Am. J. Transl. Res. 2019, 11, 2969–2982. [Google Scholar]
- Gao, S.-C.; Yin, H.-B.; Liu, H.-X.; Sui, Y.-H. Research progress on MAPK signal pathway in the pathogenesis of osteoarthritis. Zhongguo Gu Shang 2014, 27, 441–444. [Google Scholar] [PubMed]
- Moens, U.; Kostenko, S.; Sveinbjørnsson, B. The Role of Mitogen-Activated Protein Kinase-Activated Protein Kinases (MAPKAPKs) in Inflammation. Genes 2013, 4, 101–133. [Google Scholar] [CrossRef]
- Simon, T.M.; Jackson, D.W. Articular cartilage: Injury pathways and treatment options. Sports Med. Arth. Rev. 2018, 26, 31–39. [Google Scholar] [CrossRef]
- Katz, J.N.; Neogi, T.; Callahan, L.F.; Block, J.A.; Conaghan, P.G.; Simon, L.S.; Kraus, V.B.; Hochberg, M.C. Disease modification in osteoarthritis; pathways to drug approval. Osteoarthr. Cartil. Open 2020, 2, 100059. [Google Scholar] [CrossRef]
- Bannuru, R.R.; Osani, M.C.; Vaysbrot, E.E.; Arden, N.K.; Bennell, K.; Bierma-Zeinstra, S.M.A.; Kraus, V.B.; Lohmander, L.S.; Abbott, J.H.; Bhandari, M.; et al. OARSI guidelines for the non-surgical management of knee, hip, and polyarticular osteoarthritis. Osteoarthr. Cartil. 2019, 27, 1578–1589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cai, X.; Yuan, S.; Zeng, Y.; Wang, C.; Yu, N.; Ding, C. New Trends in Pharmacological Treatments for Osteoarthritis. Front. Pharmacol. 2021, 12, 701. [Google Scholar] [CrossRef]
- Oo, W.M.; Yu, S.P.-C.; Daniel, M.S.; Hunter, D.J. Disease-modifying drugs in osteoarthritis: Current understanding and future therapeutics. Expert Opin. Emerg. Drugs 2018, 23, 331–347. [Google Scholar] [CrossRef]
- Steadman, J.R.; Rodkey, W.G.; Rodrigo, J.J. Microfracture: Surgical Technique and Rehabilitation to Treat Chondral Defects. Clin. Orthop. Relat. Res. 2001, 391, S362–S369. [Google Scholar] [CrossRef] [PubMed]
- Steinwachs, M.R.; Guggi, T.; Kreuz, P.C. Marrow stimulation techniques. Injury 2008, 39, 26–31. [Google Scholar] [CrossRef]
- Kreuz, P.C.; Erggelet, C.; Steinwachs, M.R.; Krause, S.J.; Lahm, A.; Niemeyer, P.; Ghanem, N.; Uhl, M.; Südkamp, N. Is Microfracture of Chondral Defects in the Knee Associated With Different Results in Patients Aged 40 Years or Younger? Arthrosc. J. Arthrosc. Relat. Surg. 2006, 22, 1180–1186. [Google Scholar] [CrossRef]
- Brittberg, M.; Lindahl, A.; Nilsson, A.; Ohlsson, C.; Isaksson, O.; Peterson, L. Treatment of Deep Cartilage Defects in the Knee with Autologous Chondrocyte Transplantation. N. Engl. J. Med. 1994, 331, 889–895. [Google Scholar] [CrossRef] [PubMed]
- Feucht, M.J.; Izadpanah, K.; Vogt, S.; Mehl, J. Cartilage repair procedures for early osteoarthritis. Orthopade 2021, 50, 356–365. [Google Scholar] [CrossRef]
- Niemeyer, P.; Albrecht, D.; Andereya, S.; Angele, P.; Ateschrang, A.; Aurich, M.; Baumann, M.; Bosch, U.; Erggelet, C.; Fickert, S.; et al. Autologous chondrocyte implantation (ACI) for cartilage defects of the knee: A guideline by the working group “Clinical Tissue Regeneration” of the German Society of Orthopaedics and Trauma (DGOU). Knee 2016, 23, 426–435. [Google Scholar] [CrossRef] [Green Version]
- Hassan, M.N.F.B.; Yazid, M.D.; Yunus, M.H.M.; Chowdhury, S.R.; Lokanathan, Y.; Idrus, R.B.H.; Ng, A.M.H.; Law, J.X. Large-scale expansion of human mesenchymal stem cells. Stem Cells Int. 2020, 2020, 9529465. [Google Scholar] [CrossRef] [PubMed]
- Looi, Q.H.; Eng, S.P.; Liau, L.L.; Tor, Y.S.; Bajuri, M.Y.; Ng, M.H.; Xian, J. Mesenchymal stem cell therapy for sports injuries-From research to clinical practice. Sains Malays. 2020, 49, 825–838. [Google Scholar] [CrossRef]
- Le, H.; Xu, W.; Zhuang, X.; Chang, F.; Wang, Y.; Ding, J. Mesenchymal stem cells for cartilage regeneration. J. Tissue Eng. 2020, 11, 2041731420943839. [Google Scholar] [CrossRef] [PubMed]
- Zhang, R.; Ma, J.; Han, J.; Zhang, W.; Ma, J. Mesenchymal stem cell related therapies for cartilage lesions and osteoarthritis. Am. J. Transl. Res. 2019, 11, 6275. [Google Scholar]
- Jevotovsky, D.S.; Alfonso, A.R.; Einhorn, T.A.; Chiu, E.S. Osteoarthritis and stem cell therapy in humans: A systematic review. Osteoarthr. Cartil. 2018, 26, 711–729. [Google Scholar] [CrossRef] [Green Version]
- Tan, S.S.H.; Tjio, C.K.E.; Wong, J.R.Y.; Wong, K.L.; Chew, J.R.J.; Hui, J.H.P.; Toh, W.S. Mesenchymal stem cell exosomes for cartilage regeneration: A systematic review of preclinical in vivo studies. Tissue Eng. Part B Rev. 2021, 27, 1–13. [Google Scholar] [CrossRef]
- Barry, F. MSC Therapy for Osteoarthritis: An Unfinished Story. J. Orthop Res. 2019, 37, 1229–1235. [Google Scholar] [CrossRef]
- Kluzek, S.; Mattei, T.A. Machine-learning for osteoarthritis research. Osteoarthr. Cartil. 2019, 27, 977–978. [Google Scholar] [CrossRef] [PubMed]
- Fisher, C.K.; Smith, A.M.; Walsh, J.R.; Simon, A.J.; Edgar, C.; Jack, C.R.; Holtzman, D.; Russell, D.; Hill, D.; Grosset, D.; et al. Machine learning for comprehensive forecasting of Alzheimer’s Disease progression. Sci. Rep. 2019, 9, 13622. [Google Scholar] [CrossRef] [Green Version]
- Thery, 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] [PubMed] [Green Version]
- Zhang, Y.; Bi, J.; Huang, J.; Tang, Y.; Du, S.; Li, P. Exosome: A Review of Its Classification, Isolation Techniques, Storage, Diagnostic and Targeted Therapy Applications. Int. J. Nanomed. 2020, 15, 6917–6934. [Google Scholar] [CrossRef]
- Tan, K.L.; Chia, W.C.; How, C.W.; Tor, Y.S.; Show, P.L.; Looi, Q.H.D.; Foo, J.B. Benchtop Isolation and Characterisation of Small Extracellular Vesicles from Human Mesenchymal Stem Cells. Mol. Biotechnol. 2021, 63, 780–791. [Google Scholar] [CrossRef] [PubMed]
- Tschuschke, M.; Kocherova, I.; Bryja, A.; Mozdziak, P.; Angelova Volponi, A.; Janowicz, K.; Sibiak, R.; Piotrowska-Kempisty, H.; Iżycki, D.; Bukowska, D. Inclusion biogenesis, methods of isolation and clinical application of human cellular exosomes. J. Clin. Med. 2020, 9, 436. [Google Scholar] [CrossRef] [Green Version]
- Lorenc, T.; Chrzanowski, J.; Olejarz, W. Current Perspectives on Clinical Use of Exosomes as a Personalized Contrast Media and Theranostics. Cancers 2020, 12, 3386. [Google Scholar] [CrossRef]
- Katzmann, D.J.; Babst, M.; Emr, S.D. Ubiquitin-dependent sorting into the multivesicular body pathway requires the function of a conserved endosomal protein sorting complex, ESCRT-I. Cell 2001, 106, 145–155. [Google Scholar] [CrossRef] [Green Version]
- Wei, D.; Zhan, W.; Gao, Y.; Huang, L.; Gong, R.; Wang, W.; Zhang, R.; Wu, Y.; Gao, S.; Kang, T. RAB31 marks and controls an ESCRT-independent exosome pathway. Cell Res. 2021, 31, 157–177. [Google Scholar] [CrossRef]
- Hong, P.; Yang, H.; Wu, Y.; Li, K.; Tang, Z. The functions and clinical application potential of exosomes derived from adipose mesenchymal stem cells: A comprehensive review. Stem Cell Res. Ther. 2019, 10, 242. [Google Scholar] [CrossRef] [PubMed]
- Avalos-Padilla, Y.; Georgiev, V.N.; Lantero, E.; Pujals, S.; Verhoef, R.N.; Borgheti-Cardoso, L.; Albertazzi, L.; Dimova, R.; Fernandez-Busquets, X. The ESCRT-III machinery participates in the production of extracellular vesicles and protein export during Plasmodium falciparum infection. PLoS Pathog. 2021, 17, e1009455. [Google Scholar] [CrossRef]
- Morita, E.; Colf, L.A.; Karren, M.A.; Sandrin, V.; Rodesch, C.K.; Sundquist, W.I. Human ESCRT-III and VPS4 proteins are required for centrosome and spindle maintenance. Proc. Natl. Acad. Sci. USA 2010, 107, 12889–12894. [Google Scholar] [CrossRef] [Green Version]
- Tang, S.; Buchkovich, N.J.; Henne, W.M.; Banjade, S.; Kim, Y.J.; Emr, S.D. ESCRT-III activation by parallel action of ESCRT-I/II and ESCRT-0/Bro1 during MVB biogenesis. eLife 2016, 5, e15507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bryl, R.; Borowiec, B.; Siroma, R.S.; Pinto, N.; Melo, M.A.; Shibli, J.A.; Dyszkiewicz-Konwińska, M. Current application of exosomes in medicine. Med. J. Cell Biol. 2020, 8, 101–111. [Google Scholar] [CrossRef]
- Lee, I.J.; Stokasimov, E.; Dempsey, N.; Varberg, J.M.; Jacob, E.; Jaspersen, S.L.; Pellman, D. Factors promoting nuclear envelope assembly independent of the canonical ESCRT pathway. J. Cell Biol. 2020, 219, e201908232. [Google Scholar] [CrossRef]
- Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 2019, 9, 1–18. [Google Scholar] [CrossRef]
- Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brugger, B.; Simons, M. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 2008, 319, 1244–1247. [Google Scholar] [CrossRef] [PubMed]
- Wei, H.; Chen, Q.; Lin, L.; Sha, C.; Li, T.; Liu, Y.; Yin, X.; Xu, Y.; Chen, L.; Gao, W. Regulation of exosome production and cargo sorting. Int. J. Biol. Sci. 2021, 17, 163. [Google Scholar] [CrossRef]
- Sorop, A.; Constantinescu, D.; Cojocaru, F.; Dinischiotu, A.; Cucu, D.; Dima, S.O. Exosomal microRNAs as Biomarkers and Therapeutic Targets for Hepatocellular Carcinoma. Int. J. Mol. Sci. 2021, 22, 4997. [Google Scholar] [CrossRef]
- Munir, J.; Yoon, J.K.; Ryu, S. Therapeutic miRNA-Enriched Extracellular Vesicles: Current Approaches and Future Prospects. Cells 2020, 9, 2271. [Google Scholar] [CrossRef]
- Heijnen, H.F.G.; Schiel, A.E.; Fijnheer, R.; Geuze, H.J.; Sixma, J.J. Activated Platelets Release Two Types of Membrane Vesicles: Microvesicles by Surface Shedding and Exosomes Derived From Exocytosis of Multivesicular Bodies and α-Granules. Blood 1999, 94, 3791–3799. [Google Scholar] [CrossRef]
- Shenoda, B.B.; Ajit, S.K. Modulation of immune responses by exosomes derived from antigen-presenting cells. Clin. Med. Insights Pathol. 2016, 9, CPath-S39925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ha, D.; Yang, N.; Nadithe, V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: Current perspectives and future challenges. Acta Pharm. Sin. B 2016, 6, 287–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Didiot, M.-C.; Hall, L.M.; Coles, A.H.; Haraszti, R.A.; Godinho, B.M.; Chase, K.; Sapp, E.; Ly, S.; Alterman, J.F.; Hassler, M.R. Exosome-mediated delivery of hydrophobically modified siRNA for huntingtin mRNA silencing. Mol. Ther. 2016, 24, 1836–1847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wahlgren, J.; Karlson, T.D.L.; Brisslert, M.; Vaziri Sani, F.; Telemo, E.; Sunnerhagen, P.; Valadi, H. Plasma exosomes can deliver exogenous short interfering RNA to monocytes and lymphocytes. Nucleic Acids Res. 2012, 40, e130. [Google Scholar] [CrossRef] [Green Version]
- Shimbo, K.; Miyaki, S.; Ishitobi, H.; Kato, Y.; Kubo, T.; Shimose, S.; Ochi, M. Exosome-formed synthetic microRNA-143 is transferred to osteosarcoma cells and inhibits their migration. Biochem. Biophys. Res. Commun. 2014, 445, 381–387. [Google Scholar] [CrossRef] [PubMed]
- Branam, G.M.; Saber, A.Y. Osteochondral Autograft Transplantation; StatPearls, 2021. Available online: https://www.ncbi.nlm.nih.gov/books/NBK560655/ (accessed on 25 October 2021).
- Chen, Y.; Xue, K.; Zhang, X.; Zheng, Z.; Liu, K. Exosomes derived from mature chondrocytes facilitate subcutaneous stable ectopic chondrogenesis of cartilage progenitor cells. Stem Cell Res. Ther. 2018, 9, 1–14. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Ma, Y.; Zhang, J.; Yuan, Y.; Wang, J. Exosomes: A novel therapeutic agent for cartilage and bone tissue regeneration. Dose-Response 2019, 17, 1559325819892702. [Google Scholar] [CrossRef] [PubMed]
- Jiang, S.; Tian, G.; Yang, Z.; Gao, X.; Wang, F.; Li, J.; Tian, Z.; Huang, B.; Wei, F.; Sang, X.; et al. Enhancement of acellular cartilage matrix scaffold by Wharton’s jelly mesenchymal stem cell-derived exosomes to promote osteochondral regeneration. Bioact. Mater. 2021, 6, 2711–2728. [Google Scholar] [CrossRef]
- Zhang, S.; Chu, W.C.; Lai, R.C.; Lim, S.K.; Hui, J.H.; Toh, W.S. Exosomes derived from human embryonic mesenchymal stem cells promote osteochondral regeneration. Osteoarthr. Cartil. 2016, 24, 2135–2140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Vonk, L.A.; van Dooremalen, S.F.; Liv, N.; Klumperman, J.; Coffer, P.J.; Saris, D.B.; Lorenowicz, M.J. Mesenchymal stromal/stem cell-derived extracellular vesicles promote human cartilage regeneration in vitro. Theranostics 2018, 8, 906. [Google Scholar] [CrossRef]
- Lian, C.; Wang, X.; Qiu, X.; Wu, Z.; Gao, B.; Liu, L.; Liang, G.; Zhou, H.; Yang, X.; Peng, Y.; et al. Collagen type II suppresses articular chondrocyte hypertrophy and osteoarthritis progression by promoting integrin beta1-SMAD1 interaction. Bone Res. 2019, 7, 8. [Google Scholar] [CrossRef] [Green Version]
- 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, 1–12. [Google Scholar] [CrossRef]
- Zhang, S.; Chuah, S.J.; Lai, R.C.; Hui, J.H.P.; Lim, S.K.; Toh, W.S. MSC exosomes mediate cartilage repair by enhancing proliferation, attenuating apoptosis and modulating immune reactivity. Biomaterials 2018, 156, 16–27. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Pizzute, T.; Pei, M. Anti-inflammatory strategies in cartilage repair. Tissue Eng. Part B Rev. 2014, 20, 655–668. [Google Scholar] [CrossRef] [Green Version]
- Tofiño-Vian, M.; Guillén, M.I.; Del Caz, M.D.P.; Silvestre, A.; Alcaraz, M.J. Microvesicles from human adipose tissue-derived mesenchymal stem cells as a new protective strategy in osteoarthritic chondrocytes. Cell. Physiol. Biochem. 2018, 47, 11–25. [Google Scholar] [CrossRef]
- Ni, Z.; Zhou, S.; Li, S.; Kuang, L.; Chen, H.; Luo, X.; Ouyang, J.; He, M.; Du, X.; Chen, L. Exosomes: Roles and therapeutic potential in osteoarthritis. Bone Res. 2020, 8, 1–18. [Google Scholar] [CrossRef]
- 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, 165–178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, S.; Teo, K.Y.W.; Chuah, S.J.; Lai, R.C.; Lim, S.K.; Toh, W.S. MSC exosomes alleviate temporomandibular joint osteoarthritis by attenuating inflammation and restoring matrix homeostasis. Biomaterials 2019, 200, 35–47. [Google Scholar] [CrossRef] [PubMed]
- Lai, R.C.; Arslan, F.; Lee, M.M.; Sze, N.S.K.; Choo, A.; Chen, T.S.; Salto-Tellez, M.; Timmers, L.; Lee, C.N.; El Oakley, R.M. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010, 4, 214–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yin, K.; Wang, S.; Zhao, R.C. Exosomes from mesenchymal stem/stromal cells: A new therapeutic paradigm. Biomark. Res. 2019, 7, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Toh, W.S.; Lai, R.C.; Zhang, B.; Lim, S.K. MSC exosome works through a protein-based mechanism of action. Biochem. Soc. Trans. 2018, 46, 843–853. [Google Scholar] [CrossRef] [Green Version]
- Lai, R.C.; Tan, S.S.; Teh, B.J.; Sze, S.K.; Arslan, F.; De Kleijn, D.P.; Choo, A.; Lim, S.K. Proteolytic potential of the MSC exosome proteome: Implications for an exosome-mediated delivery of therapeutic proteasome. Int. J. Proteom. 2012, 2012, 971907. [Google Scholar] [CrossRef] [Green Version]
- Anderson, J.D.; Johansson, H.J.; Graham, C.S.; Vesterlund, M.; Pham, M.T.; Bramlett, C.S.; Montgomery, E.N.; Mellema, M.S.; Bardini, R.L.; Contreras, Z. Comprehensive proteomic analysis of mesenchymal stem cell exosomes reveals modulation of angiogenesis via nuclear factor-kappaB signaling. Stem Cells 2016, 34, 601–613. [Google Scholar] [CrossRef] [Green Version]
- Collino, F.; Pomatto, M.; Bruno, S.; Lindoso, R.S.; Tapparo, M.; Sicheng, W.; Quesenberry, P.; Camussi, G. Exosome and microvesicle-enriched fractions isolated from mesenchymal stem cells by gradient separation showed different molecular signatures and functions on renal tubular epithelial cells. Stem Cell Rev. 2017, 13, 226. [Google Scholar] [CrossRef] [Green Version]
- Turchinovich, A.; Drapkina, O.; Tonevitsky, A. Transcriptome of Extracellular Vesicles: State-of-the-Art. Front. Immunol. 2019, 10, 202. [Google Scholar] [CrossRef] [Green Version]
- Chen, T.; Lai, R.; Lee, M.; Choo, A.; Lee, C.; Lim, S. Mesenchymal stem cell secretes microparticles enriched in pre-microRNAs. Nucleic Acids Res. 2010, 38, 215–224. [Google Scholar] [CrossRef] [Green Version]
- Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.; Lötvall, J. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Smalheiser, N. Exosomal transfer of proteins and RNAs at synapses in the nervous system. Biol. Direct 2007, 2, 1–15. [Google Scholar] [CrossRef] [Green Version]
- Fu, G.; Brkić, J.; Hayder, H.; Peng, C. MicroRNAs in Human Placental Development and Pregnancy Complications. Int. J. Mol. Sci. 2013, 14, 5519–5544. [Google Scholar] [CrossRef] [Green Version]
- Paul, P.; Chakraborty, A.; Sarkar, D.; Langthasa, M.; Rahman, M.; Bari, M.; Singha, R.K.S.; Malakar, A.K.; Chakraborty, S. Interplay between miRNAs and human diseases. J. Cell. Physiol. 2018, 233, 2007–2018. [Google Scholar] [CrossRef] [PubMed]
- Pitt, J.M.; Kroemer, G.; Zitvogel, L. Extracellular vesicles: Masters of intercellular communication and potential clinical interventions. J. Clin. Investig. 2016, 126, 1139–1143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, T.; Gu, H.; Chen, X.; Fu, S.; Wang, C.; Xu, H.; Feng, Q.; Ni, Y. Cardiac hypertrophy and dysfunction induced by overexpression of miR-214 in vivo. J. Surg. Res. 2014, 192, 317–325. [Google Scholar] [CrossRef]
- Wang, J.; Chen, J.; Sen, S. MicroRNA as Biomarkers and Diagnostics. J. Cell. Physiol. 2016, 231, 25–30. [Google Scholar] [CrossRef] [PubMed]
- Alvarez-Garcia, I.; Miska, E. MicroRNA functions in animal development and human disease. Development 2005, 132, 4653–4662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Krek, A.; Grün, D.; Poy, M.; Wolf, R.; Rosenberg, L.; Epstein, E.; MacMenamin, P.; da Piedade, I.; Gunsalus, K.; Stoffel, M.; et al. Combinatorial microRNA target predictions. Nat. Genet. 2005, 37, 495–500. [Google Scholar] [CrossRef] [PubMed]
- Berezikov, E.; van Tetering, G.; Verheul, M.; van de Belt, J.; van Laake, L.; Vos, J.; Verloop, R.; van de Wetering, M.; Guryev, V.; Takada, S.; et al. Many novel mammalian microRNA candidates identified by extensive cloning and RAKE analysis. Genome Res. 2006, 16, 1289–1298. [Google Scholar] [CrossRef] [Green Version]
- Siomi, H.; Siomi, M.C. On the road to reading the RNA-interference code. Nature 2009, 457, 396–404. [Google Scholar] [CrossRef]
- Huntzinger, E.; Izaurralde, E. Gene silencing by microRNAs: Contributions of translational repression and mRNA decay. Nat. Rev. Genet. 2011, 12, 99–110. [Google Scholar] [CrossRef] [PubMed]
- Ipsaro, J.; Joshua-Tor, L. From guide to target: Molecular insights into eukaryotic RNA-interference machinery. Nat. Struct. Mol. Biol. 2015, 22, 20–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, W.; San Lucas, A.; Wang, Z.; Liu, Y. Identifying microRNA targets in different gene regions. BMC Bioinform. 2014, 15 (Suppl. 7), S4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Steitz, J.; Vasudevan, S. miRNPs: Versatile regulators of gene expression in vertebrate cells. Biochem. Soc. Trans. 2009, 37, 931–935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cai, Y.; Yu, X.; Hu, S.; Yu, J. A brief review on the mechanisms of miRNA regulation. Genom. Proteom. Bioinform. 2009, 7, 147–154. [Google Scholar] [CrossRef] [Green Version]
- Lewis, B.; Burge, C.; Bartel, D. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005, 120, 15–20. [Google Scholar] [CrossRef] [Green Version]
- Carroll, A.; Goodall, G.; Liu, B. Understanding principles of miRNA target recognition and function through integrated biological and bioinformatics approaches. Wiley Interdiscip. Rev. RNA 2014, 5, 361–379. [Google Scholar] [CrossRef]
- Wang, X. Composition of seed sequence is a major determinant of microRNA targeting patterns. Bioinformatics 2014, 30, 1377–1383. [Google Scholar] [CrossRef] [Green Version]
- Brodersen, P.; Voinnet, O. Revisiting the principles of microRNA target recognition and mode of action. Nat. Rev. Mol. Cell Biol. 2009, 10, 141–148. [Google Scholar] [CrossRef] [PubMed]
- Ham, O.; Song, B.; Lee, S.; Choi, E.; Cha, M.; Lee, C.; Park, J.; Kim, L.; Chang, W.; Lim, S.; et al. The role of microRNA-23b in the differentiation of MSC into chondrocyte by targeting protein kinase A signaling. Biomaterials 2012, 33, 4500–4507. [Google Scholar] [CrossRef] [PubMed]
- Ning, G.; Liu, X.; Dai, M.; Meng, A.; Wang, Q. MicroRNA-92a upholds Bmp signaling by targeting noggin3 during pharyngeal cartilage formation. Dev. Cell 2013, 24, 283–295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hou, C.; Zhang, Z.; Zhang, Z.; Wu, P.; Zhao, X.; Fu, M.; Sheng, P.; Kang, Y.; Liao, W. Presence and function of microRNA-92a in chondrogenic ATDC5 and adipose-derived mesenchymal stem cells. Mol. Med. Rep. 2015, 12, 4877–4886. [Google Scholar] [CrossRef] [Green Version]
- Matsukawa, T.; Sakai, T.; Yonezawa, T.; Hiraiwa, H.; Hamada, T.; Nakashima, M.; Ono, Y.; Ishizuka, S.; Nakahara, H.; Lotz, M.K.; et al. MicroRNA-125b regulates the expression of aggrecanase-1 (ADAMTS-4) in human osteoarthritic chondrocytes. Arthrit. Res. Ther. 2013, 15, R28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Meng, F.; Zhang, Z.; Chen, W.; Huang, G.; He, A.; Hou, C.; Long, Y.; Yang, Z.; Zhang, Z.; Liao, W. MicroRNA-320 regulates matrix metalloproteinase-13 expression in chondrogenesis and interleukin-1β-induced chondrocyte responses. Osteoarthr. Cartil. 2016, 24, 932–941. [Google Scholar] [CrossRef]
- Yang, B.; Guo, H.; Zhang, Y.; Chen, L.; Ying, D.; Dong, S. MicroRNA-145 regulates chondrogenic differentiation of mesenchymal stem cells by targeting Sox9. PLoS ONE 2011, 6, e21679. [Google Scholar] [CrossRef]
- Kim, D.; Song, J.; Jin, E. MicroRNA-221 regulates chondrogenic differentiation through promoting proteosomal degradation of slug by targeting Mdm2. J. Biol. Chem. 2010, 285, 26900–26907. [Google Scholar] [CrossRef] [Green Version]
- Iliopoulos, D.; Malizos, K.; Oikonomou, P.; Tsezou, A. Integrative microRNA and proteomic approaches identify novel osteoarthritis genes and their collaborative metabolic and inflammatory networks. PLoS ONE 2008, 3, e3740. [Google Scholar] [CrossRef]
- Mao, G.; Zhang, Z.; Hu, S.; Zhang, Z.; Chang, Z.; Huang, Z.; Liao, W.; Kang, Y. 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, 247. [Google Scholar] [CrossRef] [Green Version]
- Wang, R.; Xu, B.; Xu, H. TGF-β1 promoted chondrocyte proliferation by regulating Sp1 through MSC-exosomes derived miR-135b. Cell Cycle 2018, 17, 2756–2765. [Google Scholar] [CrossRef] [Green Version]
- Wu, J.; Kuang, L.; Chen, C.; Yang, J.; Zeng, W.; Li, T.; Chen, H.; Huang, S.; Fu, Z.; Li, J.; 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. [Google Scholar] [CrossRef]
- Tao, S.-C.; Yuan, T.; Zhang, Y.-L.; Yin, W.-J.; Guo, S.-C.; Zhang, C.-Q. 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, 180. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- 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. Arthrit. Res. Ther. 2020, 22, 1–13. [Google Scholar] [CrossRef]
- Dong, J.; Li, L.; Fang, X.; Zang, M. Exosome-Encapsulated microRNA-127-3p Released from Bone Marrow-Derived Mesenchymal Stem Cells Alleviates Osteoarthritis Through Regulating CDH11-Mediated Wnt/β-Catenin Pathway. J. Pain Res. 2021, 14, 297–310. [Google Scholar] [CrossRef] [PubMed]
- 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, 99–114. [Google Scholar] [CrossRef] [PubMed]
- Chong, Z.X.; Yeap, S.K.; Ho, W.Y. Transfection types, methods and strategies: A technical review. PeerJ 2021, 9, e11165. [Google Scholar] [CrossRef] [PubMed]
- Kim, T.K.; Eberwine, J.H. Mammalian cell transfection: The present and the future. Anal. Bioanal. Chem. 2010, 397, 3173–3178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Z.; Yan, K.; Ge, G.; Zhang, D.; Bai, J.; Guo, X.; Zhou, J.; Xu, T.; Xu, M.; Long, X.; 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, 85–96. [Google Scholar] [CrossRef] [PubMed]
- Chen, T.S.; Arslan, F.; Yin, Y.; Tan, S.S.; Lai, R.C.; Choo, A.B.; Padmanabhan, J.; Lee, C.N.; de Kleijn, D.P.; Lim, S.K. Enabling a robust scalable manufacturing process for therapeutic exosomes through oncogenic immortalization of human ESC-derived MSCs. J. Transl. Med. 2011, 9, 47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lai, R.C.; Yeo, R.W.; Padmanabhan, J.; Choo, A.; de Kleijn, D.P.; Lim, S.K. Isolation and Characterization of Exosome from Human Embryonic Stem Cell-Derived C-Myc-Immortalized Mesenchymal Stem Cells. Methods Mol. Biol. 2016, 1416, 477–494. [Google Scholar] [PubMed]
- Statello, L.; Maugeri, M.; Garre, E.; Nawaz, M.; Wahlgren, J.; Papadimitriou, A.; Lundqvist, C.; Lindfors, L.; Collén, A.; Sunnerhagen, P.; et al. Identification of RNA-binding proteins in exosomes capable of interacting with different types of RNA: RBP-facilitated transport of RNAs into exosomes. PLoS ONE 2018, 13, e0195969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Villarroya-Beltri, C.; Gutiérrez-Vázquez, C.; Sánchez-Cabo, F.; Pérez-Hernández, D.; Vázquez, J.; Martin-Cofreces, N.; Martinez-Herrera, D.J.; Pascual-Montano, A.; Mittelbrunn, M.; Sánchez-Madrid, F. Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nat. Commun. 2013, 4, 2980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Santangelo, L.; Giurato, G.; Cicchini, C.; Montaldo, C.; Mancone, C.; Tarallo, R.; Battistelli, C.; Alonzi, T.; Weisz, A.; Tripodi, M. The RNA-Binding Protein SYNCRIP Is a Component of the Hepatocyte Exosomal Machinery Controlling MicroRNA Sorting. Cell Rep. 2016, 17, 799–808. [Google Scholar] [CrossRef] [Green Version]
- Shurtleff, M.J.; Temoche-Diaz, M.M.; Karfilis, K.V.; Ri, S.; Schekman, R. Y-box protein 1 is required to sort microRNAs into exosomes in cells and in a cell-free reaction. eLife 2016, 5, e19276. [Google Scholar] [CrossRef] [PubMed]
- Lee, H.; Li, C.; Zhang, Y.; Zhang, D.; Otterbein, L.E.; Jin, Y. Caveolin-1 selectively regulates microRNA sorting into microvesicles after noxious stimuli. J. Exp. Med. 2019, 216, 2202–2220. [Google Scholar] [CrossRef]
- Bae, H.C.; Park, H.J.; Wang, S.Y.; Yang, H.R.; Lee, M.C.; Han, H.-S. Hypoxic condition enhances chondrogenesis in synovium-derived mesenchymal stem cells. Biomater. Res. 2018, 22, 28. [Google Scholar] [CrossRef] [Green Version]
- Koay, E.J.; Athanasiou, K.A. Hypoxic chondrogenic differentiation of human embryonic stem cells enhances cartilage protein synthesis and biomechanical functionality. Osteoarthr. Cartil. 2008, 16, 1450–1456. [Google Scholar] [CrossRef] [Green Version]
- Pei, M. Environmental preconditioning rejuvenates adult stem cells’ proliferation and chondrogenic potential. Biomaterials 2017, 117, 10–23. [Google Scholar] [CrossRef] [Green Version]
- Cassano, J.M.; Schnabel, L.V.; Goodale, M.B.; Fortier, L.A. Inflammatory licensed equine MSCs are chondroprotective and exhibit enhanced immunomodulation in an inflammatory environment. Stem Cell Res. Ther. 2018, 9, 82. [Google Scholar] [CrossRef] [Green Version]
- Baudry, N.; Starck, J.; Aussel, C.; Lund, K.; Aletti, M.; Duranteau, J.; Banzet, S.; Lataillade, J.J.; Vicaut, E.; Peltzer, J. Effect of Preconditioned Mesenchymal Stromal Cells on Early Microvascular Disturbance in a Mouse Sepsis Model. Stem Cells Dev. 2019, 28, 1595–1606. [Google Scholar] [CrossRef] [PubMed]
- Magne, B.; Dedier, M.; Nivet, M.; Coulomb, B.; Banzet, S.; Lataillade, J.J.; Trouillas, M. IL-1β-Primed Mesenchymal Stromal Cells Improve Epidermal Substitute Engraftment and Wound Healing via Matrix Metalloproteinases and Transforming Growth Factor-β1. J. Investig. Derm. 2020, 140, 688–698.e621. [Google Scholar] [CrossRef]
- Song, S.W.; Kim, K.E.; Choi, J.W.; Lee, C.Y.; Lee, J.; Seo, H.H.; Lim, K.H.; Lim, S.; Lee, S.; Kim, S.W.; et al. Proteomic Analysis and Identification of Paracrine Factors in Mesenchymal Stem Cell-Conditioned Media under Hypoxia. Cell. Physiol. Biochem. 2016, 40, 400–410. [Google Scholar] [CrossRef] [Green Version]
- Paquet, J.; Deschepper, M.; Moya, A.; Logeart-Avramoglou, D.; Boisson-Vidal, C.; Petite, H. Oxygen Tension Regulates Human Mesenchymal Stem Cell Paracrine Functions. Stem Cells Transl. Med. 2015, 4, 809–821. [Google Scholar] [CrossRef]
- Zhang, Q.; Fu, L.; Liang, Y.; Guo, Z.; Wang, L.; Ma, C.; Wang, H. Exosomes originating from MSCs stimulated with TGF-β and IFN-γ promote Treg differentiation. J. Cell. Physiol. 2018, 233, 6832–6840. [Google Scholar] [CrossRef] [PubMed]
- Peltzer, J.; Lund, K.; Goriot, M.-E.; Grosbot, M.; Lataillade, J.-J.; Mauduit, P.; Banzet, S. Interferon-γ and Hypoxia Priming Have Limited Effect on the miRNA Landscape of Human Mesenchymal Stromal Cells-Derived Extracellular Vesicles. Front. Cell Dev. Biol. 2020, 8, 1434. [Google Scholar] [CrossRef] [PubMed]
- Marinaro, F.; Gómez-Serrano, M.; Jorge, I.; Silla-Castro, J.C.; Vázquez, J.; Sánchez-Margallo, F.M.; Blázquez, R.; López, E.; Álvarez, V.; Casado, J.G. Unraveling the Molecular Signature of Extracellular Vesicles From Endometrial-Derived Mesenchymal Stem Cells: Potential Modulatory Effects and Therapeutic Applications. Front. Bioeng. Biotechnol. 2019, 7, 431. [Google Scholar] [CrossRef] [Green Version]
- He, X.; Deng, L. Potential of miR-25-3p in protection of chondrocytes: Emphasis on osteoarthritis. Folia Histochem. Cytobiol. 2021, 59, 30–39. [Google Scholar] [CrossRef]
- Zhang, X.; Liu, X.; Ni, X.; Feng, P.; Wang, Y.U. Long non-coding RNA H19 modulates proliferation and apoptosis in osteoarthritis via regulating miR-106a-5p. J. Biosci. 2019, 44, 128. [Google Scholar] [CrossRef]
- Zhou, Y.; Ming, J.; Li, Y.; Li, B.; Deng, M.; Ma, Y.; Chen, Z.; Zhang, Y.; Li, J.; Liu, S. Exosomes derived from miR-126-3p-overexpressing synovial fibroblasts suppress chondrocyte inflammation and cartilage degradation in a rat model of osteoarthritis. Cell Death Discov. 2021, 7, 37. [Google Scholar] [CrossRef] [PubMed]
- Wang, T.; Hao, Z.; Liu, C.; Yuan, L.; Li, L.; Yin, M.; Li, Q.; Qi, Z.; Wang, Z. LEF1 mediates osteoarthritis progression through circRNF121/miR-665/MYD88 axis via NF-кB signaling pathway. Cell Death Dis. 2020, 11, 598. [Google Scholar] [CrossRef] [PubMed]
- Endisha, H.; Datta, P.; Sharma, A.; Nakamura, S.; Rossomacha, E.; Younan, C.; Ali, S.A.; Tavallaee, G.; Lively, S.; Potla, P.; et al. MicroRNA-34a-5p Promotes Joint Destruction During Osteoarthritis. Arthritis Rheumatol. 2021, 73, 426–439. [Google Scholar] [CrossRef] [PubMed]
- Pomatto, M.A.C.; Bussolati, B.; D’Antico, S.; Ghiotto, S.; Tetta, C.; Brizzi, M.F.; Camussi, G. Improved Loading of Plasma-Derived Extracellular Vesicles to Encapsulate Antitumor miRNAs. Mol. Ther. Methods Clin. Dev. 2019, 13, 133–144. [Google Scholar] [CrossRef] [Green Version]
- Zhang, D.; Lee, H.; Zhu, Z.; Minhas, J.K.; Jin, Y. Enrichment of selective miRNAs in exosomes and delivery of exosomal miRNAs in vitro and in vivo. Am. J. Physiol. Lung Cell Mol. Physiol. 2017, 312, L110–L121. [Google Scholar] [CrossRef] [PubMed]
- Johnsen, K.B.; Gudbergsson, J.M.; Skov, M.N.; Christiansen, G.; Gurevich, L.; Moos, T.; Duroux, M. Evaluation of electroporation-induced adverse effects on adipose-derived stem cell exosomes. Cytotechnology 2016, 68, 2125–2138. [Google Scholar] [CrossRef] [Green Version]
- Rezaie, H.R.; Esnaashary, M.; Öchsner, A. The History of Drug Delivery Systems. In A Review of Biomaterials and Their Applications in Drug Delivery; Springer: Berlin/Heidelberg, Germany, 2018; pp. 1–8. [Google Scholar]
- Fang, W.H.; Vangsness, C.T., Jr. Food and Drug Administration’s Position on Commonly Injected Biologic Materials in Orthopaedic Surgery. Am. J. Sports Med. 2021, 49, 3414–3421. [Google Scholar] [CrossRef] [PubMed]
- Gupta, A.; Maffulli, N.; Rodriguez, H.C.; Carson, E.W.; Bascharon, R.A.; Delfino, K.; Levy, H.J.; El-Amin, S.F. Safety and efficacy of umbilical cord-derived Wharton’s jelly compared to hyaluronic acid and saline for knee osteoarthritis: Study protocol for a randomized, controlled, single-blind, multi-center trial. J. Orthop. Surg. Res. 2021, 16, 1–8. [Google Scholar]
- Gupta, A.; El-Amin, S.F.; Levy, H.J.; Sze-Tu, R.; Ibim, S.E.; Maffulli, N. Umbilical cord-derived Wharton’s jelly for regenerative medicine applications. J. Orthop. Surg. Res. 2020, 15, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Tao, S.-C.; Huang, J.-Y.; Gao, Y.; Li, Z.-X.; Wei, Z.-Y.; Dawes, H.; Guo, S.-C. Small extracellular vesicles in combination with sleep-related circRNA3503: A targeted therapeutic agent with injectable thermosensitive hydrogel to prevent osteoarthritis. Bioact. Mater. 2021, 6, 4455–4469. [Google Scholar] [CrossRef]
- Thomas, B.L.; Eldridge, S.E.; Nosrati, B.; Alvarez, M.; Thorup, A.S.; Nalesso, G.; Caxaria, S.; Barawi, A.; Nicholson, J.G.; Perretti, M. WNT3A-loaded exosomes enable cartilage repair. J. Extracell. Vesicles 2021, 10, e12088. [Google Scholar] [CrossRef] [PubMed]
- Wong, K.L.; Zhang, S.; Wang, M.; Ren, X.; Afizah, H.; Lai, R.C.; Lim, S.K.; Lee, E.H.; Hui, J.H.P.; Toh, W.S. Intra-articular injections of mesenchymal stem cell exosomes and hyaluronic acid improve structural and mechanical properties of repaired cartilage in a rabbit model. Arthrosc. J. Arthrosc. Relat. Surg. 2020, 36, 2215–2228.e2212. [Google Scholar] [CrossRef]
- Herrmann, M.; Diederichs, S.; Melnik, S.; Riegger, J.; Trivanović, D.; Li, S.; Jenei-Lanzl, Z.; Brenner, R.E.; Huber-Lang, M.; Zaucke, F. Extracellular vesicles in musculoskeletal pathologies and regeneration. Front. Bioeng. Biotechnol. 2021, 8, 1509. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Shao, J.; Zhu, J.; Chen, Y.; Fu, Q.; Li, L.; Ding, Z.; Wu, J.; Han, Y.; Li, H.; Qian, Q. Exosomes from Kartogenin-Pretreated Infrapatellar Fat Pad Mesenchymal Stem Cells Enhance Chondrocyte Anabolism and Articular Cartilage Regeneration. Stem Cells Int. 2021, 2021, 6624874. [Google Scholar] [CrossRef]
- Zhu, Y.; Wang, Y.; Zhao, B.; Niu, X.; Hu, B.; Li, Q.; Zhang, J.; Ding, J.; Chen, Y.; Wang, Y. 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, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gimona, M.; Brizzi, M.F.; Choo, A.B.H.; Dominici, M.; Davidson, S.M.; Grillari, J.; Hermann, D.M.; Hill, A.F.; de Kleijn, D.; Lai, R.C.; et al. Critical considerations for the development of potency tests for therapeutic applications of mesenchymal stromal cell-derived small extracellular vesicles. Cytotherapy 2021, 23, 373–380. [Google Scholar] [CrossRef]
miRNA | Target | Physiological Role | Ref. |
---|---|---|---|
miR-23b | PKA | Induce chondrogenic differentiation of human MSCs by inhibiting PKA signaling | [143] |
miR-92a | Noggin3 | Targets Noggin3 and activates the PI3K/Akt/mTOR pathway to positively regulate the proliferation and matrix synthesis of chondroprogenitors | [144] [145] |
miR-125b | ADAMTS-4 | miR-125b overexpression suppresses IL-1-induced upregulation of ADAMTS-4 in human OA chondrocytes | [146] |
miR-320 | MMP-13 | Downregulates MMP-13 expression in both the ATDC5 cell model of chondrogenesis and IL-1-treated primary mouse chondrocytes | [147] |
miR-145 | Sox9 | miR-145 inhibition upregulates Sox9 expression and promotes MSC chondrogenesis | [148] |
miR-221 | MDM2 | Downregulates MDM2 to prevent slug protein degradation that, in turn, negatively regulates chondroprogenitor proliferation | [149] |
miR-22 | PPARA, BMP-7 | miR-22 inhibition upregulates BMP-7 and PPARA expression, inhibits IL-1 expression, and suppresses MMP-13 expression in OA chondrocytes | [150] |
miR-92a-3p | Wnt5a | Regulate cartilage development and homeostasis by targeting Wnt5a | [151] |
miR-135b | Sp1a | Promote chondrocyte proliferation and cartilage repair in OA by downregulating Sp1a in chondrocytes | [152] |
miR-100-5p | mTOR | Inhibit mTOR signaling pathway to enhance chondrocyte autophagy | [153] |
miR-140-5p | YAP | Enhance ECM secretion and induce proliferation and migration of articular chondrocytes via activating YAP as well as prevent osteoarthritic joint damage | [154] |
miR-26a-5p | PTGS2 | Promote the survival of synovial fibroblasts and reduce synovitis | [155] |
miR-136-5p | ELF3 | Inhibit cartilage degeneration in traumatic osteoarthritis | [156] |
miR-127-3p | CDH11-mediated Wnt/β-catenin pathway | Inhibit CDH11, thereby blocking the Wnt/β-catenin pathway in chondrocytes and reducing the chondrocyte damage in osteoarthritic joints | [157] |
miR-9-5p | Syndecan-1 | Has anti-inflammatory and cartilage protective effects on osteoarthritis | [158] |
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
Foo, J.B.; Looi, Q.H.; How, C.W.; Lee, S.H.; Al-Masawa, M.E.; Chong, P.P.; Law, J.X. Mesenchymal Stem Cell-Derived Exosomes and MicroRNAs in Cartilage Regeneration: Biogenesis, Efficacy, miRNA Enrichment and Delivery. Pharmaceuticals 2021, 14, 1093. https://doi.org/10.3390/ph14111093
Foo JB, Looi QH, How CW, Lee SH, Al-Masawa ME, Chong PP, Law JX. Mesenchymal Stem Cell-Derived Exosomes and MicroRNAs in Cartilage Regeneration: Biogenesis, Efficacy, miRNA Enrichment and Delivery. Pharmaceuticals. 2021; 14(11):1093. https://doi.org/10.3390/ph14111093
Chicago/Turabian StyleFoo, Jhi Biau, Qi Hao Looi, Chee Wun How, Sau Har Lee, Maimonah Eissa Al-Masawa, Pei Pei Chong, and Jia Xian Law. 2021. "Mesenchymal Stem Cell-Derived Exosomes and MicroRNAs in Cartilage Regeneration: Biogenesis, Efficacy, miRNA Enrichment and Delivery" Pharmaceuticals 14, no. 11: 1093. https://doi.org/10.3390/ph14111093
APA StyleFoo, J. B., Looi, Q. H., How, C. W., Lee, S. H., Al-Masawa, M. E., Chong, P. P., & Law, J. X. (2021). Mesenchymal Stem Cell-Derived Exosomes and MicroRNAs in Cartilage Regeneration: Biogenesis, Efficacy, miRNA Enrichment and Delivery. Pharmaceuticals, 14(11), 1093. https://doi.org/10.3390/ph14111093