Next Article in Journal
Pediatric Intensive Care Unit Length of Stay Prediction by Machine Learning
Previous Article in Journal
Galectin-3/Gelatin Electrospun Scaffolds Modulate Collagen Synthesis in Skin Healing but Do Not Improve Wound Closure Kinetics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Commentary

The Use of Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles in the Treatment of Osteoarthritis: Insights from Preclinical Studies

1
Department of Chemistry, Loughborough University, Loughborough LE11 3TU, UK
2
Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS2 9JT, UK
3
Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA
4
Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
*
Authors to whom correspondence should be addressed.
Bioengineering 2024, 11(10), 961; https://doi.org/10.3390/bioengineering11100961
Submission received: 21 August 2024 / Revised: 16 September 2024 / Accepted: 24 September 2024 / Published: 26 September 2024
(This article belongs to the Section Regenerative Engineering)

Abstract

:
Osteoarthritis (OA) is a prominent cause of disability, and has severe social and economic ramifications across the globe. The main driver of OA’s pervasiveness is the fact that no current medical interventions exist to reverse or even attenuate the degeneration of cartilage within the articular joint. Crucial for cell-to-cell communication, extracellular vesicles (EVs) contribute to OA progression through the delivery of bioactive molecules in the inflammatory microenvironment. By repurposing this acellular means of signal transmission, therapeutic drugs may be administered to degenerated cartilage tissue in the hopes of encouraging regeneration. Positive outcomes are apparent in in vivo studies on this subject; however, for this therapy to prove itself in the clinical world, efforts towards standardizing the characterization, application, biological contents, and dosage are essential.

Osteoarthritis (OA) affects approximately 500 million people worldwide and is a common cause of disability and societal economic burden [1]. It is a chronic degenerative disease of the joints characterized by an irrecoverable loss of articular cartilage, low-grade synovial inflammation, and bone hypertrophy, primarily in the knee, hip, hands, and spine [2]. OA has a complex etiology depending on the initial triggers and genetic and environmental risk factors. Multiple OA phenotype classifications have been proposed to account for disease heterogeneity. However, regardless of the phenotype, patients have similar clinical endpoints (pain, stiffness, loss of function), and a common activation of low-grade joint inflammatory/immune cascades [3,4,5]. Conventional drug therapies are capable of controlling pain; however, they have not been shown to have a sustained effect on the long-term mitigation of OA symptoms or disease progression and can have deleterious effects when prescribed over long durations [6]. With this in mind, research and clinical efforts have been directed towards the development of OA therapies that do not just alleviate symptoms but target the joint proinflammatory area early following the onset of degeneration.
Newer, regeneration-oriented interventions, including orthobiologic therapy with mesenchymal stem/stromal cells (MSCs) and their derivatives, such as extracellular vesicles (EVs), require standardization, stricter regulation, and further evidence of clinical efficacy [7,8,9,10]. Deemed “injury drugstores”, MSCs have been shown to have anti-inflammatory, antifibrotic, anabolic, and analgesic therapeutic activity, all of which could prove beneficial to patients with OA [11]. While numerous in vitro studies have shown MSC-EVs’ beneficial effects on cultured chondrocytes and immune lineage cells, preclinical evidence remains at an early, though crucial, stage for identifying the key steps towards clinical translation. In this commentary, we highlight the experimental techniques that have already become standard, such as the cargo and function characterization of MSC-derived EVs, as well as areas in which experimental approaches remain highly variable. These include animal models, EV tissue sources, doses, delivery methods, and treatment schedules. The benefits of delivering certain exogenous cargos, such as microRNAs, over native EVs, are yet to be explored. The utilization of animal models, representing different etiologies of human OA, would help to rationally design clinical trials targeting different groups of OA sufferers.
Extracellular vesicles, crucial for physiological cell-to-cell communication, are seen as a new frontier in OA therapeutics, owing to their acellular nature, small sizes, and ability to transport bioactive molecules. While MSCs do secrete cytokines and chemokines directly, they also exhibit their paracrine effects via EVs. According to the International Society of Extracellular Vesicles (ISEV) guidelines, EVs are defined as “particles that are released from cells, are delimited by a lipid bilayer, and cannot replicate on their own” [12]. Specifically, EVs serve as vehicles for cellular export products, including lipids, proteins, and RNAs (mRNAs and miRNAs), and can modulate the function of other cells at proximal or distal sites [13]. Specifically deployed ‘therapeutic’ MSC-EVs, derived from healthy tissues or purposefully designed to carry specific cargos, can deliver restorative compounds to the site of damage and, in relation to OA, may aid in joint regeneration. The proposed mechanisms of action of MSC-EVs on an OA environment include the following: the regulation of chondrocytes’ senescence and apoptosis, inducing favorable changes in their bioenergetic metabolism, the enhancement of their extracellular matrix production, and regulating the immune responses in the synovium [14,15,16].
EV therapy, although only recently gaining clinical interest, has several benefits over current cell-based regenerative approaches, such as platelet-rich plasma (PRP) or MSC therapies (Figure 1). Compared to an autologous PRP [17], EVs would be produced under highly controlled good manufacturing practice (GMP) conditions [18], where quality control (QC) standards already exist. The potential limitations of MSC therapy, such as the risk of immuno-rejection, tumorigenicity, and short in situ lifespans are overcome by utilizing acellular particles instead of living cells [19]. Furthermore, the ability to produce EVs from immortalized cell lines instead of primary cells also reduces batch-to-batch variability, aids in QC assessments, and reduces overall costs [20].
Thus far, numerous in vitro studies have succeeded in demonstrating the effectiveness of MSC-EVs in reducing OA-like chondrocyte apoptosis, pyroptosis, and the release of proinflammatory and catabolic molecules, as well as in increasing their viability, migration, and anabolic abilities [21,22,23,24,25]. Additionally, recent studies have shown the effectiveness of MSC-EVs in attenuating the immune lineage cells’ proinflammatory activation and pain signaling [26,27,28,29]. While the source and lineage of the host MSCs remain variable, the generation and characterization of the produced EVs normally followed the most recent ISEV guidelines [12], and the growing consensus revealed positive outcomes in the majority of in vitro studies [8,16,30,31]. Comprehensive studies of MSC-EVs cargos have identified the most promising candidate molecules for incorporating into ‘engineered EVs’, as discussed previously [16,26,32,33,34]. However, even in advanced three-dimensional in vitro culture models, complex biomechanical and pathophysiological joint environments cannot be fully reproduced [35], necessitating preclinical evaluations in animal models.
Reviews and meta-analysis studies have described the efficacy of intra-articularly (IA) injected MSC-EVs in a variety of OA animal models [15,16,35]. When it comes to the source of parental cells and animal species, the previously used approaches remain variable (Table 1). Following successful tests of EVs from the host animals’ MSCs (commonly BM-MSC-derived [36,37]), more recent studies have focused on human MSC-EVs, predominantly from the BM [23,33,38,39,40,41,42,43,44] and adipose tissues [25,28,29,45,46], but also from the synovium [47,48,49,50,51,52,53,54] or the umbilical cord [55,56]. According to these studies, the following specific cargos have been investigated and proven effective: miR-92a-3p, miR-136-5p, miR-3960, miR-125a-5p, miR-361-5p, LncRNA NEAT1, and lncRNA MEG-3 for BM-derived MSC-EVs; miR-100-5p for adipose tissue-derived MSC-EVs; miR-26a-5p, miR-155-5p, circRNA3503, miR-140-5p, and miR-31 for synovium-derived MSC-EVs; and LncRNA H19, miR-1208 and circHIPK3 for UC-MSC-EVs (Table 1). However, direct comparisons in the same study of the effectiveness of EVs or their cargos from different MSC tissue sources, for example, BM and adipose tissue, remain infrequent [57]. Although much attention has been given to nucleic acid-based cargos, few studies have investigated specific protein cargos, such as CD10 [28] and CD90 [47], and proven the effectiveness regarding the enhanced repair of damaged cartilage and their anti-inflammatory ability.
Small animals (mice and rats) represent the most commonly utilized species model, followed by rabbits [43,47] and occasionally, minipigs [62]. The doses of delivered MSC-EVs were commonly calculated as particle numbers or, less frequently, by weight, with some studies additionally reporting the numbers of parental MSCs [28,29,38]. Treatment schedules and follow-up periods differed considerably, depending on the animal model and the study’s unique protocol (Table 1). Some studies employed single IA injections; however, the majority of studies favored multiple injections at daily or weekly intervals depending on the animal model and how OA changes had been induced (enzymatically, chemically, or surgically). Control groups commonly comprised ‘sham’ (normally PBS, the same volume as EVs) and ‘OA’ (OA-induced but not treated) groups, while some studies additionally included healthy joints. Parental MSCs as a comparator group for the assessment of relative efficacy were used less frequently [38,45]. The most common follow-up periods were 4–6 weeks (for mice) and 4–8 weeks (for rats), although shorter periods were considered for post-traumatic OA models, such as oleanolic acid- or ACL- rupture-induced models (both in mice) [39,41].
As mentioned, IA injections were the most common method, but not the only method, of EV delivery. EV-loaded hydrogels are becoming an increasingly popular means of EV delivery, because while injectable, they can be combined with 3D bioprinting [63]. For example, a study conducted by Chen et al. used an infused hydrogel scaffold to restore mitochondrial function in dysfunctional chondrocytes, attenuate chondrocyte degeneration, and rescue degenerated cartilage in an osteochondral defect model in mice [64]. The implementation of hydrogels has the benefits of delivering MSC-EVs without rapid cargo clearance and accidental cartilage disruption, as well as targeted degradation in response to OA stimuli (such as pH and temperature) that ensures precise delivery to the specific site of injury or inflammation [64,65]. The targeting ability of EVs can also be enhanced by their surface modification. This can be achieved by the genetic modification of parental MSCs, enabling EVs to express proteins and other molecules with high affinity to surface receptors on the target cells [26,66]. The systemic delivery of EVs can also be employed to target deep layer chondrocytes and subchondral bone-resident cells, such as osteocytes and osteoclasts, that are not easily accessible when utilizing IA injections [67,68].
Outcome assessments presented fairly good uniformity and commonly measured structural improvements when utilizing the Osteoarthritis Research Society International (OARSI) or International Cartilage Repair Society (ICRS) scoring systems (Table 1). Histology and immunohistochemistry were normally used to demonstrate the increase in cartilage catabolic markers, such as collagen II and aggrecan, and reductions in cartilage catabolic enzymes, such as collagenases and aggrecanases. Similarly, because it has been well described that during the progression of OA, the synovium and infrapatellar fat pad serve as a source of pain-transmitting, immune, and inflammation-modulating neuropeptides [26,69,70], many studies have focused on the effects of EVs on inflammation and pain reversal. Specifically, IA MSC-EV delivery results in a regenerative immune phenotype characterized by a higher infiltration of M2 anti-inflammatory over M1 proinflammatory macrophages, with a concomitant reduction in IL-1β and TNF-α proinflammatory and an increase in IL-10 anti-inflammatory synovial cytokines [29,47,48,50,54,56,59]. Pain or gait analyses and measurements of systemic changes in serum biomarkers remain less frequent (Table 1). When tested, pain behavior and joint motion tended to exhibit recovery, suggesting the discernible health benefits of this treatment [45,65].
Furthermore, as stated, there are major regulatory challenges when using primary MSCs as EV-producing cells [71]. To overcome these hurdles and reduce batch-to-batch heterogeneity, immortalized primary MSCs or hES-derived MSC lines represent an attractive alternative [30,72]. Larger animal models need to be increasingly utilized and have already begun to be employed to test MSC-EVs therapies for OA. For example, in a recent minipig model study, the combination of MSC-EVs and hyaluronic acid was administered intra-articularly at weekly intervals and evidenced the promotion of functional osteochondral repair using magnetic resonance imaging and microcomputed tomography, as well as using histological and biomechanical tests [62]. The design of this study offered a clinically translatable protocol for a future clinical trial. Horses may represent another suitable large test model, as they experience gravitational impacts on joints, similar to people, and are amenable to arthroscopic intervention, diagnostic imaging, and repeated sample collection [73]. With the aim of providing an effective treatment as early as possible, and given the scarcity of disease management options for physically active people or people with high-BMI OA, utilizing mild and metabolic OA animal models would be another productive way forward [62].
There are currently five ongoing clinical trials in the MSC-EV field of OA regeneration, all targeting early-to-moderate knee OA and involving IA injections (Table 2). Being in the early phases of clinical research, the trials are primarily evaluating the potential of adverse events arising from the treatment. However, two trials have progressed to dose optimization and aim to compare clinical improvements after 1 year. Due to MSC-EV treatment being in its infancy, clinical trials have yet to publish their results.
Overall, preclinical studies show that MSC-EVs can regulate and prevent symptoms in OA, notably by restoring a healthy chondrocyte phenotype and by attenuating inflammation and pain signaling. These models are very useful for dose optimization and the development of effective material carriers and delivery methods. Future work should aim to standardize animal models, EV doses, and delivery schedules in order to ensure consistency between the studies and the selection of the most efficacious EV preparations. Three-dimensional joint models should continue to be developed, as well as novel technologies for the incorporation of further therapeutic and targeting agents, such as miRNAs and proteins, into the engineered EVs. Until recently, clinical implementation has seemed distant due to the challenges related to EVs tropism, limited native therapeutic efficacy, administration routes, bioavailability, and long-term therapeutic efficacy. At present, advanced protocols to engineer MSC-EVs and their combination with novel biomaterials have shown the significant therapeutic benefits of MSC-EVs in OA animal models. The first in-human clinical trials have been initiated to test the safety and efficacy of unmodified EVs. Therefore, MSC-EVs can be considered a promising nano-immunoregulatory and anabolic modality for musculoskeletal disease treatments and especially for OA.

Author Contributions

Conception and design, M.J., E.J. and D.K.; drafting of the article, M.J., E.J. and D.K.; critical revision of the article for important intellectual content, M.J., E.J. and D.K.; final approval of the article, E.J. and D.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Institutes of Health, grant number 5R21AR080388-02.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank the Soffer Family Foundation and the DRI Foundation for their generous funding support. We would also like to thank Xiao Nong Wang for her helpful advice and critical discussion on MSC-EV therapeutic use. These funding sources were not involved in any step of the study’s design, collection, analysis, or interpretation of the data, or in the writing of the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Vos, T.; Lim, S.S.; Abbafati, C.; Abbas, K.M.; Abbasi, M.; Abbasifard, M.; Abbasi-Kangevari, M.; Abbastabar, H.; Abd-Allah, F.; Abdelalim, A.; et al. Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet 2020, 396, 1204–1222. [Google Scholar] [CrossRef] [PubMed]
  2. Sinusas, K. Osteoarthritis: Diagnosis and treatment. Am. Fam. Physician 2012, 85, 49–56. [Google Scholar]
  3. Lieberthal, J.; Sambamurthy, N.; Scanzello, C.R. Inflammation in joint injury and post-traumatic osteoarthritis. Osteoarthr. Cartil. 2015, 23, 1825–1834. [Google Scholar] [CrossRef] [PubMed]
  4. Dell’Isola, A.; Steultjens, M. Classification of patients with knee osteoarthritis in clinical phenotypes: Data from the osteoarthritis initiative. PLoS ONE 2018, 13, e0191045. [Google Scholar] [CrossRef]
  5. Dell’Isola, A.; Allan, R.; Smith, S.L.; Marreiros, S.S.; Steultjens, M. Identification of clinical phenotypes in knee osteoarthritis: A systematic review of the literature. BMC Musculoskelet. Disord. 2016, 17, 425. [Google Scholar] [CrossRef]
  6. Kompel, A.J.; Roemer, F.W.; Murakami, A.M.; Diaz, L.E.; Crema, M.D.; Guermazi, A. Intra-articular Corticosteroid Injections in the Hip and Knee: Perhaps Not as Safe as We Thought? Radiology 2019, 293, 656–663. [Google Scholar] [CrossRef]
  7. Gardashli, M.; Baron, M.; Huang, C.; Kaplan, L.D.; Meng, Z.; Kouroupis, D.; Best, T.M. Mechanical loading and orthobiologic therapies in the treatment of post-traumatic osteoarthritis (PTOA): A comprehensive review. Front. Bioeng. Biotechnol. 2024, 12, 1401207. [Google Scholar] [CrossRef] [PubMed]
  8. Rizzo, M.G.; Best, T.M.; Huard, J.; Philippon, M.; Hornicek, F.; Duan, Z.; Griswold, A.J.; Kaplan, L.D.; Hare, J.M.; Kouroupis, D. Therapeutic Perspectives for Inflammation and Senescence in Osteoarthritis using Mesenchymal Stem Cells, Mesenchymal Stem Cell-Derived Extracellular Vesicles and Senolytic Agents. Cells 2023, 12, 1421. [Google Scholar] [CrossRef]
  9. Kouroupis, D.; Wang, X.N.; El-Sherbiny, Y.; McGonagle, D.; Jones, E. The Safety of Non-Expanded Multipotential Stromal Cell Therapies. In Safety, Ethics and Regulations; Pham, P.V., Rosemann, A., Eds.; Springer International Publishing: Cham, Switzerland, 2017; pp. 91–118. [Google Scholar]
  10. Kouroupis, D.; Sanjurjo-Rodriguez, C.; Jones, E.; Correa, D. Mesenchymal Stem Cell Functionalization for Enhanced Therapeutic Applications. Tissue Eng. Part B Rev. 2019, 25, 55–77. [Google Scholar] [CrossRef]
  11. Caplan, A.I.; Correa, D. The MSC: An Injury Drugstore. Cell Stem Cell 2011, 9, 11–15. [Google Scholar] [CrossRef]
  12. Welsh, J.A.; Goberdhan, D.C.I.; O’Driscoll, L.; Buzas, E.I.; Blenkiron, C.; Bussolati, B.; Cai, H.; Di Vizio, D.; Driedonks, T.A.P.; Erdbrügger, U.; et al. Minimal information for studies of extracellular vesicles (MISEV2023): From basic to advanced approaches. J. Extracell. Vesicles 2024, 13, e12404. [Google Scholar] [CrossRef] [PubMed]
  13. Herrmann, M.; Diederichs, S.; Melnik, S.; Riegger, J.; Trivanović, D.; Li, S.; Jenei-Lanzl, Z.; Brenner, R.E.; Huber-Lang, M.; Zaucke, F.; et al. Extracellular Vesicles in Musculoskeletal Pathologies and Regeneration. Front. Bioeng. Biotechnol. 2020, 8, 624096. [Google Scholar] [CrossRef] [PubMed]
  14. Zhuang, Y.; Jiang, S.; Yuan, C.; Lin, K. The potential therapeutic role of extracellular vesicles in osteoarthritis. Front. Bioeng. Biotechnol. 2022, 10, 1022368. [Google Scholar] [CrossRef] [PubMed]
  15. Shimomura, K.; Wong, K.L.; Saseendar, S.; Muthu, S.; Concaro, S.; Fernandes, T.L.; Mahmood, A. Exploring the potential of mesenchymal stem/stromal cell-derived extracellular vesicles as cell-free therapy for osteoarthritis: A narrative review. J. Cartil. Jt. Preserv. 2024, 4, 100184. [Google Scholar] [CrossRef]
  16. Kodama, J.; Wilkinson, K.J.; Otsuru, S. MSC-EV therapy for bone/cartilage diseases. Bone Rep. 2022, 17, 101636. [Google Scholar] [CrossRef]
  17. Chahla, J.; Cinque, M.E.; Piuzzi, N.S.; Mannava, S.; Geeslin, A.G.; Murray, I.R.; Dornan, G.J.; Muschler, G.F.; LaPrade, R.F. A Call for Standardization in Platelet-Rich Plasma Preparation Protocols and Composition Reporting: A Systematic Review of the Clinical Orthopaedic Literature. J. Bone Jt. Surg. 2017, 99, 1769–1779. [Google Scholar] [CrossRef]
  18. Ramaswamy Reddy, S.H.; Reddy, R.; Babu, N.C.; Ashok, G.N. Stem-cell therapy and platelet-rich plasma in regenerative medicines: A review on pros and cons of the technologies. J. Oral Maxillofac. Pathol. 2018, 22, 367–374. [Google Scholar] [CrossRef]
  19. Liu, Z.; Zhuang, Y.; Fang, L.; Yuan, C.; Wang, X.; Lin, K. Breakthrough of extracellular vesicles in pathogenesis, diagnosis and treatment of osteoarthritis. Bioact. Mater. 2023, 22, 423–452. [Google Scholar] [CrossRef]
  20. Labusek, N.; Mouloud, Y.; Köster, C.; Diesterbeck, E.; Tertel, T.; Wiek, C.; Hanenberg, H.; Horn, P.A.; Felderhoff-Müser, U.; Bendix, I.; et al. Extracellular vesicles from immortalized mesenchymal stromal cells protect against neonatal hypoxic-ischemic brain injury. Inflamm. Regen. 2023, 43, 24. [Google Scholar] [CrossRef]
  21. Huang, Y.; Zhang, X.; Zhan, J.; Yan, Z.; Chen, D.; Xue, X.; Pan, X. Bone marrow mesenchymal stem cell-derived exosomal miR-206 promotes osteoblast proliferation and differentiation in osteoarthritis by reducing Elf3. J. Cell. Mol. Med. 2021, 25, 7734–7745. [Google Scholar] [CrossRef]
  22. Xu, H.; Xu, B. BMSC-Derived Exosomes Ameliorate Osteoarthritis by Inhibiting Pyroptosis of Cartilage via Delivering miR-326 Targeting HDAC3 and STAT1//NF-κB p65 to Chondrocytes. Mediat. Inflamm. 2021, 2021, 9972805. [Google Scholar] [CrossRef] [PubMed]
  23. Tao, Y.; Zhou, J.; Wang, Z.; Tao, H.; Bai, J.; Ge, G.; Li, W.; Zhang, W.; Hao, Y.; Yang, X.; et al. Human bone mesenchymal stem cells-derived exosomal miRNA-361-5p alleviates osteoarthritis by downregulating DDX20 and inactivating the NF-κB signaling pathway. Bioorg. Chem. 2021, 113, 104978. [Google Scholar] [CrossRef] [PubMed]
  24. Pan, C.; Huang, W.; Chen, Q.; Xu, J.; Yao, G.; Li, B.; Wu, T.; Yin, C.; Cheng, X. LncRNA Malat-1 From MSCs-Derived Extracellular Vesicles Suppresses Inflammation and Cartilage Degradation in Osteoarthritis. Front. Bioeng. Biotechnol. 2021, 9, 772002. [Google Scholar] [CrossRef] [PubMed]
  25. Wu, J.; Kuang, L.; Chen, C.; Yang, J.; Zeng, W.N.; 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]
  26. Liebmann, K.; Castillo, M.A.; Jergova, S.; Best, T.M.; Sagen, J.; Kouroupis, D. Modification of Mesenchymal Stem/Stromal Cell-Derived Small Extracellular Vesicles by Calcitonin Gene Related Peptide (CGRP) Antagonist: Potential Implications for Inflammation and Pain Reversal. Cells 2024, 13, 484. [Google Scholar] [CrossRef]
  27. Leñero, C.; Kaplan, L.D.; Best, T.M.; Kouroupis, D. CD146+ Endometrial-Derived Mesenchymal Stem/Stromal Cell Subpopulation Possesses Exosomal Secretomes with Strong Immunomodulatory miRNA Attributes. Cells 2022, 11, 4002. [Google Scholar] [CrossRef]
  28. Kouroupis, D.; Kaplan, L.D.; Huard, J.; Best, T.M. CD10-Bound Human Mesenchymal Stem/Stromal Cell-Derived Small Extracellular Vesicles Possess Immunomodulatory Cargo and Maintain Cartilage Homeostasis under Inflammatory Conditions. Cells 2023, 12, 1824. [Google Scholar] [CrossRef]
  29. Kouroupis, D.; Kaplan, L.D.; Best, T.M. Human infrapatellar fat pad mesenchymal stem cells show immunomodulatory exosomal signatures. Sci. Rep. 2022, 12, 3609. [Google Scholar] [CrossRef]
  30. Teo, K.Y.W.; Tan, R.; Wong, K.L.; Hey, D.H.W.; Hui, J.H.P.; Toh, W.S. Small extracellular vesicles from mesenchymal stromal cells: The next therapeutic paradigm for musculoskeletal disorders. Cytotherapy 2023, 25, 837–846. [Google Scholar] [CrossRef]
  31. Zhang, B.; Tian, X.; Qu, Z.; Liu, J.; Yang, L.; Zhang, W. Efficacy of Extracellular Vesicles From Mesenchymal Stem Cells on Osteoarthritis in Animal Models: A Systematic Review and Meta-Analysis. Nanomedicine 2021, 16, 1297–1310. [Google Scholar] [CrossRef]
  32. Mohd Noor, N.A.; Abdullah Nurul, A.; Ahmad Mohd Zain, M.R.; Wan Nor Aduni, W.K.; Azlan, M. Extracellular Vesicles from Mesenchymal Stem Cells as Potential Treatments for Osteoarthritis. Cells 2021, 10, 1287. [Google Scholar] [CrossRef] [PubMed]
  33. 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] [PubMed]
  34. Zhang, Y.; Cao, X.; Li, P.; Fan, Y.; Zhang, L.; Ma, X.; Sun, R.; Liu, Y.; Li, W. microRNA-935-modified bone marrow mesenchymal stem cells-derived exosomes enhance osteoblast proliferation and differentiation in osteoporotic rats. Life Sci. 2021, 272, 119204. [Google Scholar] [CrossRef] [PubMed]
  35. Colombini, A.; Perucca Orfei, C.; Kouroupis, D.; Ragni, E.; De Luca, P.; ViganÒ, M.; Correa, D.; de Girolamo, L. Mesenchymal stem cells in the treatment of articular cartilage degeneration: New biological insights for an old-timer cell. Cytotherapy 2019, 21, 1179–1197. [Google Scholar] [CrossRef]
  36. He, L.; He, T.; Xing, J.; Zhou, Q.; Fan, L.; Liu, C.; Chen, Y.; Wu, D.; Tian, Z.; Liu, B.; et al. Bone marrow mesenchymal stem cell-derived exosomes protect cartilage damage and relieve knee osteoarthritis pain in a rat model of osteoarthritis. Stem Cell Res. Ther. 2020, 11, 276. [Google Scholar] [CrossRef]
  37. 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, 16214. [Google Scholar] [CrossRef]
  38. Warmink, K.; Rios, J.L.; Varderidou-Minasian, S.; Torres-Torrillas, M.; van Valkengoed, D.R.; Versteeg, S.; Eijkelkamp, N.; Weinans, H.; Korthagen, N.M.; Lorenowicz, M.J. Mesenchymal stem/stromal cells-derived extracellular vesicles as a potentially more beneficial therapeutic strategy than MSC-based treatment in a mild metabolic osteoarthritis model. Stem Cell Res. Ther. 2023, 14, 137. [Google Scholar] [CrossRef]
  39. Chen, X.; Shi, Y.; Xue, P.; Ma, X.; Li, J.; Zhang, J. Mesenchymal stem cell-derived exosomal microRNA-136-5p inhibits chondrocyte degeneration in traumatic osteoarthritis by targeting ELF3. Arthritis Res. Ther. 2020, 22, 256. [Google Scholar] [CrossRef]
  40. Ye, P.; Mi, Z.; Wei, D.; Gao, P.; Ma, M.; Yang, H. miR-3960 from Mesenchymal Stem Cell-Derived Extracellular Vesicles Inactivates SDC1/Wnt/β-Catenin Axis to Relieve Chondrocyte Injury in Osteoarthritis by Targeting PHLDA2. Stem Cells Int. 2022, 2022, 9455152. [Google Scholar] [CrossRef]
  41. Xia, Q.; Wang, Q.; Lin, F.; Wang, J. miR-125a-5p-abundant exosomes derived from mesenchymal stem cells suppress chondrocyte degeneration via targeting E2F2 in traumatic osteoarthritis. Bioengineered 2021, 12, 11225–11238. [Google Scholar] [CrossRef]
  42. Zhang, S.; Jin, Z. Bone Mesenchymal Stem Cell-Derived Extracellular Vesicles Containing Long Noncoding RNA NEAT1 Relieve Osteoarthritis. Oxidative Med. Cell. Longev. 2022, 2022, 5517648. [Google Scholar] [CrossRef] [PubMed]
  43. Yang, H.; Cong, M.; Huang, W.; Chen, J.; Zhang, M.; Gu, X.; Sun, C.; Yang, H. The Effect of Human Bone Marrow Mesenchymal Stem Cell-Derived Exosomes on Cartilage Repair in Rabbits. Stem Cells Int. 2022, 2022, 5760107. [Google Scholar] [CrossRef] [PubMed]
  44. Jin, Y.; Xu, M.; Zhu, H.; Dong, C.; Ji, J.; Liu, Y.; Deng, A.; Gu, Z. Therapeutic effects of bone marrow mesenchymal stem cells-derived exosomes on osteoarthritis. J. Cell. Mol. Med. 2021, 25, 9281–9294. [Google Scholar] [CrossRef]
  45. Zavatti, M.; Beretti, F.; Casciaro, F.; Bertucci, E.; Maraldi, T. Comparison of the therapeutic effect of amniotic fluid stem cells and their exosomes on monoiodoacetate-induced animal model of osteoarthritis. Biofactors 2020, 46, 106–117. [Google Scholar] [CrossRef]
  46. Woo, C.H.; Kim, H.K.; Jung, G.Y.; Jung, Y.J.; Lee, K.S.; Yun, Y.E.; Han, J.; Lee, J.; Kim, W.S.; Choi, J.S.; et al. Small extracellular vesicles from human adipose-derived stem cells attenuate cartilage degeneration. J. Extracell. Vesicles 2020, 9, 1735249. [Google Scholar] [CrossRef]
  47. Li, Y.; Tu, Q.; Xie, D.; Chen, S.; Gao, K.; Xu, X.; Zhang, Z.; Mei, X. Triamcinolone acetonide-loaded nanoparticles encapsulated by CD90(+) MCSs-derived microvesicles drive anti-inflammatory properties and promote cartilage regeneration after osteoarthritis. J. Nanobiotechnol. 2022, 20, 150. [Google Scholar] [CrossRef] [PubMed]
  48. Lu, L.; Wang, J.; Fan, A.; Wang, P.; Chen, R.; Lu, L.; Yin, F. Synovial mesenchymal stem cell-derived extracellular vesicles containing microRN555A-26a-5p ameliorate cartilage damage of osteoarthritis. J. Gene Med. 2021, 23, e3379. [Google Scholar] [CrossRef]
  49. 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]
  50. 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]
  51. Duan, A.; Shen, K.; Li, B.; Li, C.; Zhou, H.; Kong, R.; Shao, Y.; Qin, J.; Yuan, T.; Ji, J.; et al. Extracellular vesicles derived from LPS-preconditioned human synovial mesenchymal stem cells inhibit extracellular matrix degradation and prevent osteoarthritis of the knee in a mouse model. Stem Cell Res. Ther. 2021, 12, 427. [Google Scholar] [CrossRef]
  52. 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, 180–195. [Google Scholar] [CrossRef] [PubMed]
  53. 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, 64. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, K.; Li, F.; Yuan, Y.; Shan, L.; Cui, Y.; Qu, J.; Lian, F. Synovial Mesenchymal Stem Cell-Derived EV-Packaged miR-31 Downregulates Histone Demethylase KDM2A to Prevent Knee Osteoarthritis. Mol. Ther. Nucleic Acids 2020, 22, 1078–1091. [Google Scholar] [CrossRef]
  55. Yan, L.; Liu, G.; Wu, X. Exosomes derived from umbilical cord mesenchymal stem cells in mechanical environment show improved osteochondral activity via upregulation of LncRNA H19. J. Orthop. Transl. 2021, 26, 111–120. [Google Scholar] [CrossRef]
  56. Zhou, H.; Shen, X.; Yan, C.; Xiong, W.; Ma, Z.; Tan, Z.; Wang, J.; Li, Y.; Liu, J.; Duan, A.; et al. Extracellular vesicles derived from human umbilical cord mesenchymal stem cells alleviate osteoarthritis of the knee in mice model by interacting with METTL3 to reduce m6A of NLRP3 in macrophage. Stem Cell Res. Ther. 2022, 13, 322. [Google Scholar] [CrossRef]
  57. Fazaeli, H.; Kalhor, N.; Naserpour, L.; Davoodi, F.; Sheykhhasan, M.; Hosseini, S.K.E.; Rabiei, M.; Sheikholeslami, A. A Comparative Study on the Effect of Exosomes Secreted by Mesenchymal Stem Cells Derived from Adipose and Bone Marrow Tissues in the Treatment of Osteoarthritis-Induced Mouse Model. Biomed. Res. Int. 2021, 2021, 9688138. [Google Scholar] [CrossRef] [PubMed]
  58. Wang, Y.; Yu, D.; Liu, Z.; Zhou, F.; Dai, J.; Wu, B.; Zhou, J.; Heng, B.C.; Zou, X.H.; Ouyang, H.; et al. Exosomes from embryonic mesenchymal stem cells alleviate osteoarthritis through balancing synthesis and degradation of cartilage extracellular matrix. Stem Cell Res. Ther. 2017, 8, 189. [Google Scholar] [CrossRef]
  59. 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]
  60. Zhang, S.; Chu, W.C.; Lai, R.C.; Lim, S.K.; Hui, J.H.P.; Toh, W.S. Exosomes derived from human embryonic mesenchymal stem cells promote osteochondral regeneration. Osteoarthr. Cartil. 2016, 24, 2135–2140. [Google Scholar] [CrossRef]
  61. Li, S.; Liu, J.; Liu, S.; Jiao, W.; Wang, X. Mesenchymal stem cell-derived extracellular vesicles prevent the development of osteoarthritis via the circHIPK3/miR-124-3p/MYH9 axis. J Nanobiotechnol. 2021, 19, 194. [Google Scholar] [CrossRef]
  62. Zhang, S.; Wong, K.L.; Ren, X.; Teo, K.Y.W.; Afizah, H.; Choo, A.B.H.; Lai, R.C.; Lim, S.K.; Hui, J.H.P.; Toh, W.S. Mesenchymal Stem Cell Exosomes Promote Functional Osteochondral Repair in a Clinically Relevant Porcine Model. Am. J. Sports Med. 2022, 50, 788–800. [Google Scholar] [CrossRef] [PubMed]
  63. Zhang, H.; Huang, J.; Alahdal, M. Exosomes loaded with chondrogenic stimuli agents combined with 3D bioprinting hydrogel in the treatment of osteoarthritis and cartilage degeneration. Biomed. Pharmacother. 2023, 168, 115715. [Google Scholar] [CrossRef] [PubMed]
  64. Chen, P.; Zheng, L.; Wang, Y.; Tao, M.; Xie, Z.; Xia, C.; Gu, C.; Chen, J.; Qiu, P.; Mei, S.; et al. Desktop-stereolithography 3D printing of a radially oriented extracellular matrix/mesenchymal stem cell exosome bioink for osteochondral defect regeneration. Theranostics 2019, 9, 2439–2459. [Google Scholar] [CrossRef] [PubMed]
  65. Scalzone, A.; Sanjurjo-Rodríguez, C.; Berlinguer-Palmini, R.; Dickinson, A.M.; Jones, E.; Wang, X.-N.; Crossland, R.E. Functional and Molecular Analysis of Human Osteoarthritic Chondrocytes Treated with Bone Marrow-Derived MSC-EVs. Bioengineering 2024, 11, 388. [Google Scholar] [CrossRef] [PubMed]
  66. Wen, S.; Huang, X.; Ma, J.; Zhao, G.; Ma, T.; Chen, K.; Huang, G.; Chen, J.; Shi, J.; Wang, S. Exosomes derived from MSC as drug system in osteoarthritis therapy. Front. Bioeng. Biotechnol. 2024, 12, 1331218. [Google Scholar] [CrossRef]
  67. Wu, Y.; Li, J.; Zeng, Y.; Pu, W.; Mu, X.; Sun, K.; Peng, Y.; Shen, B. Exosomes rewire the cartilage microenvironment in osteoarthritis: From intercellular communication to therapeutic strategies. Int. J. Oral Sci. 2022, 14, 40. [Google Scholar] [CrossRef]
  68. Salunkhe, S.; Dheeraj; Basak, M.; Chitkara, D.; Mittal, A. Surface functionalization of exosomes for target-specific delivery and in vivo imaging & tracking: Strategies and significance. J. Control. Release 2020, 326, 599–614. [Google Scholar] [CrossRef]
  69. Walsh, D.A.; Mapp, P.I.; Kelly, S. Calcitonin gene-related peptide in the joint: Contributions to pain and inflammation. Br. J. Clin. Pharmacol. 2015, 80, 965–978. [Google Scholar] [CrossRef]
  70. Mashaghi, A.; Marmalidou, A.; Tehrani, M.; Grace, P.M.; Pothoulakis, C.; Dana, R. Neuropeptide substance P and the immune response. Cell. Mol. Life Sci. CMLS 2016, 73, 4249–4264. [Google Scholar] [CrossRef]
  71. Lai, R.C.; Tan, T.T.; Sim, W.K.; Zhang, B.; Lim, S.K. A roadmap from research to clinical testing of mesenchymal stromal cell exosomes in the treatment of psoriasis. Cytotherapy 2023, 25, 815–820. [Google Scholar] [CrossRef]
  72. Toh, W.S.; Yarani, R.; El Andaloussi, S.; Cho, B.S.; Choi, C.; Corteling, R.; De Fougerolles, A.; Gimona, M.; Herz, J.; Khoury, M.; et al. A report on the International Society for Cell & Gene Therapy 2022 Scientific Signature Series, “Therapeutic advances with native and engineered human extracellular vesicles”. Cytotherapy 2023, 25, 810–814. [Google Scholar] [CrossRef] [PubMed]
  73. O’Brien, T.J.; Hollinshead, F.; Goodrich, L.R. Extracellular vesicles in the treatment and prevention of osteoarthritis: Can horses help us translate this therapy to humans? Extracell. Vesicles Circ. Nucleic Acids 2023, 4, 151–169. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Potential advantages for MSC-EV therapy (blue) compared to platelet-rich plasma (PRP) and MSC therapies (red). PRP is a constituent of a patient’s blood (IA injected) that possesses large concentrations of platelets and regenerative factors. The source, basic description, and challenges for each therapy are given, as well as their derivation. Two of the main challenges involved in the PRP approach are circumvented by MSC-EV treatment, due to EVs being primarily donor-independent and grown and produced under good manufacturing practice (GMP) conditions. The potential tumorigenicity of MSC therapy is subverted using acellular particles instead of living cells. From this, the ability to produce EVs from immortalized cell lines instead of primary cells also reduces the batch-to-batch variability of the product. Although PRP and MSC therapy have received a greater deal of research in clinical trials, MSC-EVs have begun to undergo stage 1–2 clinical trials in OA therapeutics. Image taken from Biorender.
Figure 1. Potential advantages for MSC-EV therapy (blue) compared to platelet-rich plasma (PRP) and MSC therapies (red). PRP is a constituent of a patient’s blood (IA injected) that possesses large concentrations of platelets and regenerative factors. The source, basic description, and challenges for each therapy are given, as well as their derivation. Two of the main challenges involved in the PRP approach are circumvented by MSC-EV treatment, due to EVs being primarily donor-independent and grown and produced under good manufacturing practice (GMP) conditions. The potential tumorigenicity of MSC therapy is subverted using acellular particles instead of living cells. From this, the ability to produce EVs from immortalized cell lines instead of primary cells also reduces the batch-to-batch variability of the product. Although PRP and MSC therapy have received a greater deal of research in clinical trials, MSC-EVs have begun to undergo stage 1–2 clinical trials in OA therapeutics. Image taken from Biorender.
Bioengineering 11 00961 g001
Table 1. Summary of human MSC-EV therapeutics in OA animal models.
Table 1. Summary of human MSC-EV therapeutics in OA animal models.
SourceCargoOA ModelEVs DoseControl GroupFollow Up
Period
Outcome MeasuresKey ResultsRef.
hAD-MSCVarious miRNAs- MIA induced in rats
- BMM in mice
- In rat subacute OA group:
30 μL injection once per week for 21 days of hASC-EVs (1 × 108 particles) or PBS and hyaluronic acid
- In rat chronic OA group:
30 μL injection twice per week for 40 days of hASC-EVs (1 × 108 particles) or PBS and hyaluronic acid
- 6 μL injection once per week of EVs (1 × 108 particles) or PBS
PBS4 and 8 weeksHistology (H),
immunohistochemistry (IH), OARSI scores
EVs significantly attenuated OA progression and protected cartilage from degeneration in both the monosodium iodoacetate (MIA) rat and the surgical destabilization of the medial meniscus (DMM) mouse models[46]
hAFSCNot definedMIA induced
in rats
100 μg, repeated after 10 daysOA, AFSC (5 × 105 cells)3 weeksH/IH, OARSI scores, pain assessmentEnhanced pain tolerance, lower OARSI scores comparable to AFSC-treated defects[45]
hBM-MSCmiR-92a-3pCollagenase
induced in mice
15 μL (500 μg/mL), once a weekHealthy, OA, MSC-miR-92a-3p-transfected3 weeksH/IH, WBReduced cartilage matrix loss, improved col2a1 and aggrecan expression (further improved in MSC-miR-92a-3p group[33]
hBM-MSCNot definedGroove surgery in rats on high-fat diet7.77 × 107 particles (from 2 × 106 MSCs), five doses with 5-day intervalsOA, PBS, 2 × 106 MSCs11weeksµCT scans at week 0, 12 and 24, pain behavior, H/IH, OARSI scoresUnchanged OARSI scores, pain behavior and MMP13 staining in cartilage (compared to MSC group where these were unexpectedly aggravated)[38]
hBM-MSCmiR-136-5pPost-traumatic oleanolic acid (OA) in mice100 μL single injection of 1011 particles/mL of EVs or miR-136-5p EVsHealthy, OA1 hHistology (H),
immunohistochemistry (IH)
miR-136-5p was found to reduce the degeneration of cartilage extracellular matrix[39]
hBM-MSCmiR-3960DMM in mice10 μL MSC-EVs or sterile normal saline were injected into the articular capsule for 3 weeks (once a week)Sham, OA, MSCs-EVs-agomir-NC3 weeksHistology (H), pain
assessment
miR-3960 shuttled by MSC- EVs protected against apoptosis and ECM degradation in chondrocytes[40]
hBM-MSCmiR-125a-5pACL rupture
in mice
100 μL single injection of 1011 particles/mLHealthy, OA1 hWB, qPCRAlleviation in chondrocyte extracellular matrix degradation[41]
hBM-MSCmiR-361-5pACL rupture
in rats
250 ng/5 µL EVs-miR-NC or EVs-miR-361-5p postoperatively for 7 daysSham, OA8 weeksHistology (H), WB, qPCRmiR-361-5p alleviates cartilage damage[23]
hBM-MSCLncRNA NEAT1DMM in mice10 μg EVs or an equivalent amount of PBS, twice a week for 1 monthSham, OA, DMM + Lv − NC − BMSCs − EVs group, DMM + Lv − NEAT1 – BMSCs − EVs group, DMM + Lv − NEAT1 − BMSCs − EVs + sh − Sesn2 group7 weeksHistology (H),
immunohistochemistry (IH), OARSI scores
LncRNA NEAT1 induced the proliferation and autophagy of chondrocytes but inhibited their apoptosis[42]
hBM-MSCNot definedOsteochondral
defect in rabbits
300 μL injection once a week for 4 weeks of 1 × 1010 particles/mL (low dosage) or 5 × 1010 particles/mL (high dosage) or PBSHealthy, OA5 weeksHistology (H), ICRS scoresFacilitates cartilage regeneration and enhances viability of chondrocytes[43]
hBM-MSClncRNA MEG-3DMM in rats100 µL injection per week EVs solution (100 µg) or MSCs (106 cell)Sham, OA8 weeksHistology (H),
immunohistochemistry (IH), OARSI scores,
micro-CT
- MSC and MSC-EVs alleviated cartilage destruction and subchondral bone remodeling
- lncRNA MEG-3 also reduced the senescence and apoptosis of chondrocytes
[44]
hBM-MSC and hAD-MSCNot definedCiproflaxin
induced in mice
25 mL at 100 μg/μL, once a weekHealthy, OA, PBS3 weeksH/IH, OARSI scores, real-time PCRLower OARSI scores, upregulated COLII protein and Sox9, COL2 and Aggrecan genes in cartilage, particularly in BM-MSC group[57]
hESC-MSCNot definedDMM in mice5 μL every 3 days for 4 weeksOA, PBS4 weeksHistology (H),
immunohistochemistry (IH), OARSI scores
Lower OARSI scores, stronger Col II and weaker ADAMTS5 staining of cartilage[58]
hESC-MSCNot definedOsteochondral
defect in rats
100 μL injection once a
week of 100 μg EVs or PBS
OA, PBS12 weeksHistology (H),
immunohistochemistry (IH), multiplex cytokine array
EV-treated defects displayed a regenerative immune phenotype characterized by a higher infiltration of CD163+ regenerative M2 macrophages over CD86+ M1 macrophages, with a concomitant reduction in proinflammatory synovial cytokines IL-1β and TNF-α[59]
hESC-MSCNot definedOsteochondral
defect in rats
100 μL injection once a week of 100 μg EVs or PBSOA, PBS12 weeksHistology (H), ICRS scores- Enhanced gross appearance and improved histological scores
- EV-treated defects displayed complete restoration of cartilage and subchondral bone with characteristic features including a hyaline cartilage with good surface regularity, complete bonding to adjacent cartilage, and extracellular matrix deposition
[60]
hIFP-MSCmiR-100-5pDMM in mice10 μL (1010 particles/mL) twice a weekOA, PBS4 or 6 weeksH/IH, OARSI scores, gait analysisLower OARSI scores, stronger Col II and weaker ADAMTS5 and MMP13 staining of cartilage, partial improvement of the gait patterns[25]
hIFP-MSCVarious miRNAsMIA induced
in rats
50 μL single injection of EVs derived from 5 × 105 and 5 × 106 IFP-MSCHealthy, OA4 daysHistology (H),
immunohistochemistry (IH)
MSC-EV therapeutic treatment resulted in robust macrophage polarization towards an anti-inflammatory therapeutic M2 phenotype within the synovium/IFP tissues[29]
hIFP-MSCVarious miRNAs and CD10 proteinMIA induced
in rats
50 μL single injection of EVs derived from 1 × 106 IFP-MSCHealthy, OA4 daysHistology (H),
immunohistochemistry (IH)
CD10High EV treatment resulted in robust chondroprotective effects by retaining articular cartilage structure/composition and PRG4 (lubricin)-expressing cartilage cells[28]
iMSC and hSynovium-MSC Not definedCollagenase
induced in mice
8 μL injection once per week for 21 days of iMSC-EVs (1.0 × 1010/mL) or MSC-EVs (1.0 × 1010/mL) or PBSHealthy, OA4 weeksHistology (H),
immunohistochemistry (IH), OARSI scores
The injection of iMSC-EVs and MSC-EVs both attenuated OA in the mouse OA model, but iMSC EVs had a superior therapeutic effect compared with MSC-EVs[53]
hSynovium-MSCNot definedACL rupture
in rabbits and rats
For rabbits:
12 mg once per week of TA, T-NP, T-RNP, CD90@MV, T-CD90@NP
For rats:
0.25 mg once per week of TA, T-NP, T-RNP, CD90@MV, T-CD90@NP
Sham, OA24 weeks for rabbits and 2 weeks for ratsHistology (H),
immunohistochemistry (IH), micro-CT,
RNAseq
- CD90 EVs enhanced repair of damaged cartilage and effective anti-inflammatory ability
- CD90 EVs promoted the regeneration of chondrocytes, reduced apoptosis via the FOXO pathway, and influenced type 2 macrophage polarization to regulate inflammation through IL-10
[47]
hSynovium-MSCmiR-26a-5pDMM in rats30 μL injection per week of GW inhibitor or EVs or EV-NC or EV-inhibitor (1011 particles/mL) or PBSSham, OA4 weeksHistology (H), ELISA, qPCRmiR-26a-5p MSC EVs inhibit apoptosis and inflammation and ameliorate cartilage damage of OA[48]
hSynovium-MSCmiR-155-5pDMM in mice30 μL injection of MSC-EVs (1011 EVs particles/mL) or MSC-155-5p-EVs (1011 EVs particles/mL)Healthy, OA2 weeksHistology (H),
immunohistochemistry (IH), OARSI scores
miR-155-5p EVs prevent osteoarthritis via enhancing proliferation and migration, attenuating apoptosis, and modulating extracellular matrix secretion in chondrocytes[49]
hSynovium-MSCcircRNA3503DMM in rats100 μL injection of PLEL@SMSC-EVs or PLEL@Wnt5a/b-dKO-EVs or PLEL@circRNA3503-OE-EVs or PLEL@dKO-OE-EVs or PLEL@SalineHealthy, OA24 weeksHistology (H)- circRNA3503-OE-EVs alleviate inflammation-induced apoptosis and the imbalance between ECM synthesis and ECM degradation
- circRNA3503-OE-EVs promote chondrocyte renewal to alleviate the progressive loss of chondrocytes
[50]
hSynovium-MSCNot definedDMM in mice10 μL injection twice weekly of PBS-EVs (1011 particles/mL) or PBS-LPS-pre EVs (1011 particles/mL) or PBSSham, PBS6 weeksHistology (H),
immunohistochemistry (IH), OARSI scores
EVs derived from LPS-preconditioned MSC inhibit extracellular matrix degradation and prevent osteoarthritis[51]
hSynovium-MSCmiR-140-5pDMM in rats100 μL injection of Synovium-MSC-EVs (1011 EVs particles/mL) or SMSC-140 EVs (1011 EVs particles/mL)Healthy, OA12 weeksHistology (H),
immunohistochemistry (IH), OARSI scores
miR-140-5p EVs enhance cartilage tissue regeneration and prevent osteoarthritis[52]
hSynovium-MSCmiR-31DMM in mice5 μL injection every 3 days for 4 weeks of Synovium-MSC-EVs or EVs (miR-31 mimic) or PBSSham, OA12 weeksHistology (H),
immunohistochemistry (IH), OARSI scores, ELISA
MSC EVs and EVs from miR-31-overexpressed MSC alleviated cartilage damage and inflammation in knee joints in vivo[54]
hUC-MSCLncRNA H19Osteochondral
defect in rats
100 μL injection once per week of EVs from UC-MSC transfected with siRNA H19 (si-EVs, 1 mg/mL) or EVs from UC-MSC with mechanical stimulation (S-EVs, 1 mg/mL) or PBSPBS4 and 8 weeksBehavioral analysis,
histology (H), MRI, ICRS scores
LncRNA H19 relieve pain levels during the early stages of cartilage repair via enhanced chondrocyte proliferation and matrix synthesis[55]
hUC-MSCmiR-1208DMM in mice10 μL injection twice per week of MSC-EVs (1011 particles/mL) or antagomiR-NC (200 nmol/mL) or antagomiR-1208 (200 nmol/mL) or PBSSham, PBS6 weeksHistology (H),
immunohistochemistry (IH), micro-CT, ELISA
MSC-EVs inhibited the secretion of proinflammatory factors and the degradation of cartilage ECM[56]
hUC-MSCcircHIPK3Collagenase
induced in mice
Single injection of MSC-EVs or MSC-circHIPK3-EVs or circHIPK3 or PBSHealthy, OANot reportedHistology (H),
immunohistochemistry (IH), WB, qPCR
MSC-circHIPK3 EVs inhibited cartilage degradation[61]
hAD-MSC: adipose tissue-derived MSC, hBM-MSC: bone marrow-derived MSC, hAFSC: amniotic fluid-derived MSC, hESC-MSC: embryonic stem cell-derived MSC, hIFP-MSC: infrapatellar fat pad-derived MSC, hSynovium-MSC: synovium-derived MSC, hUC-MSC: umbilical cord-derived MSC.
Table 2. Summary of MSC-EV-based clinical trials in OA therapy.
Table 2. Summary of MSC-EV-based clinical trials in OA therapy.
EV SourcePatientsDoseType of
Trial
Delivery
Method
Follow Up
Period
Primary
Outcome
Trial ID
Allogeneic UC MSCsKL 2–3 knee OA, n = 103–5 × 1011
particles
Phase 1, safety and efficacy trialSingle IA injection12 monthsAdverse events, pain and disability
reduction
NCT05060107
Allogeneic UC MSCsKL 2–3 knee OA, n = 12, n = 4 per group2 × 109 particles/dose;
6 × 109 particles/dose;
2 × 1010 particles/dose
Phase 1, open-label dose-escalation trialSingle IA injection12 monthsAdverse events, pain and disability
reduction,
percentage of responders at 52 weeks
NCT06431152
Autologous SF-MSCsBilateral degenerative meniscus, early OA, 3 groups of n = 10EVs from 106 SF MSCs;
106 SF MSCs; control
Phase 2, randomized safety and efficacy trialSingle IA injection12 monthsAdverse events, pain reduction, cytokine measurements, knee motion and physical
activity
NCT05261360
Allogeneic MSCsKL 1–3 knee OA in both knees, n = 20Not reportedPhase 1, safety and efficacy trialIA injection, day 1 and day 901, 3, and 6 monthsAdverse events, evaluation of pain, measurements of knee functionNCT06466850
Platelets:
PEP (Purified EVs Product); 2 doses; with or without 1% sodium hyaluronate
KL 2–3 knee OA, n = 241 or 2 vialsPhase 1, randomized safety and exploratory efficacy dose-escalation trialSingle IA injection90 days (primary safety); 12 months (long-term safety)Primary and long-term safety, clinical improvements after 12 monthsNCT06463132
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jones, M.; Jones, E.; Kouroupis, D. The Use of Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles in the Treatment of Osteoarthritis: Insights from Preclinical Studies. Bioengineering 2024, 11, 961. https://doi.org/10.3390/bioengineering11100961

AMA Style

Jones M, Jones E, Kouroupis D. The Use of Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles in the Treatment of Osteoarthritis: Insights from Preclinical Studies. Bioengineering. 2024; 11(10):961. https://doi.org/10.3390/bioengineering11100961

Chicago/Turabian Style

Jones, Mitch, Elena Jones, and Dimitrios Kouroupis. 2024. "The Use of Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles in the Treatment of Osteoarthritis: Insights from Preclinical Studies" Bioengineering 11, no. 10: 961. https://doi.org/10.3390/bioengineering11100961

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop