Advertisement
Research Article|Articles in Press

Small extracellular vesicles from mesenchymal stromal cells: the next therapeutic paradigm for musculoskeletal disorders

  • Kristeen Ye Wen Teo
    Affiliations
    Department of Orthopedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore

    Faculty of Dentistry, National University of Singapore, Singapore, Republic of Singapore
    Search for articles by this author
  • Rachel Tan
    Affiliations
    Department of Orthopedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
    Search for articles by this author
  • Keng Lin Wong
    Affiliations
    Department of Orthopedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore

    Department of Orthopedic Surgery, Sengkang General Hospital, Singapore Health Services, Singapore, Republic of Singapore
    Search for articles by this author
  • Dennis Hwee Weng Hey
    Affiliations
    Department of Orthopedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
    Search for articles by this author
  • James Hoi Po Hui
    Affiliations
    Department of Orthopedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore

    Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore, Republic of Singapore
    Search for articles by this author
  • Wei Seong Toh
    Correspondence
    Correspondence: Wei Seong Toh, PhD, Department of Orthopedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, National University Health System Tower Block, Level 11, Singapore 119228, Republic of Singapore.
    Affiliations
    Department of Orthopedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore

    Faculty of Dentistry, National University of Singapore, Singapore, Republic of Singapore

    Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore, Republic of Singapore

    Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Republic of Singapore

    Integrative Sciences and Engineering Program, NUS Graduate School, National University of Singapore, Singapore, Republic of Singapore
    Search for articles by this author
Open AccessPublished:May 15, 2023DOI:https://doi.org/10.1016/j.jcyt.2023.04.011

      Abstract

      Musculoskeletal disorders are one of the biggest contributors to morbidity and place an enormous burden on the health care system in an aging population. Owing to their immunomodulatory and regenerative properties, mesenchymal stromal/stem cells (MSCs) have demonstrated therapeutic efficacy for treatment of a wide variety of conditions, including musculoskeletal disorders. Although MSCs were originally thought to differentiate and replace injured/diseased tissues, it is now accepted that MSCs mediate tissue repair through secretion of trophic factors, particularly extracellular vesicles (EVs). Endowed with a diverse cargo of bioactive lipids, proteins, nucleic acids and metabolites, MSC-EVs have been shown to elicit diverse cellular responses and interact with many cell types needed in tissue repair. The present review aims to summarize the latest advances in the use of native MSC-EVs for musculoskeletal regeneration, examine the cargo molecules and mechanisms underlying their therapeutic effects, and discuss the progress and challenges in their translation to the clinic.

      Key Words

      Introduction

      Musculoskeletal disorders, with osteoarthritis (OA) and low back pain identified as the major conditions, are the leading cause of disability worldwide, resulting in loss of productivity and high socioeconomic costs [
      • James SL
      • Abate D
      • Abate KH
      • Abay SM
      • Abbafati C
      • Abbasi N
      • et al.
      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.
      ]. The prevalence and disability due to musculoskeletal disorders are expected to continue to rise with an increasingly obese and aging population. OA is the most common form of arthritis and one of the leading causes of disability, affecting over 500 million people worldwide [
      • Hunter DJ
      • March L
      • Chew M.
      Osteoarthritis in 2020 and beyond: a Lancet Commission.
      ]. Although pharmacological therapies such as non-steroidal anti-inflammatory drugs are generally effective for symptomatic relief in musculoskeletal disorders, none of these treatments are curative. Consequently, if poorly managed, surgery is the only solution at the end stage of the disease. Thus, there is a need to explore new therapeutic options for better management of musculoskeletal disorders.
      In the past decade, mesenchymal stromal/stem cells (MSCs) have emerged as a promising therapeutic agent for a wide variety of injuries and diseases, with >1400 registered clinical trials (https://clinicaltrials.gov/). As multipotent progenitor cells, these cells are able to differentiate into multiple connective tissue cell types, including adipocytes, chondrocytes and osteocytes [
      • Pittenger MF
      • Mackay AM
      • Beck SC
      • Jaiswal RK
      • Douglas R
      • Mosca JD
      • et al.
      Multilineage potential of adult human mesenchymal stem cells.
      ]. MSCs are plastic-adherent under standard conditions, express CD105, CD73 and CD90, but not CD45, CD34, CD14 or CD11b, CD79α or CD19 and HLA-DR surface molecules, and under appropriate conditions can differentiate to adipogenic, chondrogenic and osteogenic lineages [
      • Dominici M
      • Le Blanc K
      • Mueller I
      • Slaper-Cortenbach I
      • Marini F
      • Krause D
      • et al.
      Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement.
      ]. Although the use of MSCs was originally predicated on their potential to differentiate and replace lost cells in injured or diseased tissues, it is now accepted that MSCs mediate tissue repair through paracrine secretion, particularly extracellular vesicles (EVs).
      EVs are a heterogeneous class of bi-lipid membrane particles released by all cells and are broadly divided into the major subtypes known to date; namely, exosomes, microvesicles and apoptotic bodies [
      • Théry C
      • Ostrowski M
      • Segura E.
      Membrane vesicles as conveyors of immune responses.
      ]. Being of endosomal origin, exosomes are released through fusion of multivesicular bodies with the plasma membrane and are about 40–150 nm in diameter. Microvesicles (also known as ectosomes or microparticles) are formed through budding of the plasma membrane and range from 100 nm to 1000 nm in size. Apoptotic bodies range from 50 nm to 5000 nm and are formed by blebbing of apoptotic cells. The molecular composition and bioactive cargo of different EV subtypes can include various combinations of lipids, proteins, nucleic acids and metabolites depending on the type and state of the cell, as well as the biogenesis pathway. Much of the EV cargo is located inside the vesicles. However, EVs may also acquire a protein corona through diverse molecular interactions of the EV surface with the proteins present in the surrounding interstitial fluid or blood plasma [
      • Tóth EÁ
      • Turiák L
      • Visnovitz T
      • Cserép C
      • Mázló A
      • Sódar BW
      • et al.
      Formation of a protein corona on the surface of extracellular vesicles in blood plasma.
      ].
      Various isolation methods, ranging from ultracentrifugation, filtration and size-exclusion chromatography to polyethylene glycol/polymer-based enrichment and antibody-based methods, have commonly been used for EV isolation [
      • Reiner AT
      • Witwer KW
      • van Balkom BWM
      • de Beer J
      • Brodie C
      • Corteling RL
      • et al.
      Concise Review: Developing Best-Practice Models for the Therapeutic Use of Extracellular Vesicles.
      ]. However, isolation of EV subpopulations is presently challenging because of profound EV heterogeneity, with similar or overlapping biochemical or biophysical properties, and lack of definitive markers to unambiguously identify an EV subtype [
      • Reiner AT
      • Witwer KW
      • van Balkom BWM
      • de Beer J
      • Brodie C
      • Corteling RL
      • et al.
      Concise Review: Developing Best-Practice Models for the Therapeutic Use of Extracellular Vesicles.
      ,
      • Witwer KW
      • Van Balkom BWM
      • Bruno S
      • Choo A
      • Dominici M
      • Gimona M
      • et al.
      Defining mesenchymal stromal cell (MSC)-derived small extracellular vesicles for therapeutic applications.
      ]. In view of these challenges, the International Society for Extracellular Vesicles, in an effort to promote standardization, reproducibility and rigor in EV research, has recommended that the EV nomenclature, isolation and characterization follow the minimal information for studies of EVs 2018 guidelines [
      • Théry C
      • Witwer KW
      • Aikawa E
      • Alcaraz MJ
      • Anderson JD
      • Andriantsitohaina R
      • 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.
      ]. Minimal information for studies of EVs 2018 recommends use of the collective term “EVs” as well as operative terms such as “size” (small or large) to enhance the understanding of homogeneity of EV preparation. Information on sample type, culture conditions, collection and pre-processing and experimental details of EV separation, concentration and characterization should also be provided.
      Although EVs were originally thought to collect and dispose of cellular waste, it is now recognized that EVs mediate intercellular communication through the delivery of bioactive cargoes to elicit biological responses in recipient cells. With MSC-EVs, many of these biological responses translate to a therapeutic outcome in injured or diseased cells. Increasingly, native MSC-EVs have replicated the wide-ranging therapeutic efficacy of their parental MSCs in several injuries and diseases, including musculoskeletal disorders. Essentially, native MSC-EVs are EVs secreted by MSCs where the EVs or secreting MSCs are not modified or engineered to modify EV composition. In this review, we will summarize the latest advances in the use of native MSC-EVs for the treatment of various musculoskeletal disorders, examine the cargo molecules and mechanisms underlying their therapeutic effects, and discuss the progress and challenges in their translation to the clinic.

      MSC-EVs

      For many years, the search for active trophic factors in MSC secretions mainly focused on growth factors, cytokines and chemokines. However, it became evident that no single factor could sufficiently account for all of the observed therapeutic effects of MSCs in tissue repair [
      • Ghannam S
      • Bouffi C
      • Djouad F
      • Jorgensen C
      • Noël D.
      Immunosuppression by mesenchymal stem cells: mechanisms and clinical applications.
      ]. In 2009, Bruno et al. [
      • Bruno S
      • Grange C
      • Deregibus MC
      • Calogero RA
      • Saviozzi S
      • Collino F
      • et al.
      Mesenchymal Stem Cell-Derived Microvesicles Protect Against Acute Tubular Injury.
      ] first attributed the therapeutic effects of MSC-conditioned medium in a mouse model of glycerol-induced acute kidney injury to 80- to 1000-nm EVs, which at that time were called microvesicles. In 2010, Lai et al. [
      • Lai RC
      • Arslan F
      • Lee MM
      • Sze NSK
      • Choo A
      • Chen TS
      • et al.
      Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury.
      ] showed by size fractionation studies that 50- to 200-nm EVs, which were originally called exosomes, were efficacious against myocardial ischemia–reperfusion injury. In head-to-head comparisons in different animal models, EVs were shown to be therapeutically equivalent to their parental MSCs [
      • Doeppner TR
      • Herz J
      • Görgens A
      • Schlechter J
      • Ludwig AK
      • Radtke S
      • et al.
      Extracellular Vesicles Improve Post-Stroke Neuroregeneration and Prevent Postischemic Immunosuppression.
      ,
      • HE J
      • WANG Y
      • SUN S
      • YU M
      • WANG C
      • PEI X
      • et al.
      Bone marrow stem cells-derived microvesicles protect against renal injury in the mouse remnant kidney model.
      ]. To address the issue of heterogeneity of MSC-EVs, Bruno et al. [
      • Bruno S
      • Tapparo M
      • Collino F
      • Chiabotto G
      • Deregibus MC
      • Soares Lindoso R
      • et al.
      Renal Regenerative Potential of Different Extracellular Vesicle Populations Derived from Bone Marrow Mesenchymal Stromal Cells.
      ] performed differential ultracentrifugation to separate different MSC-EV subpopulations and found that a 100,000 g exosome-enriched EV population but not a 10,000 g microvesicle-enriched EV population was renal protective. More recently, Xu et al. [
      • Xu T
      • Lin Y
      • Yu X
      • Jiang G
      • Wang J
      • Xu K
      • et al.
      Comparative Effects of Exosomes and Ectosomes Isolated From Adipose-Derived Mesenchymal Stem Cells on Achilles Tendinopathy in a Rat Model.
      ] showed that EVs with a mean size of approximately 150 nm had more beneficial effects on tendon repair in a rat model of Achilles tendinopathy than EVs with a mean size of approximately 230 nm.
      Today, it is widely accepted that MSCs exert their therapeutic effects through the release of EVs, particularly small EVs (sEVs) with a size range of 50–200 nm [
      • Witwer KW
      • Van Balkom BWM
      • Bruno S
      • Choo A
      • Dominici M
      • Gimona M
      • et al.
      Defining mesenchymal stromal cell (MSC)-derived small extracellular vesicles for therapeutic applications.
      ]. This identification of sEVs as the active agent underpinning the therapeutic effects of MSCs provided the rationale to transform living MSC therapy into non-living MSC-sEV therapy, with many inherent advantages [
      • Toh WS
      • Lai RC
      • Hui JHP
      • Lim SK.
      MSC exosome as a cell-free MSC therapy for cartilage regeneration: Implications for osteoarthritis treatment.
      ]. Unlike living MSCs, which have the capacity to engraft and replicate, MSC-sEVs are non-living and non-replicative and thus have a reduced risk of tumor or aberrant tissue formation. Their small size also renders MSC-sEV preparations amenable to sterilization by filtration and administration via different routes with reduced risk of embolism. MSC-sEV production is also more scalable and amenable to process optimization. For instance, the cell source for sEV production can be selected and immortalized to generate a high sEV-yielding clonal cell line with infinite expansion potential to ensure reproducible and cost-effective production of sEVs on a large scale [
      • Chen TS
      • Arslan F
      • Yin Y
      • Tan SS
      • Lai RC
      • Choo ABH
      • et al.
      Enabling a robust scalable manufacturing process for therapeutic exosomes through oncogenic immortalization of human ESC-derived MSCs.
      ].

      MSC-sEVs for Musculoskeletal Regeneration

      Within the musculoskeletal system, MSC-sEVs have demonstrated therapeutic efficacy in a wide variety of conditions, ranging from cartilage and bone defects to debilitating conditions such as OA and intervertebral disk (IVD) degeneration. Here we will summarize key studies and highlight the MSC-sEV-associated cargo molecules that have demonstrated functional effects (Table 1, Table 2, Table 3, Figure 1).
      Table 1Overview of studies using MSC-sEVs for articular cartilage repair.
      MSC originIn vivo/in vitro modelKey outcomesPathway (EV molecule)Author
      H-EMBRat osteochondral defectEnhanced osteochondral repair.Zhang
      • Zhang S
      • Chu WC
      • Lai RC
      • Lim SK
      • Hui JHP
      • Toh WS.
      Exosomes derived from human embryonic mesenchymal stem cells promote osteochondral regeneration.
      H-EMBRat osteochondral defect/Rat chondrocytesEnhanced osteochondral repair.

      Increased cell proliferation, migration and matrix synthesis.
      AKT/ERK

      (CD73)
      Zhang
      • Zhang S
      • Chuah SJ
      • Lai RC
      • Hui JHP
      • Lim SK
      • Toh WS.
      MSC exosomes mediate cartilage repair by enhancing proliferation, attenuating apoptosis and modulating immune reactivity.
      H-EMBRabbit osteochondral defectEnhanced osteochondral repair with improved mechanical competence.Wong
      • Wong KL
      • Zhang S
      • Wang M
      • Ren X
      • Afizah H
      • Lai RC
      • et al.
      Intra-Articular Injections of Mesenchymal Stem Cell Exosomes and Hyaluronic Acid Improve Structural and Mechanical Properties of Repaired Cartilage in a Rabbit Model.
      H-EMBPig osteochondral defectEnhanced osteochondral repair with improved gross appearance, MRI, histology and mechanical competence.Zhang
      • Zhang S
      • Wong KL
      • Ren X
      • Teo KYW
      • Afizah H
      • Choo ABH
      • et al.
      Mesenchymal Stem Cell Exosomes Promote Functional Osteochondral Repair in a Clinically Relevant Porcine Model.
      H-EMBPig chondral defect with BMS/Human chondrocytesImpaired cartilage repair because of osseous ingrowth but enhanced subchondral bone healing. Improved chondrocyte proliferation.Hede
      • Hede KTC
      • Christensen BB
      • Olesen ML
      • Thomsen JS
      • Foldager CB
      • Toh WS
      • et al.
      Mesenchymal Stem Cell Extracellular Vesicles as Adjuvant to Bone Marrow Stimulation in Chondral Defect Repair in a Minipig Model.
      Ms-BMRabbit osteochondral defect/IL-1β-treated chondrocytesRestored chondrocyte mitochondrial function, increased matrix synthesis and enhanced synovial M2 macrophage infiltration.Mitochondrial biogenesis

      (–)
      Chen
      • Chen P
      • Zheng L
      • Wang Y
      • Tao M
      • Xie Z
      • Xia C
      • et al.
      Desktop-stereolithography 3D printing of a radially oriented extracellular matrix/mesenchymal stem cell exosome bioink for osteochondral defect regeneration.
      UCRat osteochondral defect/IL-1β-treated chondrocytesPromoted chondrocyte migration, matrix secretion and suppression of apoptosis and senescence.miR-29b-3p/FoxO3

      (lncRNA H19)
      Yan
      • Yan L
      • Liu G
      • Wu X.
      The umbilical cord mesenchymal stem cell-derived exosomal lncRNA H19 improves osteochondral activity through miR-29b-3p/FoxO3 axis.
      H-EMBRat TMJ OA/IL-1β-treated rat chondrocytesEnhanced TMJ repair with reduced pain and inflammation.

      Restored chondrocyte matrix homeostasis.
      AKT/ERK/AMPK

      (CD73)
      Zhang
      • Zhang S
      • Teo KYW
      • Chuah SJ
      • Lai RC
      • Lim SK
      • Toh WS.
      MSC exosomes alleviate temporomandibular joint osteoarthritis by attenuating inflammation and restoring matrix homeostasis.
      H-AFRat MIA-induced knee OA/THP1 macrophages

      Reduced pain and enhanced cartilage repair with increased matrix deposition and M2 macrophage polarization.

      (TGF-β)
      Zavatti
      • 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.
      H-MSCRat collagenase II-induced knee OA/IL-1β-treated rat chondrocytesEnhanced cartilage repair. Increased chondrocyte proliferation and migration and reduced apoptosis.

      (lncRNA KLF3-AS1)
      Liu
      • Liu Y
      • Zou R
      • Wang Z
      • Wen C
      • Zhang F
      • Lin F.
      Exosomal KLF3-AS1 from hMSCs promoted cartilage repair and chondrocyte proliferation in osteoarthritis.
      H-IPFPMouse DMM-induced knee OA/IL-1β-treated chondrocytesImproved cartilage repair and gait performance.

      Enhanced matrix synthesis, reduced apoptosis and decreased expression of catabolic factors.
      mTOR/autophagy

      (miR-100-5p)
      Wu
      • Wu J
      • Kuang L
      • Chen C
      • Yang J
      • Zeng WN
      • Li T
      • 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.
      H-BMMouse PT OA model/ChondrocytesAttenuated cartilage degeneration.

      Enhanced chondrocyte migration and matrix synthesis.
      ELF3

      (miR-136-5p)
      Chen
      • 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.
      AF, amniotic fluid; AMPK, adenosine monophosphate-activated protein kinase; BM, bone marrow; BMS, bone marrow stimulation; DMM, destabilization of medial meniscus; ELF3, E74-like factor 3; EMB, embryonic; ERK, extracellular signal-regulated kinase; H, human; IPFP, infrapatellar fat pad; lncRNA, long non-coding RNA; MIA, monoiodoacetate; MRI, magnetic resonance imaging; Ms, mouse; mTOR, mammalian target of rapamycin; PT, post-traumatic; TMJ, temporomandibular joint; UC, umbilical cord.
      Table 2Overview of studies using MSC-sEVs for bone regeneration.
      MSC originIn vivo/in vitro modelKey outcomesPathway (EV molecule)Author
      H-EMBRat calvarial defect/Human BM-MSCs, endothelial cells (EA.hy926) and rat alveolar macrophages (NR8383)Enhanced bone regeneration.

      Increased proliferation, angiogenesis, osteogenesis and M2 over M1 macrophage polarization.
      Chuah

      • Chuah SJ
      • Yong CW
      • Teo KYW
      • Chew JRJ
      • Cheow YA
      • Zhang S
      • et al.
      Mesenchymal stromal cell-derived small extracellular vesicles modulate macrophage polarization and enhance angio-osteogenesis to promote bone healing.
      H-BMMouse femur fractureAccelerated fracture healing by enhancing callus formation and bone union._

      (miR-4532, miR-125b-5p, miR-338-3p)
      Furuta
      • Furuta T
      • Miyaki S
      • Ishitobi H
      • Ogura T
      • Kato Y
      • Kamei N
      • et al.
      Mesenchymal Stem Cell-Derived Exosomes Promote Fracture Healing in a Mouse Model.
      H-iPSCRat calvarial defectEnhanced bone regeneration.

      Increased BM-MSC proliferation, migration and osteogenesis.
      PI3K/Akt

      (–)
      Zhang
      • Zhang J
      • Liu X
      • Li H
      • Chen C
      • Hu B
      • Niu X
      • et al.
      Exosomes/tricalcium phosphate combination scaffolds can enhance bone regeneration by activating the PI3K/Akt signaling pathway.
      H-UCRat femur fracture/Neonatal mouse osteoblasts, HUVECsPromoted angiogenesis and bone healing. Enhanced osteoblast differentiation and HUVEC migration and tube formation.

      HIF-1α/VEGF

      (–)
      Zhang
      • Zhang Y
      • Hao Z
      • Wang P
      • Xia Y
      • Wu J
      • Xia D
      • et al.
      Exosomes from human umbilical cord mesenchymal stem cells enhance fracture healing through HIF-1α-mediated promotion of angiogenesis in a rat model of stabilized fracture.
      H-ADRat calvarial defect/LPS/IFN-γ-treated M1 macrophages (U937)

      Increased M2 over M1 macrophage infiltration and promoted bone healing. Promoted polarization of M1 macrophages toward M2 phenotype and suppressed inflammation.miR-451a/MIF

      (miR-451a)
      Li
      • Li R
      • Li D
      • Wang H
      • Chen K
      • Wang S
      • Xu J
      • et al.
      Exosomes from adipose-derived stem cells regulate M1/M2 macrophage phenotypic polarization to promote bone healing via miR-451a/MIF.
      H-BMRat calvarial defect/Human BM-MSCsEnhanced bone regeneration. Promoted BM-MSC migration, osteogenesis and angiogenesis.

      (VEGF)
      Takeuchi
      • Takeuchi R
      • Katagiri W
      • Endo S
      • Kobayashi T.
      Exosomes from conditioned media of bone marrow-derived mesenchymal stem cells promote bone regeneration by enhancing angiogenesis.
      R-BMRat femur fracture/Mouse osteoblasts (MC3T3-E1), HUVECsAccelerated bone healing. Enhanced cell proliferation, migration and HUVEC tube formation.HIF-1α/VEGF

      BMP2/Smad1/RUNX2

      (–)
      Zhang
      • Zhang L
      • Jiao G
      • Ren S
      • Zhang X
      • Li C
      • Wu W
      • et al.
      Exosomes from bone marrow mesenchymal stem cells enhance fracture healing through the promotion of osteogenesis and angiogenesis in a rat model of nonunion.
      H-UCRat calvarial defect/Endothelial progenitor cells

      Promoted bone healing by enhancing angiogenesis and osteogenesis. Enhanced endothelial cell proliferation, migration and tube formation.NOTCH1/DLL4

      (miR-21)
      Zhang
      • Zhang Y
      • Xie Y
      • Hao Z
      • Zhou P
      • Wang P
      • Fang S
      • et al.
      Umbilical Mesenchymal Stem Cell-Derived Exosome-Encapsulated Hydrogels Accelerate Bone Repair by Enhancing Angiogenesis.
      H-BMMouse femur fracturePromoted fracture healing. Enhanced osteogenic differentiation of H-BM-MSCs.WWP1/Smurf2-mediated KLF5/β-catenin

      (miR-19b)
      Huang
      • Huang Y
      • Xu Y
      • Feng S
      • He P
      • Sheng B
      • Ni J.
      miR-19b enhances osteogenic differentiation of mesenchymal stem cells and promotes fracture healing through the WWP1/Smurf2-mediated KLF5/β-catenin signaling pathway.
      Ms-BMMouse femur fracture/Mouse osteoblasts (MC3T3-E1)Promoted fracture healing.

      Enhanced osteoblast proliferation and differentiation
      LRP4/Wnt/β-catenin

      (miR-136-5p)
      Yu
      • Yu H
      • Zhang J
      • Liu X
      • Li Y.
      microRNA-136-5p from bone marrow mesenchymal stem cell-derived exosomes facilitates fracture healing by targeting LRP4 to activate the Wnt/β-catenin pathway.
      Ms-BMMouse femur fracture/HUVECsEnhanced fracture healing. Increased HUVEC proliferation, migration and tube formation.PTEN/PI3K/AKT

      (miR-29b-3p)
      Yang
      • Yang J
      • Gao J
      • Gao F
      • Zhao Y
      • Deng B
      • Mu X
      • et al.
      Extracellular vesicles-encapsulated microRNA-29b-3p from bone marrow-derived mesenchymal stem cells promotes fracture healing via modulation of the PTEN/PI3K/AKT axis.
      H-BMRat calvarial defect/Human osteoblasts (hFOB 1.19)Enhanced bone regeneration.

      Promoted differentiation and proliferation of osteoblasts.


      (miR-196a)
      Qin
      • Qin Y
      • Wang L
      • Gao Z
      • Chen G
      • Zhang C
      Bone marrow stromal/stem cell-derived extracellular vesicles regulate osteoblast activity and differentiation in vitro and promote bone regeneration in vivo.
      AD, adipose; BM, bone marrow; EMB, embryonic; H, human; HUVEC, human umbilical vein endothelial cell; IFN-γ, interferon gamma; iPSC, induced pluripotent stem cell; LPS, lipopolysaccharide; LRP4, low-density lipoprotein receptor-related protein 4; MIF, macrophage migration inhibitory factor; Ms, mouse; PI3K, phosphatidylinositol 3-kinase; PTEN, phosphatase and tensin homolog; R, rat; UC, umbilical cord.
      Table 3Overview of studies using MSC-sEVs for IVD regeneration.
      MSC originIn vivo/in vitro modelKey outcomesPathway (EV molecule)Author
      H-BMRat IVD degeneration/Rat disk explants

      Human NP cells
      Alleviated IVD degeneration with improved disk height, MRI and histological grading. Promoted NP cell migration, proliferation and anabolic activity.Notch1

      (vasorin)
      Liao
      • Liao Z
      • Ke W
      • Liu H
      • Tong B
      • Wang K
      • Feng X
      • et al.
      Vasorin-containing small extracellular vesicles retard intervertebral disc degeneration utilizing an injectable thermoresponsive delivery system.
      H-BMRat IVD degeneration/AGE-induced ER stress in human NP cells

      Alleviated IVD degeneration with improved disk height, MRI and histological grading. Suppressed ER stress-related apoptosis in NP cells.AKT/ERK

      (–)
      Liao
      • Liao Z
      • Luo R
      • Li G
      • Song Y
      • Zhan S
      • Zhao K
      • et al.
      Exosomes from mesenchymal stem cells modulate endoplasmic reticulum stress to protect against nucleus pulposus cell death and ameliorate intervertebral disc degeneration in vivo.
      H-BMRat IVD degeneration/TNF-α-induced apoptosis in human NP cellsReduced NP cell apoptosis and IVD degeneration with improvements in MRI and histology.PTEN/PI3K/AKT

      (miR-21)
      Cheng
      • Cheng X
      • Zhang G
      • Zhang L
      • Hu Y
      • Zhang K
      • Sun X
      • et al.
      Mesenchymal stem cells deliver exogenous miR-21 via exosomes to inhibit nucleus pulposus cell apoptosis and reduce intervertebral disc degeneration.
      Ms-BMIL-1β-induced inflammation and apoptosis in mouse NP cellsSuppressed IL-1β-induced NP cell inflammation and apoptosis.MLK3/MAPK

      (miR-142-3p)
      Zhu
      • Zhu L
      • Shi Y
      • Liu L
      • Wang H
      • Shen P
      • Yang H.
      Mesenchymal stem cells-derived exosomes ameliorate nucleus pulposus cells apoptosis via delivering miR-142-3p: therapeutic potential for intervertebral disc degenerative diseases.
      H-BMMouse IVD degeneration/LPS-induced pyroptosis in mouse NP cellsReduced inflammation and IVD degeneration by inhibiting NLRP3-mediated pyroptosis.NLRP3 inflammasome

      (miR‐410)
      Zhang
      • Zhang J
      • Zhang J
      • Zhang Y
      • Liu W
      • Ni W
      • Huang X
      • et al.
      Mesenchymal stem cells-derived exosomes ameliorate intervertebral disc degeneration through inhibiting pyroptosis.
      H-BMTNF-α-induced apoptosis in human NP cellsSuppressed TNF-α-induced apoptosis in NP cells.miRNA-194-5p/TRAF6

      (miRNA-194-5p)
      Sun
      • Sun Z
      • Tang X
      • Li Q
      • Wang H
      • Sun H
      • Tian J.
      Mesenchymal stem cell extracellular vesicles-derived microRNA-194-5p delays the development of intervertebral disc degeneration by targeting TRAF6.
      Ms-BMRabbit IVD degeneration/H2O2-treated rat NP cellsAlleviated IVD degeneration with improvements in disk height, MRI and histology. Suppressed H2O2-induced inflammation, apoptosis and matrix degradation of NP cells.NLRP3 inflammasome

      (–)
      Xia [
      • Xia C
      • Zeng Z
      • Fang B
      • Tao M
      • Gu C
      • Zheng L
      • et al.
      Mesenchymal stem cell-derived exosomes ameliorate intervertebral disc degeneration via anti-oxidant and anti-inflammatory effects.
      ]
      H-UCHuman NP cellsInhibited METTL14/NLRP3-mediated pyroptosis in NP cells.METTL14/NLRP3 (miR-26a-5p)Yuan
      • Yuan X
      • Li T
      • Shi L
      • Miao J
      • Guo Y
      • Chen Y.
      Human umbilical cord mesenchymal stem cells deliver exogenous miR-26a-5p via exosomes to inhibit nucleus pulposus cell pyroptosis through METTL14/NLRP3.
      H-URat IVD degeneration/Human NP cellsAlleviated IVD degeneration with improved disk height and histological grading. Suppressed NP cell senescence while enhancing proliferation and matrix synthesis.TGF-β/Smad/AKT (matrilin-3)Guo
      • Guo Z
      • Su W
      • Zhou R
      • Zhang G
      • Yang S
      • Wu X
      • et al.
      Exosomal MATN3 of Urine-Derived Stem Cells Ameliorates Intervertebral Disc Degeneration by Antisenescence Effects and Promotes NPC Proliferation and ECM Synthesis by Activating TGF-β.
      AGE, advanced glycation end product; BM, bone marrow; ER, endoplasmic reticulum; ERK, extracellular signal-regulated kinase; H, human; H₂O₂, hydrogen peroxide; LPS, lipopolysaccharide; MAPK, mitogen-activated protein kinase; METTL14, methyltransferase-like 14; MLK3, mixed lineage kinase 3; MRI, magnetic resonance imaging; Ms, mouse; NLRP3, NLR family pyrin domain containing 3; PI3K, phosphatidylinositol 3-kinase; PTEN, phosphatase and tensin homolog; TRAF6, tumor receptor-associated factor 6; U, urine; UC, umbilical cord.
      Fig 1
      Fig. 1Cargo molecules of MSC-sEVs and their associated therapeutic effects in the repair of musculoskeletal tissues. Different colored circles show the suggested cargo proteins and RNAs mediating the immunomodulatory and regenerative effects of MSC-sEVs in the repair of cartilage, bone, IVD, skeletal muscle, tendon and ligament following disease/injury. lncRNA, long non-coding RNA. Image is created with Biorender.com.

      Articular cartilage

      Focal articular cartilage defects

      Being avascularized, articular cartilage has poor intrinsic ability to regenerate upon injury [
      • Hunziker EB.
      Articular cartilage repair: basic science and clinical progress. A review of the current status and prospects.
      ]. Current techniques, such as microfracture and autologous chondrocyte implantation (ACI), often yield an inferior fibrocartilage repair that is prone to degeneration [
      • Makris EA
      • Gomoll AH
      • Malizos KN
      • Hu JC
      • Athanasiou KA.
      Repair and tissue engineering techniques for articular cartilage.
      ]. With ACI, there are also issues of limited tissue availability, donor site morbidity and loss of chondrocyte phenotype upon ex vivo expansion [
      • Makris EA
      • Gomoll AH
      • Malizos KN
      • Hu JC
      • Athanasiou KA.
      Repair and tissue engineering techniques for articular cartilage.
      ,
      • Jiang YZ
      • Zhang SF
      • Qi YY
      • Wang LL
      • Ouyang HW.
      Cell Transplantation for Articular Cartilage Defects: Principles of Past, Present, and Future Practice.
      ].
      In the past few decades, MSCs have emerged as an alternative cell type for cartilage repair. Several clinical studies have demonstrated the safety and efficacy of MSC therapies for cartilage repair [
      • Tan SHS
      • Kwan YT
      • Neo WJ
      • Chong JY
      • Kuek TYJ
      • See JZF
      • et al.
      Intra-articular Injections of Mesenchymal Stem Cells Without Adjuvant Therapies for Knee Osteoarthritis: A Systematic Review and Meta-analysis.
      ]. Recently, a 10-year follow-up study concluded that autologous bone marrow MSC treatment resulted in clinical outcomes equivalent to ACI, with no apparent increased risk of tumor formation [
      • Teo AQA
      • Wong KL
      • Shen L
      • Lim JY
      • Toh WS
      • Lee EH
      • et al.
      Equivalent 10-Year Outcomes After Implantation of Autologous Bone Marrow–Derived Mesenchymal Stem Cells Versus Autologous Chondrocyte Implantation for Chondral Defects of the Knee.
      ]. Based on these findings, it was postulated that MSC-sEVs could recapitulate the therapeutic effects of MSCs in cartilage repair.
      We first demonstrated that weekly intra-articular injections of MSC-sEVs were efficacious in promoting osteochondral regeneration in rats [
      • Zhang S
      • Chu WC
      • Lai RC
      • Lim SK
      • Hui JHP
      • Toh WS.
      Exosomes derived from human embryonic mesenchymal stem cells promote osteochondral regeneration.
      ], and this was mediated through a well-orchestrated mechanism of augmenting cellular proliferation, attenuating apoptosis, increasing matrix synthesis and enhancing anti-inflammatory M2 over pro-inflammatory M1 macrophage infiltration with concomitant suppression of synovial inflammation [
      • Zhang S
      • Chuah SJ
      • Lai RC
      • Hui JHP
      • Lim SK
      • Toh WS.
      MSC exosomes mediate cartilage repair by enhancing proliferation, attenuating apoptosis and modulating immune reactivity.
      ]. Of these EV-mediated cellular activities, MSC-sEVs enhanced chondrocyte proliferation, migration and matrix synthesis, partly through exosomal CD73-mediated adenosine activation of pro-survival signaling pathways; namely, protein kinase B (AKT) and extracellular signal-regulated kinase. Other studies have reported MSC-sEV restoration of mitochondrial function [
      • Chen P
      • Zheng L
      • Wang Y
      • Tao M
      • Xie Z
      • Xia C
      • et al.
      Desktop-stereolithography 3D printing of a radially oriented extracellular matrix/mesenchymal stem cell exosome bioink for osteochondral defect regeneration.
      ] and suppression of apoptosis and senescence in chondrocytes [
      • Yan L
      • Liu G
      • Wu X.
      The umbilical cord mesenchymal stem cell-derived exosomal lncRNA H19 improves osteochondral activity through miR-29b-3p/FoxO3 axis.
      ] relevant for osteochondral repair (Table 1).
      To determine the functional competency of cartilage repair, further studies were performed using larger animal models, including rabbits [
      • Chen P
      • Zheng L
      • Wang Y
      • Tao M
      • Xie Z
      • Xia C
      • et al.
      Desktop-stereolithography 3D printing of a radially oriented extracellular matrix/mesenchymal stem cell exosome bioink for osteochondral defect regeneration.
      ,
      • Wong KL
      • Zhang S
      • Wang M
      • Ren X
      • Afizah H
      • Lai RC
      • et al.
      Intra-Articular Injections of Mesenchymal Stem Cell Exosomes and Hyaluronic Acid Improve Structural and Mechanical Properties of Repaired Cartilage in a Rabbit Model.
      ] and pigs [
      • Zhang S
      • Wong KL
      • Ren X
      • Teo KYW
      • Afizah H
      • Choo ABH
      • et al.
      Mesenchymal Stem Cell Exosomes Promote Functional Osteochondral Repair in a Clinically Relevant Porcine Model.
      ,
      • Hede KTC
      • Christensen BB
      • Olesen ML
      • Thomsen JS
      • Foldager CB
      • Toh WS
      • et al.
      Mesenchymal Stem Cell Extracellular Vesicles as Adjuvant to Bone Marrow Stimulation in Chondral Defect Repair in a Minipig Model.
      ]. As a prelude to a clinical trial, our group recently demonstrated the safety and efficacy of MSC-sEVs for repair of critical-size osteochondral defects in a clinically relevant porcine model [
      • Zhang S
      • Wong KL
      • Ren X
      • Teo KYW
      • Afizah H
      • Choo ABH
      • et al.
      Mesenchymal Stem Cell Exosomes Promote Functional Osteochondral Repair in a Clinically Relevant Porcine Model.
      ]. Specifically, we showed that MSC-sEVs in combination with hyaluronic acid administered at a clinically acceptable frequency of three intra-articular injections at weekly intervals were able to promote functional osteochondral repair at the weight-bearing area of medial femoral condyles, with significantly improved morphological, histological and mechanical outcomes. Notably, the newly repaired tissues associated with MSC-sEV and hyaluronic acid combination treatment yielded mostly hyaline cartilage and exhibited Young modulus and stiffness that approximated those of the native cartilage. In terms of safety assessment, we observed no adverse effects with the use of human MSC-sEVs in immunocompetent animals. This was substantiated by the blood serum analysis, which showed levels of blood urea nitrogen, creatinine, albumin, aspartate aminotransferase, alanine aminotransferase and total bilirubin (for liver and kidney function) within the healthy reference value range for pigs.
      The therapeutic outcome of tissue repair not limited to cartilage repair is likely influenced by several factors, including the defect model, dosage, scaffold, frequency, timing and route of administration that require further investigations. A recent study performed in mini-pigs observed that MSC-sEVs applied in conjunction with bone marrow stimulation failed to enhance cartilage repair over bone marrow stimulation alone, but instead promoted subchondral bone healing [
      • Hede KTC
      • Christensen BB
      • Olesen ML
      • Thomsen JS
      • Foldager CB
      • Toh WS
      • et al.
      Mesenchymal Stem Cell Extracellular Vesicles as Adjuvant to Bone Marrow Stimulation in Chondral Defect Repair in a Minipig Model.
      ]. Thus, there is a need to determine the dosing regimen of MSC-sEVs for optimal cartilage repair under different clinical scenarios.

      Osteoarthritis

      Unlike focal cartilage defects, OA is a degenerative joint disease associated with excruciating pain and degradation of cartilage and subchondral bone with varying degrees of synovitis and damage to other joint structures, including ligaments, menisci and muscles [
      • Loeser RF
      • Goldring SR
      • Scanzello CR
      • Goldring MB.
      Osteoarthritis: A disease of the joint as an organ.
      ]. Several pre-clinical studies have reported the therapeutic efficacy of MSC-EVs in OA [
      • Tan SSH
      • Tjio CKE
      • Wong JRY
      • Wong KL
      • Chew JRJ
      • Hui JHP
      • et al.
      Mesenchymal Stem Cell Exosomes for Cartilage Regeneration: A Systematic Review of Preclinical In Vivo Studies.
      ]. Using a rat model of temporomandibular joint OA, we demonstrated that intra-articular injections of MSC-sEVs alleviated pain and promoted temporomandibular joint repair through a well-coordinated mechanism of attenuating inflammation and enhancing proliferation and matrix synthesis while reducing apoptosis and matrix degradation to restore homeostasis and promote overall repair and regeneration [
      • Zhang S
      • Teo KYW
      • Chuah SJ
      • Lai RC
      • Lim SK
      • Toh WS.
      MSC exosomes alleviate temporomandibular joint osteoarthritis by attenuating inflammation and restoring matrix homeostasis.
      ]. In chondrocyte cultures, MSC-sEVs counteracted the effects of interleukin (IL)-1β on matrix degradation and nitric oxide production, partly through CD73-mediated adenosine activation of pro-survival and homeostatic signaling pathways; namely, AKT, extracellular signal-regulated kinase and adenosine monophosphate-activated protein kinase.
      In addition, the anti-inflammatory molecules, such as transforming growth factor (TGF)-β1, present in MSC-sEVs can also exert protective effects. TGF-β is critical for cartilage maintenance, and lack of TGF-β or abnormal TGF-β signaling results in OA-like changes [
      • Blaney Davidson EN
      • van der Kraan PM
      • van den Berg WB.
      TGF-β and osteoarthritis.
      ]. Given the important role of TGF-β in cartilage homeostasis, the chondroprotective effects of MSCs were attributed to the presence of TGF-β1 in secreted EVs [
      • Ruiz M
      • Toupet K
      • Maumus M
      • Rozier P
      • Jorgensen C
      • Noël D.
      TGFBI secreted by mesenchymal stromal cells ameliorates osteoarthritis and is detected in extracellular vesicles.
      ], and resolution of OA damage in rodents was positively correlated with the EV content of TGF-β [
      • Ruiz M
      • Toupet K
      • Maumus M
      • Rozier P
      • Jorgensen C
      • Noël D.
      TGFBI secreted by mesenchymal stromal cells ameliorates osteoarthritis and is detected in extracellular vesicles.
      ,
      • 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.
      ].
      Other groups have also associated the beneficial effects of MSC-EVs in OA animal models with RNA activity [
      • Liu Y
      • Zou R
      • Wang Z
      • Wen C
      • Zhang F
      • Lin F.
      Exosomal KLF3-AS1 from hMSCs promoted cartilage repair and chondrocyte proliferation in osteoarthritis.
      ,
      • Wu J
      • Kuang L
      • Chen C
      • Yang J
      • Zeng WN
      • Li T
      • 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.
      ,
      • 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.
      ]. Inhibition of miR-100-5p, miR-136-5p or long non-coding RNA KLF3-AS1 was found to attenuate the chondroprotective effects of MSC-EVs against matrix degradation in rodent models of OA [
      • Liu Y
      • Zou R
      • Wang Z
      • Wen C
      • Zhang F
      • Lin F.
      Exosomal KLF3-AS1 from hMSCs promoted cartilage repair and chondrocyte proliferation in osteoarthritis.
      ,
      • Wu J
      • Kuang L
      • Chen C
      • Yang J
      • Zeng WN
      • Li T
      • 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.
      ,
      • 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.
      ]. For example, miR-100-5p-containing MSC-sEVs were found to protect articular cartilage from damage and ameliorate gait abnormality in OA mice, possibly through miR-100-5p-regulated inhibition of the mammalian target of rapamycin/autophagy pathway [
      • Wu J
      • Kuang L
      • Chen C
      • Yang J
      • Zeng WN
      • Li T
      • 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.
      ].

      Bone

      Critical-size bone defects and non-unions

      The ability of bone to regenerate has long been recognized. However, spontaneous healing is limited in many conditions, such as in large defects of bone due to trauma, tumor and non-union. These conditions require transplantation of bone tissue or substitutes. However, current bone grafts are associated with significant drawbacks, including limited availability and donor site morbidity of autografts, risk of pathogen transmission with allografts and inferior healing and poor remodeling with synthetic grafts [
      • Parikh SN.
      Bone graft substitutes: past, present, future.
      ,
      • Salgado AJ
      • Coutinho OP
      • Reis RL.
      Bone tissue engineering: state of the art and future trends.
      ].
      In recent years, several studies have reported the therapeutic effects of MSC-sEVs on bone regeneration in different animal models of bone defects (Table 2) [
      • Chuah SJ
      • Yong CW
      • Teo KYW
      • Chew JRJ
      • Cheow YA
      • Zhang S
      • et al.
      Mesenchymal stromal cell-derived small extracellular vesicles modulate macrophage polarization and enhance angio-osteogenesis to promote bone healing.
      ,
      • Zhang Y
      • Hao Z
      • Wang P
      • Xia Y
      • Wu J
      • Xia D
      • et al.
      Exosomes from human umbilical cord mesenchymal stem cells enhance fracture healing through HIF-1α-mediated promotion of angiogenesis in a rat model of stabilized fracture.
      ,
      • Zhang Y
      • Xie Y
      • Hao Z
      • Zhou P
      • Wang P
      • Fang S
      • et al.
      Umbilical Mesenchymal Stem Cell-Derived Exosome-Encapsulated Hydrogels Accelerate Bone Repair by Enhancing Angiogenesis.
      ,
      • Zhang J
      • Liu X
      • Li H
      • Chen C
      • Hu B
      • Niu X
      • et al.
      Exosomes/tricalcium phosphate combination scaffolds can enhance bone regeneration by activating the PI3K/Akt signaling pathway.
      ,
      • Furuta T
      • Miyaki S
      • Ishitobi H
      • Ogura T
      • Kato Y
      • Kamei N
      • et al.
      Mesenchymal Stem Cell-Derived Exosomes Promote Fracture Healing in a Mouse Model.
      ,
      • Takeuchi R
      • Katagiri W
      • Endo S
      • Kobayashi T.
      Exosomes from conditioned media of bone marrow-derived mesenchymal stem cells promote bone regeneration by enhancing angiogenesis.
      ,
      • Qin Y
      • Wang L
      • Gao Z
      • Chen G
      • Zhang C
      Bone marrow stromal/stem cell-derived extracellular vesicles regulate osteoblast activity and differentiation in vitro and promote bone regeneration in vivo.
      ,
      • Zhang L
      • Jiao G
      • Ren S
      • Zhang X
      • Li C
      • Wu W
      • et al.
      Exosomes from bone marrow mesenchymal stem cells enhance fracture healing through the promotion of osteogenesis and angiogenesis in a rat model of nonunion.
      ,
      • Yang J
      • Gao J
      • Gao F
      • Zhao Y
      • Deng B
      • Mu X
      • et al.
      Extracellular vesicles-encapsulated microRNA-29b-3p from bone marrow-derived mesenchymal stem cells promotes fracture healing via modulation of the PTEN/PI3K/AKT axis.
      ,
      • Huang Y
      • Xu Y
      • Feng S
      • He P
      • Sheng B
      • Ni J.
      miR-19b enhances osteogenic differentiation of mesenchymal stem cells and promotes fracture healing through the WWP1/Smurf2-mediated KLF5/β-catenin signaling pathway.
      ,
      • Li R
      • Li D
      • Wang H
      • Chen K
      • Wang S
      • Xu J
      • et al.
      Exosomes from adipose-derived stem cells regulate M1/M2 macrophage phenotypic polarization to promote bone healing via miR-451a/MIF.
      ,
      • Yu H
      • Zhang J
      • Liu X
      • Li Y.
      microRNA-136-5p from bone marrow mesenchymal stem cell-derived exosomes facilitates fracture healing by targeting LRP4 to activate the Wnt/β-catenin pathway.
      ]. In many animal studies, MSC-sEVs were found to promote new bone formation with supporting vasculature and displayed improved morphological, biomechanical and histological outcomes coupled with positive effects on cell survival, proliferation and migration as well as osteogenesis and angiogenesis [
      • Tan SHS
      • Wong JRY
      • Sim SJY
      • Tjio CKE
      • Wong KL
      • Chew JRJ
      • et al.
      Mesenchymal stem cell exosomes in bone regenerative strategies—a systematic review of preclinical studies.
      ]. Owing to their diverse cargo, MSC-sEVs have been reported to regulate these cellular processes through multiple signaling pathways, including phosphatidylinositol 3-kinase (PI3K)/AKT, BMP/Smad, and Wnt/β-catenin.
      Bone repair and remodeling involve the activation and complex interplay between angiogenic and osteogenic pathways. MSC-sEVs have been shown to enhance angiogenesis in bone healing through angiogenic proteins such as vascular endothelial growth factor (VEGF) [
      • Takeuchi R
      • Katagiri W
      • Endo S
      • Kobayashi T.
      Exosomes from conditioned media of bone marrow-derived mesenchymal stem cells promote bone regeneration by enhancing angiogenesis.
      ] and microRNAs (miRNAs) such as miR-21 [
      • Zhang Y
      • Xie Y
      • Hao Z
      • Zhou P
      • Wang P
      • Fang S
      • et al.
      Umbilical Mesenchymal Stem Cell-Derived Exosome-Encapsulated Hydrogels Accelerate Bone Repair by Enhancing Angiogenesis.
      ] and miRNA-29b-3p [
      • Yang J
      • Gao J
      • Gao F
      • Zhao Y
      • Deng B
      • Mu X
      • et al.
      Extracellular vesicles-encapsulated microRNA-29b-3p from bone marrow-derived mesenchymal stem cells promotes fracture healing via modulation of the PTEN/PI3K/AKT axis.
      ]. For example, Takeuchi et al. [
      • Takeuchi R
      • Katagiri W
      • Endo S
      • Kobayashi T.
      Exosomes from conditioned media of bone marrow-derived mesenchymal stem cells promote bone regeneration by enhancing angiogenesis.
      ] showed that VEGF present in MSC-sEVs was critical for angiogenesis during bone healing. Specifically, rats treated with MSC-sEVs had increased amounts of newly formed bone, whereas rats treated with MSC-sEVs and anti-VEGFA antibody had significantly decreased amounts of newly formed bone, which was even lower than rats treated with saline. In the presence of anti-VEGFA, there was suppressed angiogenesis with decreased numbers of VEGF- and CD31-positive endothelial cells, reduced mobilization of endogenous CD44-positive MSCs and reduced bone formation.
      Other groups have also attributed enhanced osteogenesis by MSC-sEVs to miRNAs such as miR-196a [
      • Qin Y
      • Wang L
      • Gao Z
      • Chen G
      • Zhang C
      Bone marrow stromal/stem cell-derived extracellular vesicles regulate osteoblast activity and differentiation in vitro and promote bone regeneration in vivo.
      ], miR-19b [
      • Huang Y
      • Xu Y
      • Feng S
      • He P
      • Sheng B
      • Ni J.
      miR-19b enhances osteogenic differentiation of mesenchymal stem cells and promotes fracture healing through the WWP1/Smurf2-mediated KLF5/β-catenin signaling pathway.
      ] and miR-136-5p [
      • Yu H
      • Zhang J
      • Liu X
      • Li Y.
      microRNA-136-5p from bone marrow mesenchymal stem cell-derived exosomes facilitates fracture healing by targeting LRP4 to activate the Wnt/β-catenin pathway.
      ]. For instance, Yu et al. [
      • Yu H
      • Zhang J
      • Liu X
      • Li Y.
      microRNA-136-5p from bone marrow mesenchymal stem cell-derived exosomes facilitates fracture healing by targeting LRP4 to activate the Wnt/β-catenin pathway.
      ] showed that miR-136-5p-containing MSC-sEVs promoted mouse fracture healing and osteoblast proliferation and differentiation. Mechanistically, miR-136-5p was found to inhibit the expression of low-density lipoprotein receptor-related protein 4 to activate the Wnt/β-catenin pathway, thereby stimulating bone growth and fracture healing.
      To date, few studies have investigated the immunomodulatory effects of MSC-sEVs on bone repair [
      • Tan SHS
      • Wong JRY
      • Sim SJY
      • Tjio CKE
      • Wong KL
      • Chew JRJ
      • et al.
      Mesenchymal stem cell exosomes in bone regenerative strategies—a systematic review of preclinical studies.
      ]. Our group demonstrated that a single implantation of collagen sponge loaded with MSC-sEVs was sufficient to significantly improve bone healing in a rat calvarial defect model [
      • Chuah SJ
      • Yong CW
      • Teo KYW
      • Chew JRJ
      • Cheow YA
      • Zhang S
      • et al.
      Mesenchymal stromal cell-derived small extracellular vesicles modulate macrophage polarization and enhance angio-osteogenesis to promote bone healing.
      ]. In addition to increased proliferation, vascularization and mineralization, we observed that MSC-sEV treatment enhanced M2 over M1 macrophage infiltration with concomitant suppression of inflammatory cytokines IL-1β and TNF-α to promote bone healing. This preferential M2 over M1 macrophage infiltration could be attributed to the enhanced M2 over M1 macrophage polarization mediated by MSC-sEVs, possibly through anti-inflammatory M2-polarizing cytokines such as IL-10 [
      • Jung M
      • Ma Y
      • Iyer RP
      • DeLeon-Pennell KY
      • Yabluchanskiy A
      • Garrett MR
      • et al.
      IL-10 improves cardiac remodeling after myocardial infarction by stimulating M2 macrophage polarization and fibroblast activation.
      ] and matrix proteins such as collagen VI [
      • Lv D
      • Zhou L
      • Zheng X
      • Hu Y.
      Sustained release of collagen VI potentiates sciatic nerve regeneration by modulating macrophage phenotype.
      ,
      • Chu WC
      • Zhang S
      • Sng TJ
      • Ong YJ
      • Tan WL
      • Ang VY
      • et al.
      Distribution of pericellular matrix molecules in the temporomandibular joint and their chondroprotective effects against inflammation.
      ] present in the EV proteome, although this remains to be demonstrated. With the rich expression of different chemokine receptors, MSC-sEVs could also promote the infiltration of anti-inflammatory M2 macrophages while preventing the migration of pro-inflammatory M1 macrophages through interaction with chemokine ligands expressed on other cells and tissues [
      • Yu B
      • Shao H
      • Su C
      • Jiang Y
      • Chen X
      • Bai L
      • et al.
      Exosomes derived from MSCs ameliorate retinal laser injury partially by inhibition of MCP-1.
      ,
      • Lo Sicco C
      • Reverberi D
      • Balbi C
      • Ulivi V
      • Principi E
      • Pascucci L
      • et al.
      Mesenchymal Stem Cell-Derived Extracellular Vesicles as Mediators of Anti-Inflammatory Effects: Endorsement of Macrophage Polarization.
      ]. More recently, Li et al. [
      • Li R
      • Li D
      • Wang H
      • Chen K
      • Wang S
      • Xu J
      • et al.
      Exosomes from adipose-derived stem cells regulate M1/M2 macrophage phenotypic polarization to promote bone healing via miR-451a/MIF.
      ] attributed the augmented M2 over M1 macrophage polarization by MSC-sEVs during rat cranial bone repair to miR-451a. Using U937 cells, inhibition of miR-451a expression was found to reverse MSC-sEV effects on polarization of macrophages through increased macrophage migration inhibitory factor (MIF) expression.

      Spine

      IVD degeneration

      IVD degeneration is the leading cause of back pain. With an aging population, the prevalence of IVD degeneration is increasing drastically and is estimated to affect >90% of people over 50 years of age [
      • Cheung KM
      • Karppinen J
      • Chan D
      • Ho DW
      • Song YQ
      • Sham P
      • et al.
      Prevalence and pattern of lumbar magnetic resonance imaging changes in a population study of one thousand forty-three individuals.
      ]. Most patients are managed conservatively with physiotherapy and anti-inflammatory/analgesic medication. If conservative management fails, patients may require invasive surgical procedures, such as decompression, fusion or disk replacement. Unfortunately, these approaches only relieve symptoms and do not repair the degenerated IVD.
      MSC therapies offer considerable promise for early intervention of IVD degeneration and less invasive alternatives to spinal surgeries. In pre-clinical studies, transplanted MSCs were found to promote IVD regeneration by enhancing matrix production and increased disk height and hydration [
      • Sakai D
      • Mochida J
      • Iwashina T
      • Hiyama A
      • Omi H
      • Imai M
      • et al.
      Regenerative effects of transplanting mesenchymal stem cells embedded in atelocollagen to the degenerated intervertebral disc.
      ,
      • Miyamoto T
      • Muneta T
      • Tabuchi T
      • Matsumoto K
      • Saito H
      • Tsuji K
      • et al.
      Intradiscal transplantation of synovial mesenchymal stem cells prevents intervertebral disc degeneration through suppression of matrix metalloproteinase-related genes in nucleus pulposus cells in rabbits.
      ]. These pre-clinical data set the stage for multiple clinical trials for this indication [
      • Sun Y
      • Leung VY
      • Cheung KM.
      Clinical trials of intervertebral disc regeneration: current status and future developments.
      ,
      • Kumar H
      • Ha DH
      • Lee EJ
      • Park JH
      • Shim JH
      • Ahn TK
      • et al.
      Safety and tolerability of intradiscal implantation of combined autologous adipose-derived mesenchymal stem cells and hyaluronic acid in patients with chronic discogenic low back pain: 1-year follow-up of a phase I study.
      ,
      • Amirdelfan K
      • Bae H
      • McJunkin T
      • DePalma M
      • Kim K
      • Beckworth WJ
      • et al.
      Allogeneic mesenchymal precursor cells treatment for chronic low back pain associated with degenerative disc disease: a prospective randomized, placebo-controlled 36-month study of safety and efficacy.
      ]. Of note, a multicenter randomized controlled study conducted over a 3-year period in 100 subjects with chronic low back pain associated with moderate degenerative disk disease concluded that intradiskal injection of allogeneic bone marrow MSCs was safe and efficacious against IVD degeneration [
      • Amirdelfan K
      • Bae H
      • McJunkin T
      • DePalma M
      • Kim K
      • Beckworth WJ
      • et al.
      Allogeneic mesenchymal precursor cells treatment for chronic low back pain associated with degenerative disc disease: a prospective randomized, placebo-controlled 36-month study of safety and efficacy.
      ].
      In recent years, several groups have reported the therapeutic efficacy of MSC-sEVs for IVD regeneration in animal models (Table 3) [
      • Liao Z
      • Ke W
      • Liu H
      • Tong B
      • Wang K
      • Feng X
      • et al.
      Vasorin-containing small extracellular vesicles retard intervertebral disc degeneration utilizing an injectable thermoresponsive delivery system.
      ,
      • Zhu L
      • Shi Y
      • Liu L
      • Wang H
      • Shen P
      • Yang H.
      Mesenchymal stem cells-derived exosomes ameliorate nucleus pulposus cells apoptosis via delivering miR-142-3p: therapeutic potential for intervertebral disc degenerative diseases.
      ,
      • Zhang J
      • Zhang J
      • Zhang Y
      • Liu W
      • Ni W
      • Huang X
      • et al.
      Mesenchymal stem cells-derived exosomes ameliorate intervertebral disc degeneration through inhibiting pyroptosis.
      ,
      • Cheng X
      • Zhang G
      • Zhang L
      • Hu Y
      • Zhang K
      • Sun X
      • et al.
      Mesenchymal stem cells deliver exogenous miR-21 via exosomes to inhibit nucleus pulposus cell apoptosis and reduce intervertebral disc degeneration.
      ,
      • Sun Z
      • Tang X
      • Li Q
      • Wang H
      • Sun H
      • Tian J.
      Mesenchymal stem cell extracellular vesicles-derived microRNA-194-5p delays the development of intervertebral disc degeneration by targeting TRAF6.
      ,
      • Liao Z
      • Luo R
      • Li G
      • Song Y
      • Zhan S
      • Zhao K
      • et al.
      Exosomes from mesenchymal stem cells modulate endoplasmic reticulum stress to protect against nucleus pulposus cell death and ameliorate intervertebral disc degeneration in vivo.
      ,
      • Xia C
      • Zeng Z
      • Fang B
      • Tao M
      • Gu C
      • Zheng L
      • et al.
      Mesenchymal stem cell-derived exosomes ameliorate intervertebral disc degeneration via anti-oxidant and anti-inflammatory effects.
      ,
      • Yuan X
      • Li T
      • Shi L
      • Miao J
      • Guo Y
      • Chen Y.
      Human umbilical cord mesenchymal stem cells deliver exogenous miR-26a-5p via exosomes to inhibit nucleus pulposus cell pyroptosis through METTL14/NLRP3.
      ,
      • Guo Z
      • Su W
      • Zhou R
      • Zhang G
      • Yang S
      • Wu X
      • et al.
      Exosomal MATN3 of Urine-Derived Stem Cells Ameliorates Intervertebral Disc Degeneration by Antisenescence Effects and Promotes NPC Proliferation and ECM Synthesis by Activating TGF-β.
      ]. Using a rat disk degeneration model, Liao et al. [
      • Liao Z
      • Ke W
      • Liu H
      • Tong B
      • Wang K
      • Feng X
      • et al.
      Vasorin-containing small extracellular vesicles retard intervertebral disc degeneration utilizing an injectable thermoresponsive delivery system.
      ] demonstrated that hydrogel delivery of MSC-sEVs effectively alleviated disk degeneration, with improvements in disk height, magnetic resonance imaging and histological scores. These beneficial effects of MSC-sEVs in disk degeneration could be attributed to vasorin-enhanced proliferation, migration and matrix synthesis of nucleus pulposus (NP) cells mediated through Notch1 signaling. In another study, MSC-sEV treatment was found to alleviate rat IVD degeneration, possibly through transfer of matrilin-3, which complemented the deficit in matrilin proteins in the degenerated IVDs [
      • Guo Z
      • Su W
      • Zhou R
      • Zhang G
      • Yang S
      • Wu X
      • et al.
      Exosomal MATN3 of Urine-Derived Stem Cells Ameliorates Intervertebral Disc Degeneration by Antisenescence Effects and Promotes NPC Proliferation and ECM Synthesis by Activating TGF-β.
      ]. Consistent with the in vivo observations, MSC-sEVs containing matrilin-3 suppressed NP cell senescence while enhancing proliferation and matrix synthesis in vitro. Other groups have also ascribed the therapeutic effects of MSC-sEVs in animal models of IVD degeneration to miRNAs such as miR-21, miR-142-3p, miR-410, miR-194-5p and miR-26a-5p [
      • Zhu L
      • Shi Y
      • Liu L
      • Wang H
      • Shen P
      • Yang H.
      Mesenchymal stem cells-derived exosomes ameliorate nucleus pulposus cells apoptosis via delivering miR-142-3p: therapeutic potential for intervertebral disc degenerative diseases.
      ,
      • Zhang J
      • Zhang J
      • Zhang Y
      • Liu W
      • Ni W
      • Huang X
      • et al.
      Mesenchymal stem cells-derived exosomes ameliorate intervertebral disc degeneration through inhibiting pyroptosis.
      ,
      • Cheng X
      • Zhang G
      • Zhang L
      • Hu Y
      • Zhang K
      • Sun X
      • et al.
      Mesenchymal stem cells deliver exogenous miR-21 via exosomes to inhibit nucleus pulposus cell apoptosis and reduce intervertebral disc degeneration.
      ,
      • Sun Z
      • Tang X
      • Li Q
      • Wang H
      • Sun H
      • Tian J.
      Mesenchymal stem cell extracellular vesicles-derived microRNA-194-5p delays the development of intervertebral disc degeneration by targeting TRAF6.
      ,
      • Yuan X
      • Li T
      • Shi L
      • Miao J
      • Guo Y
      • Chen Y.
      Human umbilical cord mesenchymal stem cells deliver exogenous miR-26a-5p via exosomes to inhibit nucleus pulposus cell pyroptosis through METTL14/NLRP3.
      ]. For instance, MSC-sEV delivery of miR-21 reduced apoptosis of NP cells and alleviated IVD degeneration by targeting phosphatase and tensin homolog (PTEN) through PI3K/AKT pathway [
      • Cheng X
      • Zhang G
      • Zhang L
      • Hu Y
      • Zhang K
      • Sun X
      • et al.
      Mesenchymal stem cells deliver exogenous miR-21 via exosomes to inhibit nucleus pulposus cell apoptosis and reduce intervertebral disc degeneration.
      ], whereas delivery of miR-142-3p attenuated NP cell apoptosis and IVD degeneration by suppressing mitogen-activated protein kinase (MAPK) signaling by targeting mixed lineage kinase 3 (MLK3) [
      • Zhu L
      • Shi Y
      • Liu L
      • Wang H
      • Shen P
      • Yang H.
      Mesenchymal stem cells-derived exosomes ameliorate nucleus pulposus cells apoptosis via delivering miR-142-3p: therapeutic potential for intervertebral disc degenerative diseases.
      ]. In other studies, miR-140 present in MSC-sEVs mitigated IVD degeneration by inhibiting pyroptosis via the suppression of the NLR family pyrin domain containing 3 (NLRP3)/caspase-1 pathway [
      • Zhang J
      • Zhang J
      • Zhang Y
      • Liu W
      • Ni W
      • Huang X
      • et al.
      Mesenchymal stem cells-derived exosomes ameliorate intervertebral disc degeneration through inhibiting pyroptosis.
      ], whereas miR-194-5p inhibited tumor receptor-associated factor 6 (TRAF6), reducing NP cell apoptosis and attenuating IVD degeneration [
      • Sun Z
      • Tang X
      • Li Q
      • Wang H
      • Sun H
      • Tian J.
      Mesenchymal stem cell extracellular vesicles-derived microRNA-194-5p delays the development of intervertebral disc degeneration by targeting TRAF6.
      ].

      Other musculoskeletal tissue disorders

      Apart from therapeutic applications in cartilage, bone and spinal disorders, there are a rapidly growing number of pre-clinical studies that have explored the use of MSC-sEVs for the treatment of other musculoskeletal tissue injuries/diseases, including skeletal muscle injuries [
      • Nakamura Y
      • Miyaki S
      • Ishitobi H
      • Matsuyama S
      • Nakasa T
      • Kamei N
      • et al.
      Mesenchymal-stem-cell-derived exosomes accelerate skeletal muscle regeneration.
      ,
      • Byun S-E
      • Sim C
      • Chung Y
      • Kim HK
      • Park S
      • Kim DK
      • et al.
      Skeletal Muscle Regeneration by the Exosomes of Adipose Tissue-Derived Mesenchymal Stem Cells.
      ,
      • Cho K-A
      • Choi D-W
      • Kim Y-H
      • Kim J
      • Ryu K-H
      • Woo S-Y.
      Mesenchymal Stem Cell-Derived Exosomes Protect Muscle Loss by miR-145-5p Activity Targeting Activin A Receptors.
      ], meniscus tears [
      • Kawata K
      • Koga H
      • Tsuji K
      • Miyatake K
      • Nakagawa Y
      • Yokota T
      • et al.
      Extracellular vesicles derived from mesenchymal stromal cells mediate endogenous cell growth and migration via the CXCL5 and CXCL6/CXCR2 axes and repair menisci.
      ], growth plate defects [
      • Wong KL
      • Zhang S
      • Tan SSH
      • Cheow YA
      • Lai RC
      • Lim SK
      • et al.
      Mesenchymal Stem Cell Exosomes Promote Growth Plate Repair and Reduce Limb-Length Discrepancy in Young Rats.
      ,
      • Guan P
      • Liu C
      • Xie D
      • Mao S
      • Ji Y
      • Lin Y
      • et al.
      Exosome-loaded extracellular matrix-mimic hydrogel with anti-inflammatory property Facilitates/promotes growth plate injury repair.
      ] and tendon and ligament injuries [
      • Xu T
      • Lin Y
      • Yu X
      • Jiang G
      • Wang J
      • Xu K
      • et al.
      Comparative Effects of Exosomes and Ectosomes Isolated From Adipose-Derived Mesenchymal Stem Cells on Achilles Tendinopathy in a Rat Model.
      ,
      • Yu H
      • Cheng J
      • Shi W
      • Ren B
      • Zhao F
      • Shi Y
      • et al.
      Bone marrow mesenchymal stem cell-derived exosomes promote tendon regeneration by facilitating the proliferation and migration of endogenous tendon stem/progenitor cells.
      ,
      • Chamberlain CS
      • Kink JA
      • Wildenauer LA
      • McCaughey M
      • Henry K
      • Spiker AM
      • et al.
      Exosome-educated macrophages and exosomes differentially improve ligament healing.
      ].

      Skeletal muscle loss

      In a mouse model of skeletal muscle injury induced by cardiotoxin, local injection of MSC-sEVs improved muscle regeneration by enhancing myogenesis and angiogenesis through increased muscle fiber growth and capillary density accompanied by suppression of fibrosis [
      • Nakamura Y
      • Miyaki S
      • Ishitobi H
      • Matsuyama S
      • Nakasa T
      • Kamei N
      • et al.
      Mesenchymal-stem-cell-derived exosomes accelerate skeletal muscle regeneration.
      ]. These effects of MSC-sEVs on myogenesis and angiogenesis could be attributed at least in part to the transfer of specific cargo miRNAs such as miR-494. Using a non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mouse model of skeletal muscle defect, administration of MSC-sEVs in fibrin gel was found to promote skeletal muscle regeneration, with increased expression of myoblast determination protein 1 (MyoD) and myogenin. EV-treated animals also showed relatively preserved shapes and sizes of muscle bundles [
      • Byun S-E
      • Sim C
      • Chung Y
      • Kim HK
      • Park S
      • Kim DK
      • et al.
      Skeletal Muscle Regeneration by the Exosomes of Adipose Tissue-Derived Mesenchymal Stem Cells.
      ]. Additionally, MSC-sEVs have been shown to protect against skeletal muscle loss and atrophy. Using a mouse model of chemotherapy-induced muscle loss, MSC-sEV treatment was found to be effective in recovering body weight and skeletal muscle mass. Mechanistically, MSC-sEV treatment was found to counteract activin A-impaired myogenic differentiation by reducing the expression of activin A receptors ACVR2A and ACVR1B in a miR-145-5p-dependent manner [
      • Cho K-A
      • Choi D-W
      • Kim Y-H
      • Kim J
      • Ryu K-H
      • Woo S-Y.
      Mesenchymal Stem Cell-Derived Exosomes Protect Muscle Loss by miR-145-5p Activity Targeting Activin A Receptors.
      ].

      Growth plate defects

      Using a rat model of growth plate injury, we demonstrated that a single intra-articular injection of MSC-sEVs significantly enhanced physeal repair and reduced limb length discrepancy but did not inhibit bone bridge formation [
      • Wong KL
      • Zhang S
      • Tan SSH
      • Cheow YA
      • Lai RC
      • Lim SK
      • et al.
      Mesenchymal Stem Cell Exosomes Promote Growth Plate Repair and Reduce Limb-Length Discrepancy in Young Rats.
      ]. Notably, EV-treated animals had an approximately 35% reduction in limb length discrepancy compared with vehicle-treated animals. The clinical implications of this reduction in limb length discrepancy include possibly avoiding invasive surgical procedures that are associated with a high risk of complications. Although the exact mechanisms remain to be fully elucidated, the therapeutic effects of MSC-sEVs on growth plate repair could be partly attributed to enhanced M2 over M1 macrophage polarization and increased anabolic activity of chondrocytes against inflammation [
      • Guan P
      • Liu C
      • Xie D
      • Mao S
      • Ji Y
      • Lin Y
      • et al.
      Exosome-loaded extracellular matrix-mimic hydrogel with anti-inflammatory property Facilitates/promotes growth plate injury repair.
      ].

      Tendon and ligament injuries

      MSC-sEVs have been explored in several studies for the treatment of tendon and ligament injuries [
      • Xu T
      • Lin Y
      • Yu X
      • Jiang G
      • Wang J
      • Xu K
      • et al.
      Comparative Effects of Exosomes and Ectosomes Isolated From Adipose-Derived Mesenchymal Stem Cells on Achilles Tendinopathy in a Rat Model.
      ,
      • Yu H
      • Cheng J
      • Shi W
      • Ren B
      • Zhao F
      • Shi Y
      • et al.
      Bone marrow mesenchymal stem cell-derived exosomes promote tendon regeneration by facilitating the proliferation and migration of endogenous tendon stem/progenitor cells.
      ,
      • Chamberlain CS
      • Kink JA
      • Wildenauer LA
      • McCaughey M
      • Henry K
      • Spiker AM
      • et al.
      Exosome-educated macrophages and exosomes differentially improve ligament healing.
      ]. For instance, Yu et al. [
      • Yu H
      • Cheng J
      • Shi W
      • Ren B
      • Zhao F
      • Shi Y
      • et al.
      Bone marrow mesenchymal stem cell-derived exosomes promote tendon regeneration by facilitating the proliferation and migration of endogenous tendon stem/progenitor cells.
      ] reported that MSC-sEVs embedded in fibrin significantly improved histological scores and enhanced the expression of tendon-related markers such as mohawk, tenomodulin and type I collagen, as well as mechanical properties of the neotendon. These beneficial effects of MSC-sEVs on tendon regeneration could be attributed to enhanced proliferation, migration and tenogenic differentiation of tendon stem/progenitor cells in vitro. Similarly, MSC-sEVs have been shown to enhance ligament healing in a rat medial collateral ligament injury model by reducing scar size, upregulating type I and III collagen expression and increasing collagen production and organization [
      • Chamberlain CS
      • Kink JA
      • Wildenauer LA
      • McCaughey M
      • Henry K
      • Spiker AM
      • et al.
      Exosome-educated macrophages and exosomes differentially improve ligament healing.
      ]. More recently, using a rat model of Achilles tendinopathy, Xu et al. [
      • Xu T
      • Lin Y
      • Yu X
      • Jiang G
      • Wang J
      • Xu K
      • et al.
      Comparative Effects of Exosomes and Ectosomes Isolated From Adipose-Derived Mesenchymal Stem Cells on Achilles Tendinopathy in a Rat Model.
      ] reported that MSC-EVs with a size of approximately 150 nm had superior efficacy in tendon repair over their EV counterpart with a size of approximately 230 nm, as evidenced by improved histological scores, lower fibril density, larger collagen diameter and better biomechanical properties. Further miRNA profiling analysis of the approximately 150-nm MSC-sEVs revealed enrichment of miRNAs such as miR-29a, miR-21-5p and miR-148a-3p, which have been reported to improve tendon healing [
      • Xu T
      • Lin Y
      • Yu X
      • Jiang G
      • Wang J
      • Xu K
      • et al.
      Comparative Effects of Exosomes and Ectosomes Isolated From Adipose-Derived Mesenchymal Stem Cells on Achilles Tendinopathy in a Rat Model.
      ,
      • Liu Q
      • Zhu Y
      • Zhu W
      • Zhang G
      • Yang YP
      • Zhao C.
      The role of MicroRNAs in tendon injury, repair, and related tissue engineering.
      ].

      Diverse Cargo of MSC-sEVs

      As exemplified by several of the aforementioned studies, many musculoskeletal disorders display common pathological features of inflammation, oxidative stress, matrix degradation and apoptosis. Owing to their diverse cargo, MSC-sEVs have the ability to mount a multifaceted mechanism of action (MoA) to elicit biological effects against these diverse pathological processes. However, elucidating the MoA of MSC-sEV preparations against an injury/disease remains challenging. In elucidating a multifaceted MoA where multiple pathological processes are modulated, only processes mediated directly by MSC-sEVs should be mapped to the specific attributes in the bioactive cargo of the MSC-sEV preparation. Currently, the MoA underpinning the therapeutic efficacy of MSC-sEVs is largely attributed to the cargo of RNA or proteins. On this note, the candidate RNA or protein must be present in the EVs in functional competent configuration and concentration to elicit the activity [
      • Toh WS
      • Lai RC
      • Zhang B
      • Lim SK.
      MSC exosome works through a protein-based mechanism of action.
      ].

      miRNA- versus Protein-Based MoA

      In assessing the role of miRNAs in mediating the therapeutic effects of MSC-sEVs, it is imperative to note that miRNAs are only functional when associated with RNA-inducing silencing complexes (RISCs), and only pre-miRNA can be loaded onto RISCs. Despite several reports on miRNAs mediating the therapeutic effects of MSC-sEVs in the repair of musculoskeletal tissues, few had determined whether the miRNAs were pre-miRNAs or RISC-loaded mature miRNAs to be functional. A search of the publicly accessible databases ExoCarta (http://www.exocarta.org/) and Vesiclepedia (http://www.microvesicles.org/) revealed that RISC components, including Dicer and Argonaute, are not frequently present in EVs. Of the 41,860 protein entries in ExoCarta, there were only seven entries for Argonaute and none for Dicer. Similarly, of the 349,988 protein entries in Vesiclepedia, there was only one entry for Argonaute and two entries for Dicer. Interestingly, none of these entries were for MSC-EVs. This is consistent with an earlier report that Dicer and Argonaute are not present in MSC-sEVs [
      • Chen TS
      • Lai RC
      • Lee MM
      • Choo AB
      • Lee CN
      • Lim SK.
      Mesenchymal stem cell secretes microparticles enriched in pre-microRNAs.
      ]. Based on published data, we previously calculated that one MSC-sEV contains about 1.3 miRNA molecules [
      • Toh WS
      • Lai RC
      • Zhang B
      • Lim SK.
      MSC exosome works through a protein-based mechanism of action.
      ]. With a large cargo of more than 100 miRNAs in MSC-sEVs, the probability of a sEV having a specific miRNA is about one in 100, and consequently, one cell would have to take up 100 sEVs for one specific miRNA molecule. Based on this probability, uptake of an miRNA molecule is likely not sufficient to elicit a cellular response. Our findings are consistent with an earlier study that tested EVs from diverse sources, including plasma, seminal fluid, dendritic cells, mast cells and ovarian cells, and observed far less than one molecule of a given miRNA per EV [
      • Chevillet JR
      • Kang Q
      • Ruf IK
      • Briggs HA
      • Vojtech LN
      • Hughes SM
      • et al.
      Quantitative and stoichiometric analysis of the microRNA content of exosomes.
      ]. Similarly, in EVs isolated from human lymphoblastoid B-cell lines latently infected with Epstein–Barr virus, one miRNA molecule in 300–16,000 EVs was observed [
      • Albanese M
      • Chen YA
      • Hüls C
      • Gärtner K
      • Tagawa T
      • Mejias-Perez E
      • et al.
      MicroRNAs are minor constituents of extracellular vesicles that are rarely delivered to target cells.
      ]. Increasing the doses of EVs engineered to fuse and deliver their miRNA cargoes to HEK293T cells had no measurable effect on the target mRNAs [
      • Albanese M
      • Chen YA
      • Hüls C
      • Gärtner K
      • Tagawa T
      • Mejias-Perez E
      • et al.
      MicroRNAs are minor constituents of extracellular vesicles that are rarely delivered to target cells.
      ].
      By contrast, many proteins in MSC-sEVs are enzymes or ligands for receptors. For example, the enzymatic activity of two adenosine triphosphate (ATP)-generating glycolytic enzymes present in our MSC-sEV preparation, namely, phosphoglycerate kinase (PGK) and pyruvate kinase m2 (PKm2) were determined to be 3.59 × 10−3 U/µg EV protein and 5.5 × 10−3 U/µg EV protein, respectively. Based on the assumption that one cell has approximately 2–6 × 10−8 µmol ATP, 1 µg MSC-sEVs can generate in 1 min the same amount of ATP as in 1.5–4.6 × 105 cells. Alternatively, 1 µg MSC-sEVs can meet the ATP consumption requirements of 1 × 105 cells/s [
      • Toh WS
      • Lai RC
      • Zhang B
      • Lim SK.
      MSC exosome works through a protein-based mechanism of action.
      ]. Based on these considerations, we previously proposed that MSC-sEVs are more likely to work through a protein-based MoA.
      There are also bioactive lipids and metabolites present in the diverse cargo of MSC-sEVs. Lipidomic analysis verified the presence of bioactive lipids such as glycolipids, sphingomyelin, free fatty acids and cardiolipins in MSC-EVs [
      • Vallabhaneni KC
      • Penfornis P
      • Dhule S
      • Guillonneau F
      • Adams KV
      • Yuan Mo Y
      • et al.
      Extracellular vesicles from bone marrow mesenchymal stem/stromal cells transport tumor regulatory microRNA, proteins, and metabolites.
      ,
      • Haraszti RA
      • Didiot M-C
      • Sapp E
      • Leszyk J
      • Shaffer SA
      • Rockwell HE
      • et al.
      High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources.
      ], whereas metabolite assays identified the presence of metabolites such as lactic acid and glutamic acid [
      • Vallabhaneni KC
      • Penfornis P
      • Dhule S
      • Guillonneau F
      • Adams KV
      • Yuan Mo Y
      • et al.
      Extracellular vesicles from bone marrow mesenchymal stem/stromal cells transport tumor regulatory microRNA, proteins, and metabolites.
      ]. However, considering the molecular complexity of EVs, it remains challenging to attribute the effects of EVs to a specific lipid or metabolite [
      • Skotland T
      • Sagini K
      • Sandvig K
      • Llorente A.
      An emerging focus on lipids in extracellular vesicles.
      ].

      Clinical Translation and Challenges

      The promising therapeutic efficacy of MSC-sEVs in several pre-clinical studies has certainly increased the excitement regarding their translation to the clinic. To date, based on the search terms “extracellular vesicles” and “exosomes” (https://clinicaltrials.gov/; accessed on Oct 18, 2022), of 411 registered clinical trials using EVs for various diagnostic and therapeutic applications, 37 involve EVs derived from native MSCs. There are presently four clinical trials using MSC-EVs as therapeutics for various musculoskeletal injuries/diseases (Table 4). For example, a phase 1 trial (NCT05060107) will be conducted to evaluate the safety of EVs derived from allogeneic MSCs in the knees of patients with mild to moderate OA. Ten patients are expected to be enrolled and follow-up will be up to 12 months.
      Table 4Registered clinical trials using MSC-EVs/exosomes for musculoskeletal tissue repair.
      NCT numberTitleStatus
      NCT05060107Intra-articular Injection of MSC-derived Exosomes in Knee Osteoarthritis (ExoOA-1) (ExoOA-1)Not yet recruiting
      NCT05261360Clinical Efficacy of Exosome in Degenerative Meniscal Injury (KNEEXO)Recruiting
      NCT04223622Effects of ASC Secretome on Human Osteochondral Explants (ASC-OA)Recruiting
      NCT05520125Treatment of Patients With Bone Tissue Defects Using Mesenchymal Stem Cells Enriched by Extracellular VesiclesNot yet recruiting
      Despite the growing interest in MSC-EVs, the field is still in its infancy, and there are still several challenges that need to be addressed to achieve optimal translation to the clinic. Technical challenges represent a significant issue. Isolation and characterization of EVs and their subtypes are presently hampered by the lack of appropriate technologies and instrumentation to isolate and analyze EVs at nanoscale resolution. The scaling up of cell cultures for clinical manufacture of EVs is also challenging, as this requires optimizing the bioreactor system, defining the culture conditions and implementing the appropriate in-process controls to monitor and control the process [
      • Reiner AT
      • Witwer KW
      • van Balkom BWM
      • de Beer J
      • Brodie C
      • Corteling RL
      • et al.
      Concise Review: Developing Best-Practice Models for the Therapeutic Use of Extracellular Vesicles.
      ,
      • Gimona M
      • Pachler K
      • Laner-Plamberger S
      • Schallmoser K
      • Rohde E
      Manufacturing of Human Extracellular Vesicle-Based Therapeutics for Clinical Use.
      ].
      Although the International Society for Extracellular Vesicles has recommended use of the collective term “EVs” to circumvent the lack of definitive markers and techniques for purifying a specific EV subtype according to their biogenesis pathway, there are still issues of EV heterogeneity that EV preparations derived from different MSC sources, culture conditions, and isolation protocols are likely to contain different EV subtypes with varying composition and properties. To overcome this and other limitations, immortalizing primary MSCs and establishing monoclonal MSC lines have been proposed and demonstrated to be a feasible strategy for reducing the heterogeneity of MSCs and their derived EVs. In this regard, Chen et al. [
      • Chen TS
      • Arslan F
      • Yin Y
      • Tan SS
      • Lai RC
      • Choo ABH
      • et al.
      Enabling a robust scalable manufacturing process for therapeutic exosomes through oncogenic immortalization of human ESC-derived MSCs.
      ] previously reported the transformation of human embryonic stem cell-derived MSCs using the MYC gene to immortalize the MSCs and generate the clonal cell line E1-MYC. The immortalized E1-MYC human embryonic stem cell-derived MSCs grew faster and had increased telomerase activity while retaining the parental karyotype. The produced EVs were therapeutically similar to those derived from the parental MSCs in their ability to reduce infarct size in a mouse model of myocardial ischemia–reperfusion injury. Importantly, EVs produced by these immortalized MSCs did not promote tumor growth in a mouse model of tumor xenograft, indicating that immortalization of MSCs for EV production is a viable strategy for large-scale production of safe EV preparations for therapeutic use [
      • Tan TT
      • Lai RC
      • Padmanabhan J
      • Sim WK
      • Choo ABH
      • Lim SK.
      Assessment of Tumorigenic Potential in Mesenchymal-Stem/Stromal-Cell-Derived Small Extracellular Vesicles (MSC-sEV).
      ].
      Other parameters, such as culture conditions (i.e., culture medium, supplements, culture systems, oxygen concentration), may also affect EV yield and quality. Controlling these parameters in an EV manufacturing process is therefore necessary but remains challenging unless there are quality control metrics linked to a clearly defined MoA to guide and monitor the process. Consequently, several key identity and potency features of MSC-sEV preparations have been proposed as quality control metrics for measurement to ensure quality and batch reproducibility of the EV product before clinical testing or use. For more information on the proposed identity and potency metrics of MSC-sEVs, we refer readers to the recent reviews [
      • Witwer KW
      • Van Balkom BWM
      • Bruno S
      • Choo A
      • Dominici M
      • Gimona M
      • et al.
      Defining mesenchymal stromal cell (MSC)-derived small extracellular vesicles for therapeutic applications.
      ,
      • Gimona M
      • Brizzi MF
      • Choo ABH
      • Dominici M
      • Davidson SM
      • Grillari J
      • et al.
      Critical considerations for the development of potency tests for therapeutic applications of mesenchymal stromal cell-derived small extracellular vesicles.
      ].
      Several pre-clinical studies have demonstrated the therapeutic efficacy of MSC-sEVs for musculoskeletal tissue repair, at least for cartilage and bone defects, OA and IVD degeneration. However, most of these studies were performed using small animal models, which have very different sizes, biomechanics and healing capacities compared with humans. To enable clinical translation, clinically relevant large animal models would be necessary [
      • Chu CR
      • Szczodry M
      • Bruno S.
      Animal models for cartilage regeneration and repair.
      ].
      Another challenge involves optimization of the MSC-sEV dosing regimen. Variables such as dosage, frequency, timing, use of scaffold, and route of administration could affect the therapeutic outcome and remain to be systematically optimized for different injuries/diseases under different clinical scenarios. To optimize the dosing regimen, bioavailability and biodistribution studies should be performed but are currently limited by the lack of sensitive and specific labels. For example, lipophilic dyes (e.g., PKH26) that have been widely used for labeling cells and their EVs are frequently associated with labeling artifacts [
      • Simonsen JB.
      Pitfalls associated with lipophilic fluorophore staining of extracellular vesicles for uptake studies.
      ]. For large musculoskeletal tissue defects, MSC-sEVs are commonly used in combination with biomaterial scaffolds such as collagen sponges. However, biomaterial scaffolds could have profound effects on EV release, integrity and bioactivity that remain to be investigated, and optimal combinations of MSC-sEVs and biomaterial scaffolds should ideally be determined for each application.

      Conclusion

      Despite the ongoing challenges, significant advances have been made in the field of therapeutic MSC-EVs in recent years. Notably, an increasing number of pre-clinical studies have demonstrated the therapeutic potential of MSC-sEVs in various musculoskeletal disorders. To translate this potential to therapeutic applications, technical advancements in EV isolation and characterization will be required to define the identity and potency of MSC-sEV preparations. To elucidate the MoA of MSC-sEVs against an injury/disease, the pathological processes modulated by MSC-sEVs should be mapped to the MSC-sEV cargo molecule(s) as specific attribute(s), where critical considerations, including the biologically relevant concentration and functional configuration of the molecule(s), should be made.

      Funding

      This work was supported by the Ministry of Education (MOE) Tier 2 grant (Project ID: MOE-T2EP30122-0008), MOE Tier 1 grant (Project ID: NUHSRO/2022/091/T1/Seed-Sep/04). KLW, DHWH, and WST acknowledge support from the National Medical Research Council Singapore (TA21nov-0018, CIRG20nov-0016, and CNIG21nov-0009). JHPH and WST have received research funding from the Agency for Science, Technology and Research Singapore under Health and Biomedical Sciences Industry Alignment Fund Pre-Positioning (grant no. H19H6a0026).

      Declaration of Competing Interest

      WST is a scientific advisory board member of Paracrine Therapeutics.

      Author Contributions

      Conception and design of the study: WST. Acquisition of data: KYWT, RT and KLW. Analysis and interpretation of data: KYWT, RT, KLW, DHWH and JHPH. Drafting or revising the manuscript: KYWT, RT and WST. All authors have approved the final article.

      References

        • James SL
        • Abate D
        • Abate KH
        • Abay SM
        • Abbafati C
        • Abbasi N
        • et al.
        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.
        The Lancet. 2018; 392: 1789-1858
        • Hunter DJ
        • March L
        • Chew M.
        Osteoarthritis in 2020 and beyond: a Lancet Commission.
        Lancet. 2020; 396: 1711-1712
        • Pittenger MF
        • Mackay AM
        • Beck SC
        • Jaiswal RK
        • Douglas R
        • Mosca JD
        • et al.
        Multilineage potential of adult human mesenchymal stem cells.
        Science. 1999; 284: 143-147
        • Dominici M
        • Le Blanc K
        • Mueller I
        • Slaper-Cortenbach I
        • Marini F
        • Krause D
        • et al.
        Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement.
        Cytotherapy. 2006; 8: 315-317
        • Théry C
        • Ostrowski M
        • Segura E.
        Membrane vesicles as conveyors of immune responses.
        Nature Reviews Immunology. 2009; 9: 581-593
        • Tóth EÁ
        • Turiák L
        • Visnovitz T
        • Cserép C
        • Mázló A
        • Sódar BW
        • et al.
        Formation of a protein corona on the surface of extracellular vesicles in blood plasma.
        Journal of Extracellular Vesicles. 2021; 10: e12140
        • Reiner AT
        • Witwer KW
        • van Balkom BWM
        • de Beer J
        • Brodie C
        • Corteling RL
        • et al.
        Concise Review: Developing Best-Practice Models for the Therapeutic Use of Extracellular Vesicles.
        Stem Cells Transl Med. 2017; 6: 1730-1739
        • Witwer KW
        • Van Balkom BWM
        • Bruno S
        • Choo A
        • Dominici M
        • Gimona M
        • et al.
        Defining mesenchymal stromal cell (MSC)-derived small extracellular vesicles for therapeutic applications.
        Journal of Extracellular Vesicles. 2019; 81609206
        • Théry C
        • Witwer KW
        • Aikawa E
        • Alcaraz MJ
        • Anderson JD
        • Andriantsitohaina R
        • 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.
        Journal of Extracellular Vesicles. 2018; 71535750
        • Ghannam S
        • Bouffi C
        • Djouad F
        • Jorgensen C
        • Noël D.
        Immunosuppression by mesenchymal stem cells: mechanisms and clinical applications.
        Stem Cell Res Ther. 2010; 1: 2
        • Bruno S
        • Grange C
        • Deregibus MC
        • Calogero RA
        • Saviozzi S
        • Collino F
        • et al.
        Mesenchymal Stem Cell-Derived Microvesicles Protect Against Acute Tubular Injury.
        Journal of the American Society of Nephrology. 2009; 20: 1053
        • Lai RC
        • Arslan F
        • Lee MM
        • Sze NSK
        • Choo A
        • Chen TS
        • et al.
        Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury.
        Stem Cell Research. 2010; 4: 214-222
        • Doeppner TR
        • Herz J
        • Görgens A
        • Schlechter J
        • Ludwig AK
        • Radtke S
        • et al.
        Extracellular Vesicles Improve Post-Stroke Neuroregeneration and Prevent Postischemic Immunosuppression.
        Stem Cells Transl Med. 2015; 4: 1131-1143
        • HE J
        • WANG Y
        • SUN S
        • YU M
        • WANG C
        • PEI X
        • et al.
        Bone marrow stem cells-derived microvesicles protect against renal injury in the mouse remnant kidney model.
        Nephrology. 2012; 17: 493-500
        • Bruno S
        • Tapparo M
        • Collino F
        • Chiabotto G
        • Deregibus MC
        • Soares Lindoso R
        • et al.
        Renal Regenerative Potential of Different Extracellular Vesicle Populations Derived from Bone Marrow Mesenchymal Stromal Cells.
        Tissue Eng Part A. 2017; 23: 1262-1273
        • Xu T
        • Lin Y
        • Yu X
        • Jiang G
        • Wang J
        • Xu K
        • et al.
        Comparative Effects of Exosomes and Ectosomes Isolated From Adipose-Derived Mesenchymal Stem Cells on Achilles Tendinopathy in a Rat Model.
        Am J Sports Med. 2022; 50: 2740-2752
        • Toh WS
        • Lai RC
        • Hui JHP
        • Lim SK.
        MSC exosome as a cell-free MSC therapy for cartilage regeneration: Implications for osteoarthritis treatment.
        Seminars in Cell & Developmental Biology. 2017; 67: 56-64
        • Chen TS
        • Arslan F
        • Yin Y
        • Tan SS
        • Lai RC
        • Choo ABH
        • et al.
        Enabling a robust scalable manufacturing process for therapeutic exosomes through oncogenic immortalization of human ESC-derived MSCs.
        Journal of Translational Medicine. 2011; 9: 47
        • Hunziker EB.
        Articular cartilage repair: basic science and clinical progress. A review of the current status and prospects.
        Osteoarthritis and Cartilage. 2002; 10: 432-463
        • Makris EA
        • Gomoll AH
        • Malizos KN
        • Hu JC
        • Athanasiou KA.
        Repair and tissue engineering techniques for articular cartilage.
        Nat Rev Rheumatol. 2015; 11: 21-34
        • Jiang YZ
        • Zhang SF
        • Qi YY
        • Wang LL
        • Ouyang HW.
        Cell Transplantation for Articular Cartilage Defects: Principles of Past, Present, and Future Practice.
        Cell Transplantation. 2011; 20: 593-607
        • Tan SHS
        • Kwan YT
        • Neo WJ
        • Chong JY
        • Kuek TYJ
        • See JZF
        • et al.
        Intra-articular Injections of Mesenchymal Stem Cells Without Adjuvant Therapies for Knee Osteoarthritis: A Systematic Review and Meta-analysis.
        The American Journal of Sports Medicine. 2021; 49: 3113-3124
        • Teo AQA
        • Wong KL
        • Shen L
        • Lim JY
        • Toh WS
        • Lee EH
        • et al.
        Equivalent 10-Year Outcomes After Implantation of Autologous Bone Marrow–Derived Mesenchymal Stem Cells Versus Autologous Chondrocyte Implantation for Chondral Defects of the Knee.
        The American Journal of Sports Medicine. 2019; 47: 2881-2887
        • Zhang S
        • Chu WC
        • Lai RC
        • Lim SK
        • Hui JHP
        • Toh WS.
        Exosomes derived from human embryonic mesenchymal stem cells promote osteochondral regeneration.
        Osteoarthritis and Cartilage. 2016; 24: 2135-2140
        • Zhang S
        • Chuah SJ
        • Lai RC
        • Hui JHP
        • Lim SK
        • Toh WS.
        MSC exosomes mediate cartilage repair by enhancing proliferation, attenuating apoptosis and modulating immune reactivity.
        Biomaterials. 2018; 156: 16-27
        • Chen P
        • Zheng L
        • Wang Y
        • Tao M
        • Xie Z
        • Xia C
        • 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
        • Yan L
        • Liu G
        • Wu X.
        The umbilical cord mesenchymal stem cell-derived exosomal lncRNA H19 improves osteochondral activity through miR-29b-3p/FoxO3 axis.
        Clin Transl Med. 2021; 11: e255
        • Wong KL
        • Zhang S
        • Wang M
        • Ren X
        • Afizah H
        • Lai RC
        • et al.
        Intra-Articular Injections of Mesenchymal Stem Cell Exosomes and Hyaluronic Acid Improve Structural and Mechanical Properties of Repaired Cartilage in a Rabbit Model.
        Arthroscopy: The Journal of Arthroscopic & Related Surgery. 2020; 36 (2215-28.e2)
        • Zhang S
        • Wong KL
        • Ren X
        • Teo KYW
        • Afizah H
        • Choo ABH
        • et al.
        Mesenchymal Stem Cell Exosomes Promote Functional Osteochondral Repair in a Clinically Relevant Porcine Model.
        The American Journal of Sports Medicine. 2022; 50: 788-800
        • Hede KTC
        • Christensen BB
        • Olesen ML
        • Thomsen JS
        • Foldager CB
        • Toh WS
        • et al.
        Mesenchymal Stem Cell Extracellular Vesicles as Adjuvant to Bone Marrow Stimulation in Chondral Defect Repair in a Minipig Model.
        Cartilage. 2021; 13: 254S-266S
        • Loeser RF
        • Goldring SR
        • Scanzello CR
        • Goldring MB.
        Osteoarthritis: A disease of the joint as an organ.
        Arthritis & Rheumatism. 2012; 64: 1697-1707
        • Tan SSH
        • Tjio CKE
        • Wong JRY
        • Wong KL
        • Chew JRJ
        • Hui JHP
        • et al.
        Mesenchymal Stem Cell Exosomes for Cartilage Regeneration: A Systematic Review of Preclinical In Vivo Studies.
        Tissue Eng Part B Rev. 2021; 27: 1-13
        • Zhang S
        • Teo KYW
        • Chuah SJ
        • Lai RC
        • Lim SK
        • Toh WS.
        MSC exosomes alleviate temporomandibular joint osteoarthritis by attenuating inflammation and restoring matrix homeostasis.
        Biomaterials. 2019; 200: 35-47
        • Blaney Davidson EN
        • van der Kraan PM
        • van den Berg WB.
        TGF-β and osteoarthritis.
        Osteoarthritis and Cartilage. 2007; 15: 597-604
        • Ruiz M
        • Toupet K
        • Maumus M
        • Rozier P
        • Jorgensen C
        • Noël D.
        TGFBI secreted by mesenchymal stromal cells ameliorates osteoarthritis and is detected in extracellular vesicles.
        Biomaterials. 2020; 226119544
        • 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
        • Liu Y
        • Zou R
        • Wang Z
        • Wen C
        • Zhang F
        • Lin F.
        Exosomal KLF3-AS1 from hMSCs promoted cartilage repair and chondrocyte proliferation in osteoarthritis.
        Biochemical Journal. 2018; 475: 3629-3638
        • Wu J
        • Kuang L
        • Chen C
        • Yang J
        • Zeng WN
        • Li T
        • 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
        • 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 Research & Therapy. 2020; 22: 256
        • Parikh SN.
        Bone graft substitutes: past, present, future.
        J Postgrad Med. 2002; 48: 142-148
        • Salgado AJ
        • Coutinho OP
        • Reis RL.
        Bone tissue engineering: state of the art and future trends.
        Macromol Biosci. 2004; 4: 743-765
        • Chuah SJ
        • Yong CW
        • Teo KYW
        • Chew JRJ
        • Cheow YA
        • Zhang S
        • et al.
        Mesenchymal stromal cell-derived small extracellular vesicles modulate macrophage polarization and enhance angio-osteogenesis to promote bone healing.
        Genes & Diseases. 2022; 9: 841-844
        • Zhang Y
        • Hao Z
        • Wang P
        • Xia Y
        • Wu J
        • Xia D
        • et al.
        Exosomes from human umbilical cord mesenchymal stem cells enhance fracture healing through HIF-1α-mediated promotion of angiogenesis in a rat model of stabilized fracture.
        Cell Proliferation. 2019; 52: e12570
        • Zhang Y
        • Xie Y
        • Hao Z
        • Zhou P
        • Wang P
        • Fang S
        • et al.
        Umbilical Mesenchymal Stem Cell-Derived Exosome-Encapsulated Hydrogels Accelerate Bone Repair by Enhancing Angiogenesis.
        ACS Applied Materials & Interfaces. 2021; 13: 18472-18487
        • Zhang J
        • Liu X
        • Li H
        • Chen C
        • Hu B
        • Niu X
        • et al.
        Exosomes/tricalcium phosphate combination scaffolds can enhance bone regeneration by activating the PI3K/Akt signaling pathway.
        Stem Cell Res Ther. 2016; 7: 136
        • Furuta T
        • Miyaki S
        • Ishitobi H
        • Ogura T
        • Kato Y
        • Kamei N
        • et al.
        Mesenchymal Stem Cell-Derived Exosomes Promote Fracture Healing in a Mouse Model.
        Stem Cells Transl Med. 2016; 5: 1620-1630
        • Takeuchi R
        • Katagiri W
        • Endo S
        • Kobayashi T.
        Exosomes from conditioned media of bone marrow-derived mesenchymal stem cells promote bone regeneration by enhancing angiogenesis.
        PLoS One. 2019; 14e0225472
        • Qin Y
        • Wang L
        • Gao Z
        • Chen G
        • Zhang C
        Bone marrow stromal/stem cell-derived extracellular vesicles regulate osteoblast activity and differentiation in vitro and promote bone regeneration in vivo.
        Sci Rep. 2016; 6: 21961
        • Zhang L
        • Jiao G
        • Ren S
        • Zhang X
        • Li C
        • Wu W
        • et al.
        Exosomes from bone marrow mesenchymal stem cells enhance fracture healing through the promotion of osteogenesis and angiogenesis in a rat model of nonunion.
        Stem Cell Res Ther. 2020; 11: 38
        • Yang J
        • Gao J
        • Gao F
        • Zhao Y
        • Deng B
        • Mu X
        • et al.
        Extracellular vesicles-encapsulated microRNA-29b-3p from bone marrow-derived mesenchymal stem cells promotes fracture healing via modulation of the PTEN/PI3K/AKT axis.
        Exp Cell Res. 2022; 412113026
        • Huang Y
        • Xu Y
        • Feng S
        • He P
        • Sheng B
        • Ni J.
        miR-19b enhances osteogenic differentiation of mesenchymal stem cells and promotes fracture healing through the WWP1/Smurf2-mediated KLF5/β-catenin signaling pathway.
        Exp Mol Med. 2021; 53: 973-985
        • Li R
        • Li D
        • Wang H
        • Chen K
        • Wang S
        • Xu J
        • et al.
        Exosomes from adipose-derived stem cells regulate M1/M2 macrophage phenotypic polarization to promote bone healing via miR-451a/MIF.
        Stem Cell Research & Therapy. 2022; 13: 149
        • Yu H
        • Zhang J
        • Liu X
        • Li Y.
        microRNA-136-5p from bone marrow mesenchymal stem cell-derived exosomes facilitates fracture healing by targeting LRP4 to activate the Wnt/β-catenin pathway.
        Bone Joint Res. 2021; 10: 744-758
        • Tan SHS
        • Wong JRY
        • Sim SJY
        • Tjio CKE
        • Wong KL
        • Chew JRJ
        • et al.
        Mesenchymal stem cell exosomes in bone regenerative strategies—a systematic review of preclinical studies.
        Materials Today Bio. 2020; 7100067
        • Jung M
        • Ma Y
        • Iyer RP
        • DeLeon-Pennell KY
        • Yabluchanskiy A
        • Garrett MR
        • et al.
        IL-10 improves cardiac remodeling after myocardial infarction by stimulating M2 macrophage polarization and fibroblast activation.
        Basic Res Cardiol. 2017; 112: 33
        • Lv D
        • Zhou L
        • Zheng X
        • Hu Y.
        Sustained release of collagen VI potentiates sciatic nerve regeneration by modulating macrophage phenotype.
        Eur J Neurosci. 2017; 45: 1258-1267
        • Chu WC
        • Zhang S
        • Sng TJ
        • Ong YJ
        • Tan WL
        • Ang VY
        • et al.
        Distribution of pericellular matrix molecules in the temporomandibular joint and their chondroprotective effects against inflammation.
        Int J Oral Sci. 2017; 9: 43-52
        • Yu B
        • Shao H
        • Su C
        • Jiang Y
        • Chen X
        • Bai L
        • et al.
        Exosomes derived from MSCs ameliorate retinal laser injury partially by inhibition of MCP-1.
        Sci Rep. 2016; 6: 34562
        • Lo Sicco C
        • Reverberi D
        • Balbi C
        • Ulivi V
        • Principi E
        • Pascucci L
        • et al.
        Mesenchymal Stem Cell-Derived Extracellular Vesicles as Mediators of Anti-Inflammatory Effects: Endorsement of Macrophage Polarization.
        Stem Cells Transl Med. 2017; 6: 1018-1028
        • Cheung KM
        • Karppinen J
        • Chan D
        • Ho DW
        • Song YQ
        • Sham P
        • et al.
        Prevalence and pattern of lumbar magnetic resonance imaging changes in a population study of one thousand forty-three individuals.
        Spine (Phila Pa 1976). 2009; 34: 934-940
        • Sakai D
        • Mochida J
        • Iwashina T
        • Hiyama A
        • Omi H
        • Imai M
        • et al.
        Regenerative effects of transplanting mesenchymal stem cells embedded in atelocollagen to the degenerated intervertebral disc.
        Biomaterials. 2006; 27: 335-345
        • Miyamoto T
        • Muneta T
        • Tabuchi T
        • Matsumoto K
        • Saito H
        • Tsuji K
        • et al.
        Intradiscal transplantation of synovial mesenchymal stem cells prevents intervertebral disc degeneration through suppression of matrix metalloproteinase-related genes in nucleus pulposus cells in rabbits.
        Arthritis Res Ther. 2010; 12: R206
        • Sun Y
        • Leung VY
        • Cheung KM.
        Clinical trials of intervertebral disc regeneration: current status and future developments.
        International Orthopaedics. 2019; 43: 1003-1010
        • Kumar H
        • Ha DH
        • Lee EJ
        • Park JH
        • Shim JH
        • Ahn TK
        • et al.
        Safety and tolerability of intradiscal implantation of combined autologous adipose-derived mesenchymal stem cells and hyaluronic acid in patients with chronic discogenic low back pain: 1-year follow-up of a phase I study.
        Stem Cell Res Ther. 2017; 8: 262
        • Amirdelfan K
        • Bae H
        • McJunkin T
        • DePalma M
        • Kim K
        • Beckworth WJ
        • et al.
        Allogeneic mesenchymal precursor cells treatment for chronic low back pain associated with degenerative disc disease: a prospective randomized, placebo-controlled 36-month study of safety and efficacy.
        The Spine Journal. 2021; 21: 212-230
        • Liao Z
        • Ke W
        • Liu H
        • Tong B
        • Wang K
        • Feng X
        • et al.
        Vasorin-containing small extracellular vesicles retard intervertebral disc degeneration utilizing an injectable thermoresponsive delivery system.
        Journal of Nanobiotechnology. 2022; 20: 420
        • Zhu L
        • Shi Y
        • Liu L
        • Wang H
        • Shen P
        • Yang H.
        Mesenchymal stem cells-derived exosomes ameliorate nucleus pulposus cells apoptosis via delivering miR-142-3p: therapeutic potential for intervertebral disc degenerative diseases.
        Cell Cycle. 2020; 19: 1727-1739
        • Zhang J
        • Zhang J
        • Zhang Y
        • Liu W
        • Ni W
        • Huang X
        • et al.
        Mesenchymal stem cells-derived exosomes ameliorate intervertebral disc degeneration through inhibiting pyroptosis.
        Journal of Cellular and Molecular Medicine. 2020; 24: 11742-11754
        • Cheng X
        • Zhang G
        • Zhang L
        • Hu Y
        • Zhang K
        • Sun X
        • et al.
        Mesenchymal stem cells deliver exogenous miR-21 via exosomes to inhibit nucleus pulposus cell apoptosis and reduce intervertebral disc degeneration.
        Journal of Cellular and Molecular Medicine. 2018; 22: 261-276
        • Sun Z
        • Tang X
        • Li Q
        • Wang H
        • Sun H
        • Tian J.
        Mesenchymal stem cell extracellular vesicles-derived microRNA-194-5p delays the development of intervertebral disc degeneration by targeting TRAF6.
        Regenerative Therapy. 2022; 19: 88-96
        • Liao Z
        • Luo R
        • Li G
        • Song Y
        • Zhan S
        • Zhao K
        • et al.
        Exosomes from mesenchymal stem cells modulate endoplasmic reticulum stress to protect against nucleus pulposus cell death and ameliorate intervertebral disc degeneration in vivo.
        Theranostics. 2019; 9: 4084-4100
        • Xia C
        • Zeng Z
        • Fang B
        • Tao M
        • Gu C
        • Zheng L
        • et al.
        Mesenchymal stem cell-derived exosomes ameliorate intervertebral disc degeneration via anti-oxidant and anti-inflammatory effects.
        Free Radical Biology and Medicine. 2019; 143: 1-15
        • Yuan X
        • Li T
        • Shi L
        • Miao J
        • Guo Y
        • Chen Y.
        Human umbilical cord mesenchymal stem cells deliver exogenous miR-26a-5p via exosomes to inhibit nucleus pulposus cell pyroptosis through METTL14/NLRP3.
        Molecular Medicine. 2021; 27: 91
        • Guo Z
        • Su W
        • Zhou R
        • Zhang G
        • Yang S
        • Wu X
        • et al.
        Exosomal MATN3 of Urine-Derived Stem Cells Ameliorates Intervertebral Disc Degeneration by Antisenescence Effects and Promotes NPC Proliferation and ECM Synthesis by Activating TGF-β.
        Oxid Med Cell Longev. 2021; 20215542241
        • Nakamura Y
        • Miyaki S
        • Ishitobi H
        • Matsuyama S
        • Nakasa T
        • Kamei N
        • et al.
        Mesenchymal-stem-cell-derived exosomes accelerate skeletal muscle regeneration.
        FEBS Lett. 2015; 589: 1257-1265
        • Byun S-E
        • Sim C
        • Chung Y
        • Kim HK
        • Park S
        • Kim DK
        • et al.
        Skeletal Muscle Regeneration by the Exosomes of Adipose Tissue-Derived Mesenchymal Stem Cells.
        Current Issues in Molecular Biology. 2021; 43: 1473-1488
        • Cho K-A
        • Choi D-W
        • Kim Y-H
        • Kim J
        • Ryu K-H
        • Woo S-Y.
        Mesenchymal Stem Cell-Derived Exosomes Protect Muscle Loss by miR-145-5p Activity Targeting Activin A Receptors.
        Cells. 2021; 10: 2169
        • Kawata K
        • Koga H
        • Tsuji K
        • Miyatake K
        • Nakagawa Y
        • Yokota T
        • et al.
        Extracellular vesicles derived from mesenchymal stromal cells mediate endogenous cell growth and migration via the CXCL5 and CXCL6/CXCR2 axes and repair menisci.
        Stem Cell Res Ther. 2021; 12: 414
        • Wong KL
        • Zhang S
        • Tan SSH
        • Cheow YA
        • Lai RC
        • Lim SK
        • et al.
        Mesenchymal Stem Cell Exosomes Promote Growth Plate Repair and Reduce Limb-Length Discrepancy in Young Rats.
        JBJS. 2022; 104: 1098-1106
        • Guan P
        • Liu C
        • Xie D
        • Mao S
        • Ji Y
        • Lin Y
        • et al.
        Exosome-loaded extracellular matrix-mimic hydrogel with anti-inflammatory property Facilitates/promotes growth plate injury repair.
        Bioact Mater. 2022; 10: 145-158
        • Yu H
        • Cheng J
        • Shi W
        • Ren B
        • Zhao F
        • Shi Y
        • et al.
        Bone marrow mesenchymal stem cell-derived exosomes promote tendon regeneration by facilitating the proliferation and migration of endogenous tendon stem/progenitor cells.
        Acta Biomater. 2020; 106: 328-341
        • Chamberlain CS
        • Kink JA
        • Wildenauer LA
        • McCaughey M
        • Henry K
        • Spiker AM
        • et al.
        Exosome-educated macrophages and exosomes differentially improve ligament healing.
        Stem Cells. 2021; 39: 55-61
        • Liu Q
        • Zhu Y
        • Zhu W
        • Zhang G
        • Yang YP
        • Zhao C.
        The role of MicroRNAs in tendon injury, repair, and related tissue engineering.
        Biomaterials. 2021; 277121083
        • Toh WS
        • Lai RC
        • Zhang B
        • Lim SK.
        MSC exosome works through a protein-based mechanism of action.
        Biochemical Society Transactions. 2018; 46: 843-853
        • Chen TS
        • Lai RC
        • Lee MM
        • Choo AB
        • Lee CN
        • Lim SK.
        Mesenchymal stem cell secretes microparticles enriched in pre-microRNAs.
        Nucleic Acids Res. 2010; 38: 215-224
        • Chevillet JR
        • Kang Q
        • Ruf IK
        • Briggs HA
        • Vojtech LN
        • Hughes SM
        • et al.
        Quantitative and stoichiometric analysis of the microRNA content of exosomes.
        Proc Natl Acad Sci U S A. 2014; 111: 14888-14893
        • Albanese M
        • Chen YA
        • Hüls C
        • Gärtner K
        • Tagawa T
        • Mejias-Perez E
        • et al.
        MicroRNAs are minor constituents of extracellular vesicles that are rarely delivered to target cells.
        PLoS Genet. 2021; 17e1009951
        • Vallabhaneni KC
        • Penfornis P
        • Dhule S
        • Guillonneau F
        • Adams KV
        • Yuan Mo Y
        • et al.
        Extracellular vesicles from bone marrow mesenchymal stem/stromal cells transport tumor regulatory microRNA, proteins, and metabolites.
        Oncotarget. 2015; 6: 4953-4967
        • Haraszti RA
        • Didiot M-C
        • Sapp E
        • Leszyk J
        • Shaffer SA
        • Rockwell HE
        • et al.
        High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources.
        Journal of Extracellular Vesicles. 2016; 5: 32570
        • Skotland T
        • Sagini K
        • Sandvig K
        • Llorente A.
        An emerging focus on lipids in extracellular vesicles.
        Adv Drug Deliv Rev. 2020; 159: 308-321
        • Gimona M
        • Pachler K
        • Laner-Plamberger S
        • Schallmoser K
        • Rohde E
        Manufacturing of Human Extracellular Vesicle-Based Therapeutics for Clinical Use.
        Int J Mol Sci. 2017; 18: 1190
        • Tan TT
        • Lai RC
        • Padmanabhan J
        • Sim WK
        • Choo ABH
        • Lim SK.
        Assessment of Tumorigenic Potential in Mesenchymal-Stem/Stromal-Cell-Derived Small Extracellular Vesicles (MSC-sEV).
        Pharmaceuticals. 2021; 14: 345
        • Gimona M
        • Brizzi MF
        • Choo ABH
        • Dominici M
        • Davidson SM
        • Grillari J
        • 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
        • Chu CR
        • Szczodry M
        • Bruno S.
        Animal models for cartilage regeneration and repair.
        Tissue Eng Part B Rev. 2010; 16: 105-115
        • Simonsen JB.
        Pitfalls associated with lipophilic fluorophore staining of extracellular vesicles for uptake studies.
        Journal of Extracellular Vesicles. 2019; 81582237