Open Access

Potential and challenges of utilizing exosomes in osteoarthritis therapy (Review)

  • Authors:
    • Xuesong Chen
    • Bin Tian
    • Yiqun Wang
    • Jiang Zheng
    • Xin Kang
  • View Affiliations

  • Published online on: January 8, 2025     https://doi.org/10.3892/ijmm.2025.5484
  • Article Number: 43
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Exosomes are integral to the pathophysiology of osteoarthritis (OA) due to their roles in mediating intercellular communication and regulating inflammatory processes. Exosomes are integral to the transport of bioactive molecules, such as proteins, lipids and nucleic acids, which can influence chondrocyte behavior and joint homeostasis. Given their properties of regeneration and ability to target damaged tissues, exosomes represent a promising therapeutic avenue for OA treatment. Exosomes have potential in promoting cartilage repair, reducing inflammation and improving overall joint function. However, several challenges remain, including the need for standardized isolation and characterization methods, variability in exosomal content, and regulatory hurdles. The present review aims to provide a comprehensive overview of the current understanding of exosome mechanisms in OA and their therapeutic potential, while also addressing the ongoing challenges faced in translating these findings into clinical practice. By consolidating existing research, the present review aims to pave the way for future studies aimed at optimizing exosome‑based therapies for effective OA management.

Introduction

Osteoarthritis (OA) is the most common degenerative joint disease, characterized by the progressive breakdown of articular cartilage, subchondral bone changes and synovial inflammation (1). OA predominantly affects weight-bearing joints such as the knees, hips and spine, leading to pain, stiffness and functional impairment (2). According to recent epidemiological studies, the prevalence of OA is increasing globally, especially among aging populations (3,4). The World Health Organization has reported that approximately one third of individuals over the age of 65 years will experience some form of OA, contributing to healthcare burdens and diminished quality of life (5).

The pathophysiology of OA is multifactorial, involving mechanical, biochemical and genetic factors. Mechanical stress can lead to cartilage degradation through increased cytokine and MMP production, resulting in an imbalance between anabolic and catabolic processes within the joint (6). Additionally, synovial inflammation serves a pivotal role in OA progression, as it not only contributes to the degradation of cartilage but also stimulates pain perception and dysfunction in the affected joints (7). The inflammatory milieu is often exacerbated by the presence of pro-inflammatory mediators such as interleukins and TNF-α, promoting further degradation and pain generation (7). Furthermore, chondrocyte senescence and apoptosis contribute to the decline of cartilage repair mechanisms, perpetuating the cycle of joint degeneration (8). OA treatment currently relies on a multimodal approach, including pharmacological interventions such as nonsteroidal anti-inflammatory drugs, opioids and corticosteroids, alongside physical therapy, lifestyle modifications and surgical options (9). However, challenges remain, including the limited efficacy of medications, potential side effects and the progressive nature of the disease (5). These factors often lead to inadequate pain relief and functional improvement (10), prompting the need for innovative therapies beyond traditional methods.

Extracellular vesicles (EVs), particularly exosomes, have garnered attention in recent years as vital mediators in cell communication and intercellular signaling (11). EVs are membrane-bound vesicles released by various cell types, and they can be classified based on their biogenesis: Exosomes (30-150 nm) are derived from the endosomal system, while microvesicles and apoptotic bodies originate from cell membrane shedding and apoptosis, respectively (12). Exosomes contain a variety of biomolecules, including proteins, lipids and RNAs, which reflect the physiological state of their originating cells (13). This cargo allows exosomes to participate in diverse biological processes, including immune responses, tissue repair and inflammation, making them important players in pathology (13).

The significance of EVs in the context of OA is emerging, as they possess the potential to modulate inflammatory responses, influence chondrocyte behavior and facilitate communication between cells within the joint (14). There is growing evidence indicating that EVs released from degenerative joint tissues may contribute to the pathogenesis of OA by carrying pro-inflammatory signals (15-17). Conversely, therapeutic application of EVs derived from healthy cells has shown promise in attenuating OA progression by promoting chondrocyte survival and function (18).

The present review aims to elucidate the role of exosomes in the mechanisms underlying OA, highlighting their potential as novel biomarkers and therapeutic targets. By consolidating current research findings, the present review assesses the dual nature of exosomes in OA: Both as contributors to disease pathology and as vehicles for potential regenerative strategies. Understanding these dynamics may pave the way for innovative therapeutic approaches, improving clinical outcomes for patients suffering from OA and enhancing the comprehension of joint biology.

Role of exosomes in OA

Exosomes are important carriers of information transfer between cells (19). Exosomes enter target cells through membrane fusion, receptor binding and endocytosis, facilitating intercellular communication, immune modulation, waste removal, and transfer of proteins, lipids and RNA to influence cellular functions (19). Exosomes are integral players in the pathophysiology of OA, with their biogenesis, composition and interactive dynamics influencing disease progression (20), and serve as vehicles for a diverse range of biomolecules, providing critical insights into the mechanisms underlying OA and presenting novel opportunities for therapeutic intervention (21). Existing research focuses on delineating the specific roles of exosomal components in cellular interactions and signaling pathways, which is essential for developing innovative strategies to combat this debilitating condition (22).

Exosome biogenesis and composition

Exosomes are small EVs that range in size from 30 to 150 nm. They are formed through the multivesicular bodies (MVBs) pathway within the endosomes of various cell types (23). The MVBs can either be targeted to lysosomes for degradation or fuse with the plasma membrane to release exosomes into the extracellular space (23). Exosomes can be classified based on their cellular origin, including exosomes derived from chondrocytes, synovial cells and bone marrow stem cells, and others associated with the joint microenvironment (24). Chondrocyte-derived exosomes are particularly important in the context of OA, as they reflect cartilage health and contribute to disease progression through the release of inflammatory cytokines and matrix-degrading enzymes (25,26). In addition, subchondral bone-derived exosomes contain factors that influence osteoclast and osteoblast activity, potentially exacerbating cartilage degeneration (27). Synovial cell-derived exosomes are implicated in modulating inflammation within the synovial fluid, thereby impacting clinical outcomes in patients with OA (14). Furthermore, exosomes from bone marrow stem cells have shown promise in promoting tissue regeneration and reducing inflammation, offering a potential therapeutic avenue in OA management (28). Understanding the origins and functions of exosomes around OA is crucial for their application as biomarkers and therapeutic agents in OA.

The biochemical composition of exosomes encompasses a diverse array of biomolecules, including proteins, lipids and nucleotides (19). Proteins found in exosomes can include receptors, cytokines, growth factors and enzymes that serve critical roles in cell signaling and intercellular communication (19). Furthermore, exosomes are enriched in distinct lipid compositions, including sphingolipids, phospholipids and cholesterol, which are essential for membrane integrity and fluidity (29). Nucleic acids present in exosomes, particularly microRNAs (miRNAs/miRs) and mRNAs, are critical for gene regulation (30). The unique composition of each exosome is influenced by its cell of origin and the microenvironment, which makes exosomes valuable indicators of pathological changes in OA (15).

Function of exosomal components in OA progression
miRNA payloads and gene regulation

The influence of exosomal miRNAs in OA has garnered attention due to their potential roles in mediating intercellular communication and gene regulation. Extensive research has been conducted to understand how exosomal miRNAs contribute to the progression of OA and their therapeutic implications (Table I). The dysregulation of these miRNAs may lead to an imbalance in matrix synthesis and degradation, promoting the pathological processes of OA (21). Additionally, exosomal miRNAs can influence neighboring cell types, such as synovial cells and macrophages, contributing to the chronic inflammatory environment characteristic of OA (21). By modulating gene expression in target cells, exosomal miRNAs coordinate a wide range of cellular responses, thereby serving an integral role in the progression of OA (Fig. 1) (31).

Table I

Regulatory mechanism of OA regulated by miRNAs loaded with exosomes.

Table I

Regulatory mechanism of OA regulated by miRNAs loaded with exosomes.

First author/s, yearmiRNAsExosome-derived cellsBiological effect(Refs.)
Kong et al, 2023miR-19b-3pSynovial cellsmiR-19b-3p enhances cartilage ferroptosis and damage by sponging SLC7A11 in OA.(32)
Mao et al, 2018miR-92a-3pMSCsmiR-92a-3p promotes chondrocyte proliferation, while suppressing cartilage degradation in OA models.(33)
Qiu et al, 2021miR-129-5pHuman synovial MSCsChondrocyte apoptosis and inflammation are reduced by miR-129-5p.(34)
Xia et al, 2021miR-125a-5pMSCsBy promoting ECM secretion, miR-125a-5p alleviates chondrocyte degeneration.(35)
Tao et al, 2021miR-361-5pHuman bone MSCsChondrocyte damage is mitigated by miR-361-5p.(36)
Li et al, 2021miR-100-5pHuman umbilical cord MSCsExosomal miR-100-5p alleviates cyclic strain-induced chondrocyte damage in OA.(37)
Ye et al, 2022miR-3960MSCsmiR-3960 relieves chondrocyte injury in OA.(38)
Wang et al, 2021miR-155-5pSynovial MSCsmiR-155-5p promotes proliferation, inhibits apoptosis and regulates ECM secretion.(40)
Qian et al, 2024miR-26b-5pM2 macrophagesM2 macrophage-derived exosomal miR-26b-5p can alleviate OA by targeting TLR3 and COL10A1, and regulates macrophage polarization and chondrocyte hypertrophy.(41)
Li et al, 2023miR-376c-3pAdipose MSCsmiR-376c-3p inhibits the WNT-β-catenin pathway by targeting WNT3 or WNT9a, thereby suppressing chondrocyte apoptosis and alleviating OA-induced chondrocyte degradation and synovial fibrosis.(79)
Tao et al, 2017miR-140-5pHuman synovial MSCsEarly-stage OA is delayed by miR-140-5p, which promotes chondrocyte proliferation and migration.(146)
Sun et al, 2019miR-320cMSCsBy downregulating MMP-13 and upregulating SOX9 expression, miR-320c promotes chondrocyte proliferation.(147)
Lu et al, 2021miR-26a-5pSynovial MSCsApoptosis and inflammation are inhibited by miR-26a-5p, which ameliorates cartilage injury in OA.(148)
Li et al, 2022miR-338-3pMSCsmiR-338-3p promotes cell proliferation and inhibits apoptosis.(149)
Zhou et al, 2022miR-1208BMSCsThrough a decrease in pro-inflammatory factors, miR-1208 suppresses cartilage ECM degradation.(150)
Zhang et al, 2023miR-3473bBone marrow MSCsBy targeting PTEN, miR-3473b promotes the migration of OA, improves the anabolism of chondrocytes and inhibits the apoptosis of chondrocytes.(151)
Lai et al, 2023miR-214-3pSynovial fibroblastsExosomal miR-214-3p suppresses the formation of osteophytes, prevents degeneration of cartilage, and exerts anti-inflammatory and anti-apoptotic effects in articular cartilage tissue.(152)
Ji et al, 2023miR-182-5pSynovial fluidSynovial fluid exosome-derived miR-182-5p alleviates OA by downregulating TNFAIP8 and promoting autophagy via LC3 signaling.(153)
Wu et al, 2024miR-182Synovial fluidmiR-182 may directly regulate OA progression by modulating FOXO3 production, inflammation and apoptosis.(154)
Qiu et al, 2024miR-485-3pSynovial MSCsSynovial mesenchymal stem cell-derived exosomal miR-485-3p relieves cartilage damage in OA by targeting the NRP1-mediated PI3K/Akt pathway.(155)

[i] BMSC, bone marrow mesenchymal stem cell; COL10A1, collagen type X α1 chain; ECM, extracellular matrix; MSCs, mesenchymal stem cells; miRNA/miR, microRNA; NRP1, neuropilin 1; OA, osteoarthritis; ROS, reactive oxygen species; SLC7A11, solute carrier family 7 member 11; TLR3, toll like receptor 3; TNFAIP8, TNF α induced protein 8.

Several studies have highlighted exosomal miRNAs that facilitate the progression of OA by targeting key regulatory pathways (32-34). Kong et al (32) illustrated that exosomes secreted from osteoarthritic fibroblast-like synoviocytes contain miR-19b-3p, which promotes cartilage ferroptosis by targeting solute carrier family 7 member 11, thereby exacerbating cartilage damage. This finding underscores the contribution of specific miRNAs derived from pathologically altered tissue to the overall OA pathology. In another pivotal study, Mao et al (33) demonstrated that miR-92a-3p derived from exosomes of human mesenchymal stem cells (MSCs) enhanced chondrogenesis while simultaneously suppressing cartilage degradation through targeted modulation of WNT5A. This research indicates that certain exosomal miRNAs can serve a dual role, depending on the cellular context, complicating their impact on cartilage health. Furthermore, Xia et al (35) reported that miR-125a-5p-rich exosomes from MSCs suppress chondrocyte degeneration in traumatic OA by targeting E2F2, presenting evidence that even dysregulated miRNAs can have protective roles under certain conditions. Overall, these studies emphasize the critical role that exosomal miRNAs serve in propagating OA through the modulation of signaling pathways involved in cartilage homeostasis. Understanding these mechanisms is crucial for identifying potential therapeutic targets as well as biomarkers for monitoring disease progression.

A growing body of research has demonstrated the ability of certain exosomal miRNAs to inhibit the progression of OA, providing avenues for potential therapeutic interventions (36-38). Tao et al (36) revealed that exosomal miR-361-5p derived from human bone MSCs alleviates OA by downregulating DEAD-box helicase 20 and inactivating the NF-κB signaling pathway, consequently reducing inflammation and promoting chondrocyte viability. The study highlights the impact of MSC-derived exosomes in mitigating OA pathology through specific miRNA-mediated mechanisms. Further exploring protective roles, Li et al (37) observed that exosomal miR-100-5p derived from human umbilical cord MSCs alleviated cyclic strain-induced chondrocyte damage in OA by inhibiting reactive oxygen species production and apoptosis through direct targeting of NADPH oxidase 4 (NOX4). This finding not only provides insights into how exosomal miRNAs can counteract oxidative stress but also establishes potential therapeutic uses for MSC-derived exosomal products in OA treatment. Furthermore, Ye et al (38) demonstrated that exosomal miR-3960 can target the syndecan 1/Wnt/β-catenin axis, leading to the alleviation of chondrocyte injury in OA, further supporting the protective potential of exosomal miRNAs against degeneration in joint tissues. This dichotomy of miRNAs, both promoting and inhibiting OA progression, points towards a complex interplay where the recruitment, secretion and action of specific exosomes can be leveraged for therapeutic purposes. The choice of exosomal miRNA delivery systems may thus prove pivotal in therapeutic strategies aimed at OA management.

The diagnostic and therapeutic potential of exosomal miRNAs extends beyond their role in OA progression. As highlighted by a study, alterations in the levels of specific exosomal miRNAs, including miR-140-5p, miR-92a-3p, miR-100-5p and miR-155-5p, may serve as biomarkers for OA severity and response to treatment (39). These miRNAs can elucidate the molecular landscape of joint degeneration, providing insights into disease mechanisms and guiding therapeutic decisions (31). The therapeutic application of exosomal miRNAs offers promising strategies to combat OA. Utilizing exosomes enriched with protective miRNAs can be a novel approach to modulate inflammatory responses and enhance chondrocyte function. For instance, Wang et al (40) demonstrated that exosomal miR-155-5p derived from synovial MSCs enhanced proliferation, migration and extracellular matrix (ECM) secretion in chondrocytes, suggesting their potential for tissue engineering and regenerative medicine applications in OA. Furthermore, Qian et al (41) illustrated that M2 macrophage-derived exosomal miR-26b-5p modulates macrophage polarization to alleviate OA, underscoring the therapeutic potential of targeting inflammation in OA treatment using miRNA-based strategies. Overall, the ongoing exploration of exosomal miRNAs holds the potential to revolutionize the understanding and treatment of OA. The ability of miRNAs to act as diagnostic markers and therapeutic agents signals a pivotal shift in how OA may be managed in clinical settings, emphasizing the importance of further research to harness their full therapeutic potential.

Proteins and their signaling pathways

Exosomes have emerged as key mediators of intercellular communication in OA (14). These vesicles carry a cargo rich in proteins that can influence the pathophysiological processes of OA (22). The role of exosomal proteins in OA is multifaceted and involves various signaling pathways essential for maintaining cartilage integrity, regulating inflammation and promoting chondrocyte survival (1,42). Through their ability to mediate complex cellular communication and modulate critical signaling pathways such as the TGF-β, Wnt/β-catenin, PI3K/Akt, MAPK/ERK and NF-κB signaling pathways, exosomes offer a novel avenue for therapeutic intervention in OA (43-47) (Fig. 2). Focusing on specific exosomal cargoes and their respective signaling pathways may reveal novel therapeutic targets for OA, paving the way for innovative treatment strategies that address the underlying mechanisms of the disease.

TGF-β pathway

TGF-β is a well-characterized protein present in exosomes, with implications in the regulation of chondrocyte homeostasis and cartilage repair mechanisms (42). Studies have demonstrated that exosomal TGF-β promotes chondrocyte proliferation and matrix synthesis while inhibiting apoptosis (48,49). The activation of the TGF-β signaling pathway leads to the phosphorylation of SMAD proteins, which subsequently regulate the expression of ECM components, such as collagen and aggrecan (43). Furthermore, TGF-β signaling has been implicated in the modulation of inflammation within the OA joint, helping to counteract pro-inflammatory cytokine effects, including those mediated by TNF-α and IL-1β (50).

Wnt/β-catenin signaling pathway

Exosomal proteins can also influence the Wnt/β-catenin signaling pathway, which serves a role in maintaining cartilage integrity and regulating osteophyte formation (44). For example, Wnt-5a has been shown to be present in MSC-derived exosomes and to activate β-catenin signaling in chondrocytes, promoting their differentiation and proliferation (44). This pathway may also enhance the synthesis of cartilage-specific markers while suppressing the expression of MMPs that contribute to cartilage degradation (1). In OA, the dysregulation of Wnt/β-catenin signaling is evident, and thus, exosomal proteins modulating this pathway may offer therapeutic benefits.

PI3K/Akt pathway

The PI3K/Akt signaling pathway is another crucial pathway influenced by exosomal proteins in OA (51). Exosomal delivery of growth factors such as VEGF can activate the PI3K/Akt pathway in target cells, leading to enhanced cell survival and proliferation (45). In the context of OA, activation of this pathway in chondrocytes can promote not only cell survival but also the production of anti-apoptotic factors such as Bcl-2 (45). Additionally, the PI3K/Akt pathway has been shown to inhibit the expression of inflammatory mediators, thus providing a protective effect against the inflammatory milieu associated with OA (52).

MAPK/ERK pathway

The MAPK/ERK pathway is another signaling pathway activated by exosomal proteins (53). Proteins such as fibroblast growth factor 2 carried by exosomes can activate this pathway in chondrocytes, leading to cellular responses that support cartilage anabolism and integrity (54). Activation of MAPK/ERK signaling leads to increased expression of chondrocyte-specific markers and ECM components, while downregulating inflammatory responses (46). Dysregulation of the MAPK pathway has been associated with OA progression (55), thus showcasing the therapeutic potential of exosomal proteins that can modulate this pathway effectively.

NF-κB signaling pathway

The NF-κB signaling pathway is central to the inflammatory response observed in OA (56). Exosomal proteins can modulate the activity of NF-κB, thereby influencing the expression of pro-inflammatory cytokines. For instance, exosomal delivery of cytokines such as IL-10 can inhibit the activation of NF-κB, leading to suppressed expression of inflammatory mediators such as IL-1β and TNF-α (47). This inhibitory effect not only protects chondrocytes from inflammation-induced apoptosis but also promotes a favorable microenvironment for cartilage regeneration (47).

miRNA and protein interactions

In addition to direct signaling effects, exosomal proteins also interact with miRNAs that can post-transcriptionally regulate gene expression (57). For example, the exosomal protein cargo may deliver specific miRNAs, such as miR-140, which have been shown to serve a regulatory role in maintaining cartilage integrity and modulating inflammatory responses in OA (58). Similarly, miR-100-5p regulates NOX4 levels, reducing oxidative stress and apoptosis in response to cyclic strain (37). These findings illustrate how exosomal miRNAs can modulate protein activity, influencing OA pathology and presenting potential therapeutic targets for treatment strategies.

Lipids and impact on inflammation

The lipid composition of exosomes also serves a pivotal role in the inflammatory response associated with OA (59). In OA, aberrant lipid profiles in exosomes can influence inflammation by activating pro-inflammatory pathways (59). For instance, the presence of specific sphingolipids can modulate inflammatory cytokine production in synovial cells, contributing to synovitis (60). These lipids can also affect the stability and functionality of exosomes, subsequently influencing their interaction with target cells (60). By altering the lipid composition in exosomes, researchers may be able to design therapeutic strategies to attenuate inflammation and promote cartilage health (Fig. 3). Further research into the lipid profiles of exosomes in OA could reveal novel insights into their role in inflammation and promote the development of lipid-based therapies targeting inflammatory mechanisms to improve joint health.

Role of lipids in exosome biogenesis and function

Exosomes are lipid bilayer vesicles that originate from MVBs and facilitate the transfer of bioactive molecules (molecules that are able to interact with molecules in living organisms to produce specific biological effects) between cells (61). The lipid composition of exosomes, which includes phospholipids, cholesterol and sphingolipids, is pivotal for their biogenesis, stability and cargo selection (61). For instance, certain lipids, including phosphoinositides and ether lipids, contribute to the fluidity of the exosomal membrane and influence the loading of proteins and RNA molecules (61). Mori et al (59) highlighted that exosomes can serve as nanocarriers for the delivery of therapeutic agents in inflammatory diseases, indicating that manipulating lipid composition could be an effective novel strategy for drug delivery.

Lipid dysregulation in OA

In OA, the lipid metabolism is altered, which affects the properties and functionality of synovial fluid and cartilage (62). Sphingosine-1-phosphate (S1P) is a sphingolipid that has been implicated in OA pathogenesis (63). El Jamal et al (60) demonstrated that S1P signaling contributes to cartilage degradation and promotes inflammation in joint tissues, revealing that lipid alterations may directly impact disease mechanisms. Furthermore, alterations in the lipid composition of exosomes derived from MSCs may modulate the progression of OA, as shown by Toh et al (64), suggesting that therapeutic MSC-derived exosomes could improve cartilage repair by restoring normal lipid profiles.

Immunomodulatory role of exosomal lipids

Exosomal lipids serve crucial roles in immunomodulation, particularly in the context of OA, where inflammation is a central feature (22). Lipid components, including phospholipids and sphingolipids, are known to influence immune cell activation and cytokine release (22). Cosenza et al (65) emphasized the importance of MSCs-derived exosomes in modulating the inflammatory microenvironment. The bioactive lipids within these exosomes can reduce pro-inflammatory cytokines and promote an anti-inflammatory milieu (65). This effect suggests that lipid-carrying exosomes might be harnessed to mitigate OA-related inflammation and pain.

Therapeutic potential of exosomal lipids in OA management

The therapeutic potential of exosomes, particularly those derived from MSCs, is underscored by their ability to deliver lipids and other bioactive molecules to affected joint tissues (66). Yang et al (67) provided evidence that exosomal treatments can enhance cartilage regeneration and joint function in OA models. Exosomes derived from MSCs can promote chondrogenesis and inhibit cartilage degradation by delivering specific lipids (59). Furthermore, Fan et al (15) indicated the possibility of utilizing exosomes as novel drug delivery systems, where lipid manipulation could optimize their therapeutic effects.

Interaction with cartilage cells
Chondrocytes

Exosomes derived from various sources can exert effects on chondrocyte proliferation and apoptosis (68,69) (Figs. 1 and 2). For example, exosomes from stem cells have been shown to enhance chondrocyte proliferation and survival, providing a potential avenue for therapeutic application in OA (28). These exosomes contain factors, such as TGF-β and MMPs, that can stimulate anabolism in chondrocytes and promote the synthesis of ECM components (42,70). Conversely, exosomes from degenerative cartilage can contain catabolic signals that trigger chondrocyte apoptosis and inhibit their repair capabilities (25,71). This dichotomy underscores the importance of identifying the origin and content of circulating exosomes in the context of OA, as their influence can determine the fate of chondrocytes and the overall joint health.

Chondrocytes are intrinsically sensitive to the inflammatory signals present in the OA milieu (8). Exosomes serve a crucial role in modulating this response by delivering pro-inflammatory or anti-inflammatory signals. For example, exosomes from inflamed joints may carry TNF-α or IL-1β, perpetuating the inflammatory feedback loop and resulting in further cartilage degradation (72). On the other hand, certain exosomal cargoes may have the potential to mitigate inflammation. MSCs-derived exosomes can reduce the infiltration of inflammatory cells and inhibit the production of specific inflammatory mediators by providing anti-inflammatory factors such as TGF-β and prostaglandin E2 (73). Furthermore, exosomes harboring anti-inflammatory miRNAs can promote resistance to apoptosis and enhance chondrocyte function (74). This balance between pro-inflammatory and anti-inflammatory exosome signaling is pivotal for maintaining cartilage homeostasis in OA (74).

Synovial cells

The accumulation of exosomes in the synovial fluid during OA contributes to synovial inflammation (synovitis), a hallmark of the disease (75). Synovial cells, including fibroblasts and macrophages, respond to exosomal signals, leading to increased production of inflammatory mediators (32). This culminates in a self-perpetuating cycle of inflammation that contributes to joint pain and dysfunction. Exosomes from osteoarthritic synovial cells can influence local immunity and inflammation, promoting the recruitment of immune cells to the joint (32). This heightened inflammatory response can exacerbate tissue damage and further propagate the cycle of joint deterioration (76).

Exosomes also hold therapeutic potential for immune modulation within the context of OA (77). For instance, exosomes derived from MSCs can induce an anti-inflammatory response in synovial cells, downregulating the production of pro-inflammatory cytokines. This property makes them an attractive candidate for cell-based therapies aimed at reducing synovitis and promoting cartilage regeneration (77). Understanding the interaction between exosomes and synovial cells can open up novel avenues for targeting OA inflammation. Therapeutic approaches leveraging exosomes as delivery vehicles for anti-inflammatory agents or regenerative factors may ultimately enhance joint healing and function.

Signaling cascades or biomarkers associated with exosomes present in OA

Exosomes have emerged as crucial mediators in the pathophysiology of OA, influencing key signaling pathways and serving as potential biomarkers (78). In OA, exosomes derived from chondrocytes, synovial cells and cartilage have been shown to contain various biomolecules, including proteins, lipids, mRNA and miRNA, which can modulate inflammatory responses and cartilage homeostasis (78).

One signaling pathway associated with exosome activity in OA is the Wnt/β-catenin pathway (79). Research has indicated that exosomes from OA chondrocytes can transfer Wnt ligands, activating this pathway, which contributes to chondrocyte hypertrophy and cartilage degradation (79). Additionally, exosomal miRNAs, particularly miR-21 and miR-140, have been implicated in the regulation of this pathway, further promoting catabolic processes in cartilage (80,81).

Exosomes serve a vital role in mediating the inflammatory response in OA. They facilitate communication between synovial cells and chondrocytes, promoting the release of pro-inflammatory cytokines such as TNF-α and IL-1β (82). Exosomal miRNAs, including miR-146a, can modulate the NF-κB pathway, enhancing inflammatory signaling and exacerbating joint damage (83).

Previous studies have also identified specific exosomal biomarkers that could help in OA diagnosis and prognosis. For instance, increased levels of exosomal proteins such as TGF-β and cyclooxygenase-2 have been reported in patients with OA, and are associated with disease severity (42,72). Furthermore, circulating exosomal miRNAs are being investigated as non-invasive biomarkers for OA progression and response to treatment (84).

In conclusion, exosomes are pivotal in the signaling cascades and molecular mechanisms underlying OA. The interactions between exosomal cargo and various signaling pathways reveal their dual role as mediators of cartilage degeneration and potential biomarkers for disease monitoring (85). Future research targeting exosomal contents could pave the way for novel therapeutic strategies aimed at modulating exosomal signaling and mitigating OA progression. This highlights the necessity for continued exploration of the role of exosomes in OA and their therapeutic prospects.

Exosome applications in OA treatment

EVs are important mediators of intercellular communication and show promising potential in the treatment of OA (86). Compared with conventional therapies, exosome-based treatments offer several advantages, including enhanced cartilage regeneration, reduced inflammatory responses and modulated immune reactions (87). They can facilitate the delivery of bioactive molecules such as proteins, RNAs and lipids directly to target cells, promoting tissue repair (88). Furthermore, compared with traditional drugs and biological agents, exosomes have lower immunogenicity and are associated with fewer side effects, making them a potentially safer alternative to pharmacological interventions in OA management (87). Thus, utilizing exosomes can represent a novel and effective strategy for OA therapy.

EVs have shown promising potential in preclinical studies for the treatment of OA, demonstrating their ability to mediate cellular communication and regeneration within joint tissues (89-94). These studies have indicated that the administration of vesicles can attenuate cartilage degradation and promote chondrocyte proliferation (89-94) (Table II). Additionally, a limited number of clinical studies have begun to reveal the therapeutic potential of EVs in managing OA (95-99), suggesting a novel avenue for intervention in this degenerative joint disease.

Table II

Preclinical studies on the application of exosomes in the treatment of OA.

Table II

Preclinical studies on the application of exosomes in the treatment of OA.

First author/s, yearExosome-derived cellsProcessing strategyOA modelEfficacy(Refs.)
Wang et al, 2017ESC-MSCsExosomes were extracted from the supernatant of ESC-MSC culture.Mouse knee OA modelExosomes of ESC-MSCs serve a beneficial therapeutic role in OA by balancing the synthesis and degradation of chondrocyte extracellular matrix.(90)
Zhang et al, 2019MSCsExosomes were extracted from the supernatant of MSC culture.Rat model of temporomandibular joint OAMSCs exosomes promote OA repair andregeneration, thereby restoring overall joint homeostasis.(91)
He et al, 2020BMSCsExosomes were extracted from the supernatant of MSC culture.Rat model of OABone marrow mesenchymal stem cell-derived exosomes can effectively promote cartilage repair and extracellular matrix synthesis.(92)
Xu and Xu, 2021BMSCsExosomes were extracted from the supernatant of BMSC culture.Rat OA modelBMSCs-derived exosomes ameliorate OA by delivering miR-326 to chondrocytes to inhibit pyroptosis of chondrocytes and cartilage.(93)
Jin et al, 2021BMSCsInjection of BMSCs or their exosomes.Rat OA modelExosomes from BMSCs exert beneficial therapeutic effects on OA by reducing the senescence and apoptosis of chondrocytes.(94)
Jiang et al, 2021MSCsExosomes extracted from the bone marrow MSCs of the mouse OA model.Mouse knee OA modelMSCs-derived exosomes modulate chondrocyte glutamine metabolism to alleviate OA progression.(97)
Yin et al, 2023SCAT-derived stem cellsHydrogel microparticles containing ADSCs-derived exosomes enriched with miR-99a-3p.Mouse knee OA modelEncapsulation of exosomes in hydrogel microparticles provides an injectable continuous local drug delivery system.(100)
Wang et al, 2022ADSCsExosomes were extracted from the supernatant of ADSC culture.Rat OA modelADSCs-derived exosomes modulate chondrocyte metabolism, thus alleviating the progression of OA.(101)
Zhao et al, 2023ADSCsHypoxia-treated ADSCs-derived exosomes.MiceHypoxia is an effective method to improve the therapeutic effect of ADSC-exosomes in ameliorating spinal pain and lumbar facet joint OA progression.(102)
Fu et al, 2023DPSCsExosomes were extracted from the supernatant of DPSC culture.Mouse knee OA modelDPSCs-derived exosomes effectively improve abnormal subchondral bone remodeling, inhibit the occurrence of bone sclerosis and osteophytes, and alleviate cartilage degradation and synovial inflammation in vivo.(103)
Meng et al, 2023ADSCsADSCs were isolated and cultured from rats to extract exosomesRat knee OA modelADSCs exosomes promote chondrocyte proliferation through miR-429. miR-429 improves OA cartilage injury by targeting FEZ2 and promoting autophagy.(104)
Yang et al, 2024hUC-MSCshUC-MSCs were isolated from the umbilical cord and exosomes were extracted.Rat knee OA modelhUC-MSCs-exosomes promote cartilage regeneration in OA rats.(105)
Huang et al, 2023Human placenta-derived exosomesPlacenta-derived exosomes were generated from full-term human placenta tissues.Rat knee OA modelpExo has multiple potential therapeutic effects, including symptom control and disease-modifying characteristics.(106)
Zhao et al, 2023Subcutaneous fat MSCsAs a delivery vector of miR-199a-3p.RatsExosomes enriched with miR-199a-3p improve the therapeutic outcomes of cartilage injury and OA by targeting delivery of the molecule and regulating the mTOR-autophagy pathway.(107)
Yang et al, 2024MSCsExosomes were utilized as therapeutic agents for OA by being encapsulated within magnetic polysaccharide hydrogel microcarriers.Rat knee OA modelReleased exosomes from the microcarriers have a synergistic effect in alleviating OA symptoms and promoting cartilage repair.(108)
Ma et al, 2024MSCsEngineered exosomes with ATF5-modified mRNA loaded in injectable thermogels.Rat knee OA modelGenetically engineered exosome-treated cartilage has an intact surface, low OARSI scores, fewer osteophytes, mild subchondral osteosclerosis and cystic degeneration.(109)
Zhang et al, 2022PRP-exosomesPRP-exosomes were coated with thermal gel.Mouse knee OA modelExosome-gel increases the local retention of exosomes, inhibits the apoptosis and hypertrophy of chondrocytes, enhances their proliferation, and potentially serves a role in stem cell recruitment to delay the development of STOA.(156)
Lou et al, 2023MSCsFucoidan-pretreated MSCs-derived exosomes.Rat knee OA modelExosomes loaded with miR-146b-5p can effectively inhibit the inflammatory response and extracellular matrix degradation, and promote chondrocyte autophagy to protect OA chondrocytes.(157)
Meng et al, 2023ADSCsExosomes derived from TE-pretreated ADSCs.Rat knee OA modelTE-exosomes help maintain the chondrocyte phenotype in vitro and promote cartilage repair in vivo. These therapeutic effects may be related to the expression changes of miR-451-5p.(158)
Wang et al, 2023hUC-MSCshUC-MSCs-exosomes were isolated from hUC-MSC culture supernatant.Cartilage inflammation chondrocyte modelhUC-MSC-exosomes have an anti-inflammatory effect in a human articular chondrocyte inflammation model.(159)
Chen et al, 2024CSPCsExosomes were prepared from CSPCs.Rat knee OA modelThe application of exosomes promotes cartilage regeneration.(160)

[i] ADSC, adipose mesenchymal stem cell; ATF5, activating transcription factor 5; BMSCs, bone marrow mesenchymal stem cells; CSPCs, cartilage stem/progenitor cells; DPSCs, dental pulp stem cells; ESC-MSCs, human embryonic stem cell-induced mesenchymal stem cells; FEZ2, fasciculation and elongation protein ζ2; hUC-MSCs, human umbilical cord-derived mesenchymal stem cells; miR, microRNA; MSCs, mesenchymal stem cells; OA, osteoarthritis; OARSI, Osteoarthritis Research Society International; pExo, placenta-derived exosome; PRP-exosomes, platelet-rich plasma-derived exosomes; SCAT, subcutaneous adipose tissue; STOA, subtalar osteoarthritis; TE, tropoelastin.

Preclinical studies
Exosomes derived from embryonic and adult stem cells

Exosomes derived from embryonic MSCs (eMSCs) have been shown to alleviate OA by balancing the synthesis and degradation of the cartilage ECM (90). This study demonstrated that eMSC-derived exosomes promote chondrocyte proliferation and enhanced matrix synthesis, while inhibiting catabolic pathways. Similarly, He et al (92) reported that bone marrow MSC-derived exosomes possess protective effects against cartilage damage in a rat OA model. The authors found that these exosomes reduce knee pain and enhanced joint function, suggesting a therapeutic potential for OA management. Furthermore, Xu et al (93) provided insights into the anti-inflammatory potential of bone marrow MSC-derived exosomes by demonstrating their ability to inhibit pyroptosis in chondrocytes through the specific delivery of miR-326, targeting the signaling pathways involved in inflammatory processes. This indicates a critical role of exosomal miRNAs in mediating the therapeutic effects of MSC-derived exosomes.

Exosomes from other stem cell types

Adipose-derived stem cells (ADSCs) have also emerged as a source of therapeutic exosomes in OA (100). Wang et al (101) demonstrated that ADSCs-derived exosomes modulate chondrocyte metabolism, particularly glutamine metabolism, thus alleviating the progression of OA. Hypoxia-treated ADSCs-derived exosomes have been found to attenuate lumbar facet joint OA by delivering pro-survival signals (102). These findings suggest that the microenvironment and hypoxic preconditioning may enhance the exosomal therapeutic potential in joint diseases. Notably, Fu et al (103) identified that dental pulp stem cell-derived exosomes alleviate OA by inhibiting transient receptor potential cation channel subfamily V member 4-mediated osteoclast activation, underscoring the immunomodulatory capabilities of exosomal therapies in joint disease. Additionally, Meng et al (104) showcased that exosomal miR-429 from ADSCs can ameliorate chondral injury by targeting fasciculation and elongation protein ζ2 and promoting autophagy, highlighting the role of miRNAs in cartilage repair mechanisms.

Various novel sources of exosomes are emerging, including exosomes derived from human umbilical cord MSCs (105) and placenta-derived exosomes (106). The latter study characterized placenta-derived exosomes as a potential disease-modifying therapy for OA, emphasizing their unique biochemical composition and low immunogenicity, which make them suitable candidates for therapeutic applications.

Engineered exosomes and modern delivery systems

Advancements in bioengineering have enhanced the therapeutic efficacy of exosomes. Zhao et al (107) reported that engineering exosomes from subcutaneous fat MSCs specifically promotes cartilage repair through targeted delivery of miR-199a-3p, showcasing an innovative approach to enhance the specificity and efficacy of exosomal therapy. Similarly, Yang et al (108) developed magnetic polysaccharide-based microcarriers loaded with MSC-derived exosomes, thereby enhancing targeted delivery and synergistic effects for OA treatment. Furthermore, Ma et al (109) explored the use of engineered exosomes with activating transcription factor 5-modified mRNA loaded into thermosensitive hydrogels, demonstrating that this novel delivery system can alleviate OA through mitochondrial signaling pathways. Such advancements indicate a shift towards more tailored exosomal therapies that could optimize treatment outcomes in OA.

In previous studies, the preconditioning of donor cells (hypoxia preconditioning or biochemical treatment) has been shown to enhance the therapeutic efficacy of exosomes (102), as highlighted by the findings of Zhao et al (102), where hypoxia enhanced the protective effect of ADSC-derived exosomes on lumbar facet joint OA. The application of exosomes derived from various stem cells represents a promising strategy for OA treatment, with preclinical studies revealing their potential to modulate inflammatory responses, promote cartilage repair and improve joint function (92,101,105,106). As research continues to unveil the complexities of exosomal biology and engineering, future studies should focus on optimizing exosomal therapies and translating these findings into clinical settings to address the considerable burden of OA.

Clinical trials

Recent advancements in regenerative medicine have highlighted the potential of EVs, particularly those derived from umbilical cord tissues, as innovative therapeutic agents in the treatment of OA (110). The present review summarizes the current clinical research on the application of exosomes in OA therapy, focusing on findings and implications, while also highlighting the need for further studies.

Clinical trials have begun to explore the safety and efficacy of umbilical cord-derived Wharton's jelly (UCWJ) as a novel treatment for knee OA (95,96). In a study, UCWJ has been compared with traditional therapies, such as hyaluronic acid and saline, with the study design being a randomized, controlled, single-blind, multi-center trial (95). This approach is significant as it aims to evaluate not only the safety but also the potential benefits of UCWJ in improving knee function and alleviating pain in patients with OA. The anticipated results of the study are expected to shed light on the therapeutic benefits of UCWJ exosomes. Preliminary findings from earlier research have indicated that exosomes derived from Wharton's jelly may possess anti-inflammatory properties and the ability to enhance cartilage regeneration, providing a biological advantage over conventional treatments (95). These exosomes may stimulate cellular processes responsible for cartilage repair and modulate inflammatory pathways in the joint environment. Furthermore, Gupta et al (96) also conducted a non-randomized, open-label, multi-center trial to further assess the therapeutic potential of UCWJ in OA treatment. The focus was on the functional outcomes of patients as well as the severity of symptoms post-treatment. Collectively, these studies have highlighted the promising role of UCWJ in reducing pain and improving joint function in patients with OA. The findings underscore the potential for UCWJ to revolutionize current treatment options and enhance the quality of life of patients.

Currently, the research landscape regarding exosomal therapies in OA is expanding, with various studies underway (NCT06466850, NCT06431152 and NCT05060107). The emphasis is shifting towards understanding the underlying mechanisms by which exosomes influence cartilage health, inflammatory responses and joint function (73,111). In addition to UCWJ, studies are also exploring exosomes from other sources, such as ADSCs and bone marrow-derived stem cells (98,99). However, the current body of research remains limited, focusing primarily on specific types of exosomes rather than a broader spectrum of availed cellular sources. A number of ongoing studies (NCT06466850, NCT06431152 and NCT06463132) are expected to assess the impact of these therapeutic agents on various patient populations, different OA stages and their combinatory effects with other treatment modalities.

While the current clinical research indicates a promising direction for exosome-based therapies in OA (98,99), there is only one randomized controlled study and only preliminary efficacy has been achieved (95). Future studies should adopt more rigorous methodologies, including larger sample sizes and randomized controlled designs, to validate the efficacy and safety of exosome treatments. Additionally, research should focus on identifying predictive biomarkers that can help select the most suitable patients for exosome therapy, enhancing personalized treatment approaches. Continued clinical research and refinement of methodologies will be crucial for translating these findings into safe and effective therapies for patients, ultimately improving the management of OA.

Challenges in exosome-based OA treatment

The challenges associated with exosome-based therapies for OA are multifaceted, encompassing technical, biological and regulatory dimensions (112). Addressing these challenges will require collaborative efforts among researchers, clinicians, regulatory bodies and ethical committees to ensure that exosome therapies can be effectively developed and translated into clinical practice. By overcoming these hurdles, exosome-based strategies can ultimately lead to innovative and improved treatments that benefit patients suffering from OA, potentially altering the management landscape of this debilitating condition.

Technical challenges
Isolation and characterization of exosomes

A technical challenge in the field of exosome therapy lies in the isolation and characterization of exosomes (113), which can impact research and clinical applications. Exosomes are nanoscale EVs that vary in size, composition and functional roles, making standardization of isolation methods crucial (114). Currently, a variety of isolation techniques exist, including ultracentrifugation, ultrafiltration and commercial isolation kits, each with its advantages and disadvantages (114). Ultracentrifugation is highly effective for isolating pure exosomes but is time-consuming and requires specialized equipment. Ultrafiltration offers faster processing times and can concentrate exosomes but may lead to some loss of sample quality due to filter retention. Commercial separation kits are user-friendly and convenient but can be expensive and may result in lower yields. Overall, the choice of method depends on the specific research needs, sample type and available resources (114). One major issue is the lack of standardized protocols, leading to variability in yield and purity (115). As noted by Yadav et al (115), standardized reporting is crucial for ensuring rigor and reproducibility in exosome research. Furthermore, existing purification methods, such as ultracentrifugation and affinity chromatography, have limitations in specificity and efficiency (116). These challenges must be addressed to enhance the reliability of exosome studies and facilitate the eventual application of exosomes in OA.

The isolation process can lead to contamination with other EVs or proteins, compromising the purity and functional efficacy of the exosome preparation (117). Furthermore, variations in yield between and within different biological sources such as stem cells, platelets or other tissues can introduce inconsistencies in exosomal properties (118). As a result, the difficulty in achieving consistent and reproducible isolation hampers the ability to conduct comparative studies and can complicate the validation of treatment protocols.

In addition to isolation, there is a pressing need for reliable characterization techniques to assess the size, concentration and content of exosomes. Characterization methodologies typically involve nanoparticle tracking analysis, dynamic light scattering and electron microscopy, as well as profiling bioactive molecules through proteomic and genomic analyses (113). Fluctuations in these parameters can markedly influence the anticipated efficacy of the therapy (113), underscoring the need for robust quality control measures to ensure that only high-quality exosome preparations are utilized in clinical applications.

Delivery mechanisms and targeting

Another crucial technical hurdle is the development of effective delivery mechanisms that can ensure the successful targeting of exosomes to the desired tissues in patients with OA (119). While exosomes possess inherent targeting capabilities due to their lipid bilayer and surface markers that can facilitate cellular uptake, these natural targeting mechanisms alone are often insufficient for achieving optimal therapeutic outcomes (120). One major concern is the bioavailability of exosomes following administration. Traditional delivery routes such as intravenous or intra-articular injections may result in rapid systemic distribution and clearance, thereby limiting the local therapeutic concentration of exosomes at the target site, specifically the affected joints (121). Researchers are exploring innovative delivery strategies, including the use of hydrogels (110), nanoparticles (122) or other biomaterials as carriers for exosomes, which could enhance the retention and controlled release of exosomes at the joint site.

Optimizing the surface modification of exosomes to improve targeting specificity is critical. Genetic engineering or chemical modifications may enhance receptor-mediated targeting, increasing the likelihood of exosomes interacting with chondrocytes and other relevant cell types in the joint (123,124). Nonetheless, these adaptations may introduce complex regulatory and safety considerations, necessitating further research and validation before clinical use.

Biological hurdles
Variability in exosome content

Biological variability in exosome content poses a challenge to the standardization and predictability of exosome-based therapies (125). The molecular makeup of exosomes, including proteins, lipids, mRNAs and non-coding RNAs, can differ based on their cellular source, culture conditions and even the physiological state of the donor organism (126). This variability can lead to inconsistent therapeutic effects when using exosomes derived from different sources or batches (127). For instance, exosomes derived from MSCs exhibit similar characteristic; however, their regenerative potential may differ based on the tissue source such as the bone marrow or adipose tissue. This inconsistency can make it challenging to predict treatment responses in patients with OA, as different exosomal preparations may yield varying degrees of anti-inflammatory effects or regenerative outcomes (127). Additionally, factors such as age, sex and the severity of OA can lead to alterations in exosome composition, further contributing to variability in experimental outcomes (127).

Variability in exosome isolation techniques can introduce discrepancies in the assessments of exosomal content (128). Methods such as ultrafiltration, precipitation and affinity-based techniques can yield exosomes of different purity and yield, complicating comparisons across studies (129). This variability poses challenges for the reproducibility and reliability of therapeutic results, as different exosomal profiles may yield divergent biological effects when applied in therapeutic contexts (129).

To tackle the challenges of exosome variability, it is crucial to standardize isolation and characterization protocols. Strict quality control measures, including the use of reference materials and calibration standards, alongside the establishment of consensus guidelines for exosome research, can enhance reproducibility (125). Additionally, integrating advanced techniques such as single-exosome characterization and multi-omics approaches will yield deeper insights into exosomal heterogeneity and its relevance to OA treatment (130). Coupling machine learning algorithms with high-throughput screening could further identify specific exosomal signatures associated with OA (131). Implementing standardized exosome handling and storage protocols will ensure consistency, ultimately aiding in the validation of exosomes as reliable biomarkers or therapeutic agents in OA management (132).

Long-term effects of treatment

While the short-term effects of exosome therapies are gaining positive preliminary results, understanding the long-term outcomes and safety is crucial for establishing their clinical relevance (133). At present, to the best of our knowledge, there are no clinical data on long-term administration of exosomes for OA, especially in terms of immune response, potential immunogenicity and tissue side effects (78).

One concern is the possibility of unknown long-term effects on local tissues or systemic immunity (134). The administration of foreign exosomal material, even from autologous sources, may elicit immune responses in some patients, particularly in those with underlying inflammatory conditions (135). Long-term follow-up studies are essential to monitor potential adverse events associated with prolonged use of exosome-based therapies. Additionally, the regenerative effects of exosome therapies need thorough investigation in terms of sustainability. It remains unclear whether the benefits observed after treatment will persist over time or if repeat administrations will be required for sustained therapeutic effects. Ongoing research must focus on defining the optimal dosing strategies, frequency of administration and long-term monitoring to mitigate any adverse effects while maximizing regenerative benefits.

Regulatory and ethical considerations
Approval processes for clinical applications

One of the most significant hurdles faced by exosome-based therapies is navigating the complex regulatory landscape. As biologics derived from living cells, exosomes fall under stringent regulations which vary across different regions and countries (136). Regulatory agencies require comprehensive data on the safety, efficacy, manufacturing and quality control of exosome-based products, which can be daunting for developers (137). The Food and Drug Administration categorizes exosomes as biological products (138); however, their specific classification can be ambiguous, leading to uncertainties in regulatory pathways. Unlike traditional pharmaceutical products, exosomes are derived from living cells, which introduces variability in their composition, function and manufacturing process, complicating the establishment of standardized protocols for their development and approval (139).

Regulations related to cell therapies impact exosome research. Current guidelines primarily focus on the source, manufacturing and clinical application of cells, often neglecting the unique aspects of exosome therapeutics. For example, compliance with Good Manufacturing Practices in the production of exosomes requires rigorous documentation and control of starting materials, processing, storage and quality testing (140). This can be particularly challenging given the inherent variability in exosome isolation techniques and characterization methodologies.

Safety assessments for exosome-based therapeutics must address risks associated with immune reactions or unintended biological effects, which are not yet fully understood (141). This necessitates extensive preclinical studies and clinical trials, aligning with a risk-benefit analysis approach typically reserved for more conventional therapies.

To navigate these regulatory hurdles, researchers must advocate for the development of specific guidelines tailored to exosome-based therapies. By engaging with regulatory agencies early in the research process, scientists can better clarify regulatory expectations and streamline the pathway for exosome applications in the treatment of OA. Establishing collaborative frameworks among researchers, clinicians and regulators will be crucial for advancing this field and ensuring that innovative exosome-based therapies can be safely and effectively translated into clinical practice.

Ethical concerns related to isolation sources

The ethical considerations surrounding the sources of exosome isolation present another challenge in the development of exosome-based therapies. Exosomes can be derived from various sources, including stem cells, body fluids and tissues. However, ethical concerns may arise, especially when it comes to the need to obtain tissue samples or cells from human donors (142).

The use of embryonic stem cells for exosome production raises complex ethical dilemmas and concerns related to consent, donor rights and the implications of using embryonic tissue (143). Additionally, the collection of exosomes from human adipose or bone marrow sources necessitates ensuring that such procedures are performed ethically, with informed consent from donors and robust transparency regarding the implications of their contributions (144). Furthermore, as exosome therapies move forward, establishing clear guidelines and ethical frameworks for the use of both animal and human-derived materials will be crucial. This ensures that researchers and clinicians prioritize patient rights and societal norms while advancing the scientific understanding and therapeutic applications (145).

Discussion

Exosomes are nano-sized EVs derived from various cell types, have emerged as crucial mediators of intercellular communication in the bone microenvironment and serve a role in the pathogenesis of OA (20). Previous studies have demonstrated that exosomes released from chondrocytes, synovial cells and MSCs contain a variety of bioactive molecules, including proteins, lipids and miRNAs, which can modulate inflammation, cartilage repair and apoptosis, which are key processes in OA development and progression (22,32,59).

The therapeutic potential of exosomes lies in their ability to deliver regenerative signals to target cells. For instance, MSC-derived exosomes have shown promise in promoting cartilage regeneration and inhibiting inflammatory responses in OA models (90,91). Exosomes can facilitate chondrocyte proliferation and enhance the production of ECM components, thereby holding potential as a novel regenerative therapy (104). Furthermore, their inherent ability to mimic the properties of their parent cells while providing a safer alternative due to lower risk of immune rejection further solidifies their role in OA treatment (104).

However, the clinical application of exosomes in OA management is still faces several challenges. First, the heterogeneity of exosomes derived from different cell types and the variations in their composition raise concerns regarding standardization and quality control (114). Additionally, the mechanisms underlying their therapeutic actions remain poorly understood, which complicates the design of targeted therapies. Furthermore, the pharmacokinetics and biodistribution of exosomes in OA models need to be extensively evaluated to optimize their effectiveness.

Therefore, while exosomes represent a promising avenue for OA therapy, further research is essential to unlock their full potential. Future studies should focus on elucidating the specific roles of exosomal cargo in OA, standardizing exosome preparation techniques and conducting clinical trials to assess their safety and efficacy. By addressing these challenges, exosomal therapies may transform the current landscape of OA treatment, potentially improving patient outcomes.

Conclusion

In conclusion, exosomes serve a crucial role in the pathophysiology of OA, influencing inflammation and cartilage degradation. However, gaps remain in understanding the specific molecular mechanisms of exosomal signaling in OA progression. Future research should focus on identifying key exosomal biomarkers for early diagnosis and developing targeted exosome-based therapies. Additionally, optimizing exosome isolation and characterization methods will enhance the therapeutic potential of exosomes. Collaborative efforts to standardize protocols and validate their efficacy in clinical trials are essential for advancing exosome-centered innovations in OA treatment.

Availability of data and materials

Not applicable.

Authors' contributions

XC, BT, YW and XK made substantial contributions to the conception and design of the manuscript. XC and XK performed acquisition, analysis and interpretation of data. XC, BT, YW, JZ and XK drafted and wrote the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

No funding was received.

References

1 

Yao Q, Wu X, Tao C, Gong W, Chen M, Qu M, Zhong Y, He T, Chen S and Xiao G: Osteoarthritis: Pathogenic signaling pathways and therapeutic targets. Signal Transduct Target Ther. 8:562023. View Article : Google Scholar : PubMed/NCBI

2 

Barnett R: Osteoarthritis. Lancet. 391:19852018. View Article : Google Scholar : PubMed/NCBI

3 

Englund M: Osteoarthritis, part of life or a curable disease? A bird's-eye view. J Intern Med. 293:681–693. 2023. View Article : Google Scholar : PubMed/NCBI

4 

Schlenk EA, Fitzgerald GK, Rogers JC, Kwoh CK and Sereika SM: Promoting physical activity in older adults with knee osteoarthritis and hypertension: A randomized controlled trial. J Aging Phys Act. 29:207–218. 2021. View Article : Google Scholar :

5 

Hawker GA and King LK: The burden of osteoarthritis in older adults. Clin Geriatr Med. 38:181–192. 2022. View Article : Google Scholar : PubMed/NCBI

6 

Jiang W, Chen H, Lin Y, Cheng K, Zhou D, Chen R, Song C, Zeng L and Yu H: Mechanical stress abnormalities promote chondrocyte senescence-the pathogenesis of knee osteoarthritis. Biomed Pharmacother. 167:1155522023. View Article : Google Scholar

7 

Sanchez-Lopez E, Coras R, Torres A, Lane NE and Guma M: Synovial inflammation in osteoarthritis progression. Nat Rev Rheumatol. 18:258–275. 2022. View Article : Google Scholar : PubMed/NCBI

8 

Rim YA, Nam Y and Ju JH: The role of chondrocyte hypertrophy and senescence in osteoarthritis initiation and progression. Int J Mol Sci. 21:23582020. View Article : Google Scholar : PubMed/NCBI

9 

Abramoff B and Caldera FE: Osteoarthritis: Pathology, diagnosis, and treatment options. Med Clin North Am. 104:293–311. 2020. View Article : Google Scholar : PubMed/NCBI

10 

Emami A, Namdari H, Parvizpour F and Arabpour Z: Challenges in osteoarthritis treatment. Tissue Cell. 80:1019922023. View Article : Google Scholar

11 

Danilushkina AA, Emene CC, Barlev NA and Gomzikova MO: Strategies for engineering of extracellular vesicles. Int J Mol Sci. 24:132472023. View Article : Google Scholar : PubMed/NCBI

12 

Zhang Y, Dou Y, Liu Y, Di M, Bian H, Sun X and Yang Q: Advances in therapeutic applications of extracellular vesicles. Int J Nanomedicine. 18:3285–3307. 2023. View Article : Google Scholar : PubMed/NCBI

13 

Gurunathan S, Kang MH, Song H, Kim NH and Kim JH: The role of extracellular vesicles in animal reproduction and diseases. J Anim Sci Biotechnol. 13:622022. View Article : Google Scholar : PubMed/NCBI

14 

Ni Z, Zhou S, Li S, Kuang L, Chen H, Luo X, Ouyang J, He M, Du X and Chen L: Exosomes: Roles and therapeutic potential in osteoarthritis. Bone Res. 8:252020. View Article : Google Scholar : PubMed/NCBI

15 

Fan Y, Li Z and He Y: Exosomes in the pathogenesis, progression, and treatment of osteoarthritis. Bioengineering (Basel). 9:992022. View Article : Google Scholar : PubMed/NCBI

16 

Yan J, Shen M, Sui B, Lu W, Han X, Wan Q, Liu Y, Kang J, Qin W, Zhang Z, et al: Autophagic LC3+ calcified extracellular vesicles initiate cartilage calcification in osteoarthritis. Sci Adv. 8:eabn15562022. View Article : Google Scholar

17 

Zhou H, Shen X, Yan C, Xiong W, Ma Z, Tan Z, Wang J, Li Y, Liu J, Duan A and Liu F: Extracellular vesicles derived from human umbilical cord mesenchymal stem cells alleviate osteoarthritis of the knee in mice model by interacting with METTL3 to reduce m6A of NLRP3 in macrophage. Stem Cell Res Ther. 13:3222022. View Article : Google Scholar : PubMed/NCBI

18 

Zou J, Yang W, Cui W, Li C, Ma C, Ji X, Hong J, Qu Z, Chen J, Liu A and Wu H: Therapeutic potential and mechanisms of mesenchymal stem cell-derived exosomes as bioactive materials in tendon-bone healing. J Nanobiotechnology. 21:142023. View Article : Google Scholar : PubMed/NCBI

19 

Krylova SV and Feng D: The machinery of exosomes: Biogenesis, release, and uptake. Int J Mol Sci. 24:13372023. View Article : Google Scholar : PubMed/NCBI

20 

Xie F, Liu YL, Chen XY, Li Q, Zhong J, Dai BY, Shao XF and Wu GB: Role of MicroRNA, LncRNA, and exosomes in the progression of osteoarthritis: A review of recent literature. Orthop Surg. 12:708–716. 2020. View Article : Google Scholar : PubMed/NCBI

21 

Miao C, Zhou W, Wang X and Fang J: The research progress of exosomes in osteoarthritis, with particular emphasis on the mediating roles of miRNAs and lncRNAs. Front Pharmacol. 12:6856232021. View Article : Google Scholar : PubMed/NCBI

22 

Asghar S, Litherland GJ, Lockhart JC, Goodyear CS and Crilly A: Exosomes in intercellular communication and implications for osteoarthritis. Rheumatology (Oxford). 59:57–68. 2020.

23 

Sheta M, Taha EA, Lu Y and Eguchi T: Extracellular vesicles: New classification and tumor immunosuppression. Biology (Basel). 12:1102023.PubMed/NCBI

24 

Zhou Q, Cai Y, Jiang Y and Lin X: Exosomes in osteoarthritis and cartilage injury: Advanced development and potential therapeutic strategies. Int J Biol Sci. 16:1811–1820. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Liu Q, Wang R, Hou S, He F, Ma Y, Ye T, Yu S, Chen H, Wang H and Zhang M: Chondrocyte-derived exosomes promote cartilage calcification in temporomandibular joint osteoarthritis. Arthritis Res Ther. 24:442022. View Article : Google Scholar : PubMed/NCBI

26 

Sang X, Zhao X, Yan L, Jin X, Wang X, Wang J, Yin Z, Zhang Y and Meng Z: Thermosensitive hydrogel loaded with primary chondrocyte-derived exosomes promotes cartilage repair by regulating macrophage polarization in osteoarthritis. Tissue Eng Regen Med. 19:629–642. 2022. View Article : Google Scholar : PubMed/NCBI

27 

Wu X, Crawford R, Xiao Y, Mao X and Prasadam I: Osteoarthritic subchondral bone release exosomes that promote cartilage degeneration. Cells. 10:2512021. View Article : Google Scholar : PubMed/NCBI

28 

Kim YG, Choi J and Kim K: Mesenchymal stem cell-derived exosomes for effective cartilage tissue repair and treatment of osteoarthritis. Biotechnol J. 15:e20000822020. View Article : Google Scholar : PubMed/NCBI

29 

Skotland T, Sandvig K and Llorente A: Lipids in exosomes: Current knowledge and the way forward. Prog Lipid Res. 66:30–41. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Kalluri R and LeBleu VS: The biology, function, and biomedical applications of exosomes. Science. 367:eaau69772020. View Article : Google Scholar : PubMed/NCBI

31 

Mihanfar A, Shakouri SK, Khadem-Ansari MH, Fattahi A, Latifi Z, Nejabati HR and Nouri M: Exosomal miRNAs in osteoarthritis. Mol Biol Rep. 47:4737–4748. 2020. View Article : Google Scholar : PubMed/NCBI

32 

Kong R, Ji L, Pang Y, Zhao D and Gao J: Exosomes from osteoarthritic fibroblast-like synoviocytes promote cartilage ferroptosis and damage via delivering microRNA-19b-3p to target SLC7A11 in osteoarthritis. Front Immunol. 14:11811562023. View Article : Google Scholar : PubMed/NCBI

33 

Mao G, Zhang Z, Hu S, Zhang Z, Chang Z, Huang Z, Liao W and Kang Y: Exosomes derived from miR-92a-3p-overexpressing human mesenchymal stem cells enhance chondrogenesis and suppress cartilage degradation via targeting WNT5A. Stem Cell Res Ther. 9:2472018. View Article : Google Scholar : PubMed/NCBI

34 

Qiu M, Liu D and Fu Q: MiR-129-5p shuttled by human synovial mesenchymal stem cell-derived exosomes relieves IL-1β induced osteoarthritis via targeting HMGB1. Life Sci. 269:1189872021. View Article : Google Scholar

35 

Xia Q, Wang Q, Lin F and Wang J: miR-125a-5p-abundant exosomes derived from mesenchymal stem cells suppress chondrocyte degeneration via targeting E2F2 in traumatic osteoarthritis. Bioengineered. 12:11225–11238. 2021. View Article : Google Scholar : PubMed/NCBI

36 

Tao Y, Zhou J, Wang Z, Tao H, Bai J, Ge G, Li W, Zhang W, Hao Y, Yang X and Geng D: Human bone mesenchymal stem cells-derived exosomal miRNA-361-5p alleviates osteoarthritis by downregulating DDX20 and inactivating the NF-κB signaling pathway. Bioorg Chem. 113:1049782021. View Article : Google Scholar

37 

Li X, Wang Y, Cai Z, Zhou Q, Li L and Fu P: Exosomes from human umbilical cord mesenchymal stem cells inhibit ROS production and cell apoptosis in human articular chondrocytes via the miR-100-5p/NOX4 axis. Cell Biol Int. 45:2096–2106. 2021. View Article : Google Scholar : PubMed/NCBI

38 

Ye P, Mi Z, Wei D, Gao P, Ma M and Yang H: miR-3960 from mesenchymal stem cell-derived extracellular vesicles inactivates SDC1/Wnt/β-catenin axis to relieve chondrocyte injury in osteoarthritis by targeting PHLDA2. Stem Cells Int. 2022:94551522022. View Article : Google Scholar

39 

Foo JB, Looi QH, How CW, Lee SH, Al-Masawa ME, Chong PP and Law JX: Mesenchymal stem cell-derived exosomes and MicroRNAs in cartilage regeneration: Biogenesis, efficacy, miRNA enrichment and delivery. Pharmaceuticals (Basel). 14:10932021. View Article : Google Scholar : PubMed/NCBI

40 

Wang Z, Yan K, Ge G, Zhang D, Bai J, Guo X, Zhou J, Xu T, Xu M, Long X, et al: Exosomes derived from miR-155-5p-overexpressing synovial mesenchymal stem cells prevent osteoarthritis via enhancing proliferation and migration, attenuating apoptosis, and modulating extracellular matrix secretion in chondrocytes. Cell Biol Toxicol. 37:85–96. 2021. View Article : Google Scholar

41 

Qian Y, Chu G, Zhang L, Wu Z, Wang Q, Guo JJ and Zhou F: M2 macrophage-derived exosomal miR-26b-5p regulates macrophage polarization and chondrocyte hypertrophy by targeting TLR3 and COL10A1 to alleviate osteoarthritis. J Nanobiotechnology. 22:722024. View Article : Google Scholar : PubMed/NCBI

42 

Yoo KH, Thapa N, Chwae YJ, Yoon SH, Kim BJ, Lee JO, Jang YN and Kim J: Transforming growth factor-β family and stem cell-derived exosome therapeutic treatment in osteoarthritis (Review). Int J Mol Med. 49:622022. View Article : Google Scholar

43 

Du X, Cai L, Xie J and Zhou X: The role of TGF-beta3 in cartilage development and osteoarthritis. Bone Res. 11:22023. View Article : Google Scholar : PubMed/NCBI

44 

Lambert C, Dubuc JE, Montell E, Vergés J, Munaut C, Noël A and Henrotin Y: Gene expression pattern of cells from inflamed and normal areas of osteoarthritis synovial membrane. Arthritis Rheumatol. 66:960–968. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Tsai CH, Liu SC, Chung WH, Wang SW, Wu MH and Tang CH: Visfatin increases VEGF-dependent angiogenesis of endothelial progenitor cells during osteoarthritis progression. Cells. 9:13152020. View Article : Google Scholar : PubMed/NCBI

46 

Zou ZL, Sun MH, Yin WF, Yang L and Kong LY: Avicularin suppresses cartilage extracellular matrix degradation and inflammation via TRAF6/MAPK activation. Phytomedicine. 91:1536572021. View Article : Google Scholar : PubMed/NCBI

47 

Wang L and He C: Nrf2-mediated anti-inflammatory polarization of macrophages as therapeutic targets for osteoarthritis. Front Immunol. 13:9671932022. View Article : Google Scholar : PubMed/NCBI

48 

Lin S, Li H, Wu B, Shang J, Jiang N, Peng R, Xing B, Xu X and Lu H: TGF-β1 regulates chondrocyte proliferation and extracellular matrix synthesis via circPhf21a-Vegfa axis in osteoarthritis. Cell Commun Signal. 20:752022. View Article : Google Scholar

49 

Ge Y, Xu W, Chen Z, Zhang H, Zhang W, Chen J, Huang J, Du W, Tong P, Shan L and Zhou L: Nanofat lysate ameliorates pain and cartilage degradation of osteoarthritis through activation of TGF-β-Smad2/3 signaling of chondrocytes. Front Pharmacol. 14:9002052023. View Article : Google Scholar

50 

Zhong Y, Xu Y, Xue S, Zhu L, Lu H, Wang C, Chen H, Sang W and Ma J: Nangibotide attenuates osteoarthritis by inhibiting osteoblast apoptosis and TGF-β activity in subchondral bone. Inflammopharmacology. 30:1107–1117. 2022. View Article : Google Scholar : PubMed/NCBI

51 

Sun K, Luo J, Guo J, Yao X, Jing X and Guo F: The PI3K/AKT/mTOR signaling pathway in osteoarthritis: A narrative review. Osteoarthritis Cartilage. 28:400–409. 2020. View Article : Google Scholar : PubMed/NCBI

52 

Wang L, Xu H, Li X, Chen H, Zhang H, Zhu X, Lin Z, Guo S, Bao Z, Rui H, et al: Cucurbitacin E reduces IL-1β-induced inflammation and cartilage degeneration by inhibiting the PI3K/Akt pathway in osteoarthritic chondrocytes. J Transl Med. 21:8802023. View Article : Google Scholar

53 

Zhang Y, Pizzute T and Pei M: A review of crosstalk between MAPK and Wnt signals and its impact on cartilage regeneration. Cell Tissue Res. 358:633–649. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Yan D, Chen D and Im HJ: Fibroblast growth factor-2 promotes catabolism via FGFR1-Ras-Raf-MEK1/2-ERK1/2 axis that coordinates with the PKCδ pathway in human articular chondrocytes. J Cell Biochem. 113:2856–2865. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Ge Q, Wang H, Xu X, Xu L, Zhai L and Tao R: PDK1 promotes apoptosis of chondrocytes via modulating MAPK pathway in osteoarthritis. Tissue Cell. 49:719–725. 2017. View Article : Google Scholar : PubMed/NCBI

56 

Saha S and Rebouh NY: Anti-osteoarthritis mechanism of the Nrf2 signaling pathway. Biomedicines. 11:31762023. View Article : Google Scholar : PubMed/NCBI

57 

Shang X, Fang Y, Xin W and You H: The application of extracellular vesicles mediated miRNAs in osteoarthritis: Current knowledge and perspective. J Inflamm Res. 15:2583–2599. 2022. View Article : Google Scholar : PubMed/NCBI

58 

Duan L, Liang Y, Xu X, Xiao Y and Wang D: Recent progress on the role of miR-140 in cartilage matrix remodelling and its implications for osteoarthritis treatment. Arthritis Res Ther. 22:1942020. View Article : Google Scholar : PubMed/NCBI

59 

Mori T, Giovannelli L, Bilia AR and Margheri F: Exosomes: Potential next-generation nanocarriers for the therapy of inflammatory diseases. Pharmaceutics. 15:22762023. View Article : Google Scholar : PubMed/NCBI

60 

El Jamal A, Bougault C, Mebarek S, Magne D, Cuvillier O and Brizuela L: The role of sphingosine 1-phosphate metabolism in bone and joint pathologies and ectopic calcification. Bone. 130:1150872020. View Article : Google Scholar

61 

Skotland T, Hessvik NP, Sandvig K and Llorente A: Exosomal lipid composition and the role of ether lipids and phosphoinositides in exosome biology. J Lipid Res. 60:9–18. 2019. View Article : Google Scholar :

62 

Wei G, Lu K, Umar M, Zhu Z, Lu WW, Speakman JR, Chen Y, Tong L and Chen D: Risk of metabolic abnormalities in osteoarthritis: A new perspective to understand its pathological mechanisms. Bone Res. 11:632023. View Article : Google Scholar : PubMed/NCBI

63 

Wang G and Bieberich E: Sphingolipids in neurodegeneration (with focus on ceramide and S1P). Adv Biol Regul. 70:51–64. 2018. View Article : Google Scholar : PubMed/NCBI

64 

Toh WS, Lai RC, Hui JHP and Lim SK: MSC exosome as a cell-free MSC therapy for cartilage regeneration: Implications for osteoarthritis treatment. Semin Cell Dev Biol. 67:56–64. 2017. View Article : Google Scholar

65 

Cosenza S, Ruiz M, Maumus M, Jorgensen C and Noël D: Pathogenic or therapeutic extracellular vesicles in rheumatic diseases: Role of mesenchymal stem cell-derived vesicles. Int J Mol Sci. 18:8892017. View Article : Google Scholar : PubMed/NCBI

66 

Chen M, Wang Q, Wang Y, Fan Y and Zhang X: Biomaterials-assisted exosomes therapy in osteoarthritis. Biomed Mater. 17:0220012022. View Article : Google Scholar

67 

Yang B, Li X, Fu C, Cai W, Meng B, Qu Y, Kou X and Zhang Q: Extracellular vesicles in osteoarthritis of peripheral joint and temporomandibular joint. Front Endocrinol (Lausanne). 14:11587442023. View Article : Google Scholar : PubMed/NCBI

68 

Liu Y, Lin L, Zou R, Wen C, Wang Z and Lin F: MSC-derived exosomes promote proliferation and inhibit apoptosis of chondrocytes via lncRNA-KLF3-AS1/miR-206/GIT1 axis in osteoarthritis. Cell Cycle. 17:2411–2422. 2018. View Article : Google Scholar : PubMed/NCBI

69 

Xie L, Chen Z, Liu M, Huang W, Zou F, Ma X, Tao J, Guo J, Xia X, Lyu F, et al: MSC-derived exosomes protect vertebral endplate chondrocytes against apoptosis and calcification via the miR-31-5p/ATF6 axis. Mol Ther Nucleic Acids. 22:601–614. 2020. View Article : Google Scholar : PubMed/NCBI

70 

Tang S, Tang T, Gao G, Wei Q, Sun K and Huang W: Bone marrow mesenchymal stem cell-derived exosomes inhibit chondrocyte apoptosis and the expression of MMPs by regulating Drp1-mediated mitophagy. Acta Histochem. 123:1517962021. View Article : Google Scholar : PubMed/NCBI

71 

Zheng L, Wang Y, Qiu P, Xia C, Fang Y, Mei S, Fang C, Shi Y, Wu K, Chen Z, et al: Primary chondrocyte exosomes mediate osteoarthritis progression by regulating mitochondrion and immune reactivity. Nanomedicine (Lond). 14:3193–3212. 2019. View Article : Google Scholar : PubMed/NCBI

72 

Withrow J, Murphy C, Liu Y, Hunter M, Fulzele S and Hamrick MW: Extracellular vesicles in the pathogenesis of rheumatoid arthritis and osteoarthritis. Arthritis Res Ther. 18:2862016. View Article : Google Scholar : PubMed/NCBI

73 

Yu H, Huang Y and Yang L: Research progress in the use of mesenchymal stem cells and their derived exosomes in the treatment of osteoarthritis. Ageing Res Rev. 80:1016842022. View Article : Google Scholar : PubMed/NCBI

74 

Zhou Y, Ming J, Li Y, Li B, Deng M, Ma Y, Chen Z, Zhang Y, Li J and Liu S: Exosomes derived from miR-126-3p-overexpressing synovial fibroblasts suppress chondrocyte inflammation and cartilage degradation in a rat model of osteoarthritis. Cell Death Discov. 7:372021. View Article : Google Scholar : PubMed/NCBI

75 

Szponder T, Latalski M, Danielewicz A, Krać K, Kozera A, Drzewiecka B, Nguyen Ngoc D, Dobko D and Wessely-Szponder J: Osteoarthritis: Pathogenesis, animal models, and new regenerative therapies. J Clin Med. 12:52022. View Article : Google Scholar

76 

Liu B, Xian Y, Chen X, Shi Y, Dong J, Yang L, An X, Shen T, Wu W, Ma Y, et al: Inflammatory fibroblast-like synoviocyte-derived exosomes aggravate osteoarthritis via enhancing macrophage glycolysis. Adv Sci (Weinh). 11:e23073382024. View Article : Google Scholar : PubMed/NCBI

77 

Wang J, Guo X, Kang Z, Qi L, Yang Y, Wang J, Xu J and Gao S: Roles of exosomes from mesenchymal stem cells in treating osteoarthritis. Cell Reprogram. 22:107–117. 2020. View Article : Google Scholar : PubMed/NCBI

78 

Huang L, Dong G, Peng J, Li T, Zou M, Hu K, Shu Y, Cheng T and Hao L: The role of exosomes and their enhancement strategies in the treatment of osteoarthritis. Hum Cell. 36:1887–1900. 2023. View Article : Google Scholar : PubMed/NCBI

79 

Li F, Xu Z, Xie Z, Sun X, Li C, Chen Y, Xu J and Pi G: Adipose mesenchymal stem cells-derived exosomes alleviate osteoarthritis by transporting microRNA-376c-3p and targeting the WNT-beta-catenin signaling axis. Apoptosis. 28:362–378. 2023. View Article : Google Scholar

80 

Zhang Y, Jia J, Yang S, Liu X, Ye S and Tian H: MicroRNA-21 controls the development of osteoarthritis by targeting GDF-5 in chondrocytes. Exp Mol Med. 46:e792014. View Article : Google Scholar : PubMed/NCBI

81 

Chen Y, Huang H, Zhong W, Li L, Lu Y and Si HB: miR-140-5p protects cartilage progenitor/stem cells from fate changes in knee osteoarthritis. Int Immunopharmacol. 114:1095762023. View Article : Google Scholar

82 

Kim M, Shin DI, Choi BH and Min BH: Exosomes from IL-1β-primed mesenchymal stem cells inhibited IL-1β- and TNF-α-mediated inflammatory responses in osteoarthritic SW982 cells. Tissue Eng Regen Med. 18:525–536. 2021. View Article : Google Scholar : PubMed/NCBI

83 

Wang H, Zhang Y, Zhang C, Zhao Y, Shu J and Tang X: Exosomes derived from miR-146a-overexpressing fibroblast-like synoviocytes in cartilage degradation and macrophage M1 polarization: A novel protective agent for osteoarthritis? Front Immunol. 15:13616062024. View Article : Google Scholar : PubMed/NCBI

84 

Fan WJ, Liu D, Pan LY, Wang WY, Ding YL, Zhang YY, Ye RX, Zhou Y, An SB and Xiao WF: Exosomes in osteoarthritis: Updated insights on pathogenesis, diagnosis, and treatment. Front Cell Dev Biol. 10:9496902022. View Article : Google Scholar : PubMed/NCBI

85 

Selvadoss A, Baby HM, Zhang H and Bajpayee AG: Harnessing exosomes for advanced osteoarthritis therapy. Nanoscale. 16:19174–19191. 2024. View Article : Google Scholar : PubMed/NCBI

86 

Liu Z, Zhuang Y, Fang L, Yuan C, Wang X and Lin K: Breakthrough of extracellular vesicles in pathogenesis, diagnosis and treatment of osteoarthritis. Bioact Mater. 22:423–452. 2022.PubMed/NCBI

87 

Yin B, Ni J, Witherel CE, Yang M, Burdick JA, Wen C and Wong SHD: Harnessing tissue-derived extracellular vesicles for osteoarthritis theranostics. Theranostics. 12:207–231. 2022. View Article : Google Scholar : PubMed/NCBI

88 

Nagelkerke A, Ojansivu M, van der Koog L, Whittaker TE, Cunnane EM, Silva AM, Dekker N and Stevens MM: Extracellular vesicles for tissue repair and regeneration: Evidence, challenges and opportunities. Adv Drug Deliv Rev. 175:1137752021. View Article : Google Scholar : PubMed/NCBI

89 

Wu C, He Y, Yao Y, Yang H and Lu F: Exosomes treating osteoarthritis: Hope with challenge. Heliyon. 9:e131522023. View Article : Google Scholar : PubMed/NCBI

90 

Wang Y, Yu D, Liu Z, Zhou F, Dai J, Wu B, Zhou J, Heng BC, Zou XH, Ouyang H and Liu H: Exosomes from embryonic mesenchymal stem cells alleviate osteoarthritis through balancing synthesis and degradation of cartilage extracellular matrix. Stem Cell Res Ther. 8:1892017. View Article : Google Scholar : PubMed/NCBI

91 

Zhang S, Teo KYW, Chuah SJ, Lai RC, Lim SK and Toh WS: MSC exosomes alleviate temporomandibular joint osteoarthritis by attenuating inflammation and restoring matrix homeostasis. Biomaterials. 200:35–47. 2019. View Article : Google Scholar : PubMed/NCBI

92 

He L, He T, Xing J, Zhou Q, Fan L, Liu C, Chen Y, Wu D, Tian Z, Liu B and Rong L: Bone marrow mesenchymal stem cell-derived exosomes protect cartilage damage and relieve knee osteoarthritis pain in a rat model of osteoarthritis. Stem Cell Res Ther. 11:2762020. View Article : Google Scholar : PubMed/NCBI

93 

Xu H and Xu B: BMSC-derived exosomes ameliorate osteoarthritis by inhibiting pyroptosis of cartilage via delivering mir-326 targeting HDAC3 and STAT1//NF-κB p65 to chondrocytes. Mediators Inflamm. 2021:99728052021. View Article : Google Scholar

94 

Jin Y, Xu M, Zhu H, Dong C, Ji J, Liu Y, Deng A and Gu Z: Therapeutic effects of bone marrow mesenchymal stem cells-derived exosomes on osteoarthritis. J Cell Mol Med. 25:9281–9294. 2021. View Article : Google Scholar : PubMed/NCBI

95 

Gupta A, Maffulli N, Rodriguez HC, Carson EW, Bascharon RA, Delfino K, Levy HJ and El-Amin SF III: Safety and efficacy of umbilical cord-derived Wharton's jelly compared to hyaluronic acid and saline for knee osteoarthritis: Study protocol for a randomized, controlled, single-blind, multi-center trial. J Orthop Surg Res. 16:3522021. View Article : Google Scholar : PubMed/NCBI

96 

Gupta A, Maffulli N, Rodriguez HC, Lee CE, Levy HJ and El-Amin SF III: Umbilical cord-derived Wharton's jelly for treatment of knee osteoarthritis: Study protocol for a non-randomized, open-label, multi-center trial. J Orthop Surg Res. 16:1432021. View Article : Google Scholar : PubMed/NCBI

97 

Jiang K, Jiang T, Chen Y and Mao X: Mesenchymal stem cell-derived exosomes modulate chondrocyte glutamine metabolism to alleviate osteoarthritis progression. Mediators Inflamm. 2021:29791242021. View Article : Google Scholar

98 

Storti G, Scioli MG, Kim BS, Orlandi A and Cervelli V: Adipose-derived stem cells in bone tissue engineering: Useful tools with new applications. Stem Cells Int. 2019:36738572019. View Article : Google Scholar : PubMed/NCBI

99 

Liu M, Sun Y and Zhang Q: Emerging role of extracellular vesicles in bone remodeling. J Dent Res. 97:859–868. 2018. View Article : Google Scholar : PubMed/NCBI

100 

Yin Z, Qin C, Pan S, Shi C, Wu G, Feng Y, Zhang J, Yu Z, Liang B and Gui J: Injectable hyperbranched PEG crosslinked hyaluronan hydrogel microparticles containing mir-99a-3p modified subcutaneous ADSCs-derived exosomes was beneficial for long-term treatment of osteoarthritis. Mater Today Bio. 23:1008132023. View Article : Google Scholar : PubMed/NCBI

101 

Wang Y, Fan A, Lu L, Pan Z, Ma M, Luo S, Liu Z, Yang L, Cai J and Yin F: Exosome modification to better alleviates endoplasmic reticulum stress induced chondrocyte apoptosis and osteoarthritis. Biochem Pharmacol. 206:1153432022. View Article : Google Scholar : PubMed/NCBI

102 

Zhao J, Sun Y, Sheng X, Xu J, Dai G, He R, Jin Y, Liu Z, Xie Y, Wu T, et al: Hypoxia-treated adipose mesenchymal stem cell-derived exosomes attenuate lumbar facet joint osteoarthritis. Mol Med. 29:1202023. View Article : Google Scholar : PubMed/NCBI

103 

Fu Y, Cui S, Zhou Y and Qiu L: Dental pulp stem cell-derived exosomes alleviate mice knee osteoarthritis by inhibiting TRPV4-mediated osteoclast activation. Int J Mol Sci. 24:48262023.

104 

Meng C, Na Y, Han C, Ren Y, Liu M, Ma P and Bai R: Exosomal miR-429 derived from adipose-derived stem cells ameliorated chondral injury in osteoarthritis via autophagy by targeting FEZ2. Int Immunopharmacol. 120:1103152023. View Article : Google Scholar : PubMed/NCBI

105 

Yang H, Zhou Y, Ying B, Dong X, Qian Q and Gao S: Effects of human umbilical cord mesenchymal stem cell-derived exosomes in the rat osteoarthritis models. Stem Cells Transl Med. 13:803–811. 2024. View Article : Google Scholar : PubMed/NCBI

106 

Huang C, Zhao Y, Lin S, Li L, Guo X, Yumiseba S, Yang JD, Hariri R, Ye Q, He S and Kilcoyne A: Characterization of human placenta-derived exosome (pExo) as a potential osteoarthritis disease modifying therapeutic. Arthritis Res Ther. 25:2292023. View Article : Google Scholar : PubMed/NCBI

107 

Zhao S, Xiu G, Wang J, Wen Y, Lu J, Wu B, Wang G, Yang D, Ling B, Du D and Xu J: Engineering exosomes derived from subcutaneous fat MSCs specially promote cartilage repair as miR-199a-3p delivery vehicles in osteoarthritis. J Nanobiotechnology. 21:3412023. View Article : Google Scholar : PubMed/NCBI

108 

Yang L, Li W, Zhao Y and Shang L: Magnetic polysaccharide mesenchymal stem cells exosomes delivery microcarriers for synergistic therapy of osteoarthritis. ACS Nano. Jul 22–2024.Epub ahead of print.

109 

Ma T, Xu G, Gao T, Zhao G, Huang G, Shi J, Chen J, Song J, Xia J and Ma X: Engineered exosomes with ATF5-modified mRNA loaded in injectable thermogels alleviate osteoarthritis by targeting the mitochondrial unfolded protein response. ACS Appl Mater Interfaces. 16:21383–21399. 2024. View Article : Google Scholar : PubMed/NCBI

110 

Ju Y, Hu Y, Yang P, Xie X and Fang B: Extracellular vesicle-loaded hydrogels for tissue repair and regeneration. Mater Today Bio. 18:1005222022. View Article : Google Scholar

111 

Chen M, Lu Y, Liu Y, Liu Q, Deng S, Liu Y, Cui X, Liang J, Zhang X, Fan Y and Wang Q: Injectable microgels with hybrid exosomes of chondrocyte-targeted FGF18 gene-editing and self-renewable lubrication for osteoarthritis therapy. Adv Mater. 36:e23125592024. View Article : Google Scholar : PubMed/NCBI

112 

Wen S, Huang X, Ma J, Zhao G, Ma T, Chen K, Huang G, Chen J, Shi J and Wang S: Exosomes derived from MSC as drug system in osteoarthritis therapy. Front Bioeng Biotechnol. 12:13312182024. View Article : Google Scholar : PubMed/NCBI

113 

Lai JJ, Chau ZL, Chen SY, Hill JJ, Korpany KV, Liang NW, Lin LH, Lin YH, Liu JK, Liu YC, et al: Exosome processing and characterization approaches for research and technology development. Adv Sci (Weinh). 9:e21032222022. View Article : Google Scholar : PubMed/NCBI

114 

Kimiz-Gebologlu I and Oncel SS: Exosomes: Large-scale production, isolation, drug loading efficiency, and biodistribution and uptake. J Control Release. 347:533–543. 2022. View Article : Google Scholar : PubMed/NCBI

115 

Yadav A, Xuan Y, Sen CK and Ghatak S: Standardized reporting of research on exosomes to ensure rigor and reproducibility. Adv Wound Care (New Rochelle). 13:584–599. 2024. View Article : Google Scholar : PubMed/NCBI

116 

Kilgore RE, Moore BD, Sripada SA, Chu W, Shastry S, Barbieri E, Hu S, Tian W, Petersen H, Mohammadifar M, et al: Peptide ligands for the universal purification of exosomes by affinity chromatography. Biotechnol Bioeng. 121:3484–3501. 2024. View Article : Google Scholar : PubMed/NCBI

117 

Hassanpour Tamrin S, Sanati Nezhad A and Sen A: Label-free isolation of exosomes using microfluidic technologies. ACS Nano. 15:17047–17079. 2021. View Article : Google Scholar : PubMed/NCBI

118 

Jiang Y, Lv H, Shen F, Fan L, Zhang H, Huang Y, Liu J, Wang D, Pan H and Yang J: Strategies in product engineering of mesenchymal stem cell-derived exosomes: Unveiling the mechanisms underpinning the promotive effects of mesenchymal stem cell-derived exosomes. Front Bioeng Biotechnol. 12:13637802024. View Article : Google Scholar : PubMed/NCBI

119 

Shao J, Zaro J and Shen Y: Advances in exosome-based drug delivery and tumor targeting: From tissue distribution to intra-cellular fate. Int J Nanomedicine. 15:9355–9371. 2020. View Article : Google Scholar :

120 

Fan X, Zhang Y, Liu W, Shao M, Gong Y, Wang T, Xue S and Nian R: A comprehensive review of engineered exosomes from the preparation strategy to therapeutic applications. Biomater Sci. 12:3500–3521. 2024. View Article : Google Scholar : PubMed/NCBI

121 

He J, Ren W, Wang W, Han W, Jiang L, Zhang D and Guo M: Exosomal targeting and its potential clinical application. Drug Deliv Transl Res. 12:2385–2402. 2022. View Article : Google Scholar : PubMed/NCBI

122 

Barzin M, Bagheri AM, Ohadi M, Abhaji AM, Salarpour S and Dehghannoudeh G: Application of plant-derived exosome-like nanoparticles in drug delivery. Pharm Dev Technol. 28:383–402. 2023. View Article : Google Scholar : PubMed/NCBI

123 

Liang Y, Duan L, Lu J and Xia J: Engineering exosomes for targeted drug delivery. Theranostics. 11:3183–3195. 2021. View Article : Google Scholar : PubMed/NCBI

124 

Luo R, Liu M, Tan T, Yang Q, Wang Y, Men L, Zhao L, Zhang H, Wang S, Xie T and Tian Q: Emerging significance and therapeutic potential of extracellular vesicles. Int J Biol Sci. 17:2476–2486. 2021. View Article : Google Scholar : PubMed/NCBI

125 

Tiruvayipati S, Wolfgeher D, Yue M, Duan F, Andrade J, Jiang H and Schuger L: Variability in protein cargo detection in technical and biological replicates of exosome-enriched extracellular vesicles. PLoS One. 15:e02288712020. View Article : Google Scholar : PubMed/NCBI

126 

Ranjan P and Verma SK: Exosomes isolation, purification, and characterization. Methods Mol Biol. 2835:173–180. 2024. View Article : Google Scholar : PubMed/NCBI

127 

Gandham S, Su X, Wood J, Nocera AL, Alli SC, Milane L, Zimmerman A, Amiji M and Ivanov AR: Technologies and standardization in research on extracellular vesicles. Trends Biotechnol. 38:1066–1098. 2020. View Article : Google Scholar : PubMed/NCBI

128 

Singh S, Dansby C, Agarwal D, Bhat PD, Dubey PK and Krishnamurthy P: Exosomes: Methods for isolation and characterization in biological samples. Methods Mol Biol. 2835:181–213. 2024. View Article : Google Scholar : PubMed/NCBI

129 

Ansari FJ, Tafti HA, Amanzadeh A, Rabbani S, Shokrgozar MA, Heidari R, Behroozi J, Eyni H, Uversky VN and Ghanbari H: Comparison of the efficiency of ultrafiltration, precipitation, and ultracentrifugation methods for exosome isolation. Biochem Biophys Rep. 38:1016682024.PubMed/NCBI

130 

Claridge B, Lozano J, Poh QH and Greening DW: Development of extracellular vesicle therapeutics: Challenges, considerations, and opportunities. Front Cell Dev Biol. 9:7347202021. View Article : Google Scholar : PubMed/NCBI

131 

Lin X and Zhu J, Shen J, Zhang Y and Zhu J: Advances in exosome plasmonic sensing: Device integration strategies and AI-aided diagnosis. Biosens Bioelectron. 266:1167182024. View Article : Google Scholar : PubMed/NCBI

132 

Wang X, Xia J, Yang L, Dai J and He L: Recent progress in exosome research: Isolation, characterization and clinical applications. Cancer Gene Ther. 30:1051–1065. 2023. View Article : Google Scholar : PubMed/NCBI

133 

Rezaie J, Ajezi S, Avci ÇB, Karimipour M, Geranmayeh MH, Nourazarian A, Sokullu E, Rezabakhsh A and Rahbarghazi R: Exosomes and their application in biomedical field: Difficulties and advantages. Mol Neurobiol. 55:3372–3393. 2018. View Article : Google Scholar

134 

Saravanan PB, Kalivarathan J, Khan F, Shah R, Levy MF and Kanak MA: Exosomes in transplantation: Role in allograft rejection, diagnostic biomarker, and therapeutic potential. Life Sci. 324:1217222023. View Article : Google Scholar : PubMed/NCBI

135 

Awadasseid A, Wu Y and Zhang W: Extracellular vesicles (exosomes) as immunosuppressive mediating variables in tumor and chronic inflammatory microenvironments. Cells. 10:25332021. View Article : Google Scholar : PubMed/NCBI

136 

Codispoti B, Marrelli M, Paduano F and Tatullo M: NANOmetric BIO-Banked MSC-derived exosome (NANOBIOME) as a novel approach to regenerative medicine. J Clin Med. 7:3572018. View Article : Google Scholar : PubMed/NCBI

137 

Krut Z, Pelled G, Gazit D and Gazit Z: Stem cells and exosomes: New therapies for intervertebral disc degeneration. Cells. 10:22412021. View Article : Google Scholar : PubMed/NCBI

138 

Hartman N, Loyal J and Fabi S: Update on exosomes in aesthetics. Dermatol Surg. 48:862–865. 2022. View Article : Google Scholar : PubMed/NCBI

139 

Syed NH, Misbah I, Azlan M, Ahmad Mohd Zain MR and Nurul AA: Exosomes in osteoarthritis: A review on their isolation techniques and therapeutic potential. Indian J Orthop. 58:866–875. 2024. View Article : Google Scholar : PubMed/NCBI

140 

Ma CY, Zhai Y, Li CT, Liu J, Xu X, Chen H, Tse HF and Lian Q: Translating mesenchymal stem cell and their exosome research into GMP compliant advanced therapy products: Promises, problems and prospects. Med Res Rev. 44:919–938. 2024. View Article : Google Scholar

141 

Xia Y, Zhang J, Liu G and Wolfram J: Immunogenicity of extracellular vesicles. Adv Mater. 36:e24031992024. View Article : Google Scholar : PubMed/NCBI

142 

Huang J, Xiong J, Yang L, Zhang J, Sun S and Liang Y: Cell-free exosome-laden scaffolds for tissue repair. Nanoscale. 13:8740–8750. 2021. View Article : Google Scholar : PubMed/NCBI

143 

Mi P, Liu JL, Qi BP, Wei BM, Xu CZ and Zhu L: Stem cell-derived exosomes for chronic wound repair. Cell Tissue Res. 391:419–423. 2023. View Article : Google Scholar : PubMed/NCBI

144 

Yadav A, Nandy A, Sharma A and Ghatak S: Exosome mediated cell-cell crosstalk in tissue injury and repair. Results Probl Cell Differ. 73:249–297. 2024. View Article : Google Scholar : PubMed/NCBI

145 

Tan F, Li X, Wang Z, Li J, Shahzad K and Zheng J: Clinical applications of stem cell-derived exosomes. Signal Transduct Target Ther. 9:172024. View Article : Google Scholar : PubMed/NCBI

146 

Tao SC, Yuan T, Zhang YL, Yin WJ, Guo SC and Zhang CQ: Exosomes derived from miR-140-5p-overexpressing human synovial mesenchymal stem cells enhance cartilage tissue regeneration and prevent osteoarthritis of the knee in a rat model. Theranostics. 7:180–195. 2017. View Article : Google Scholar : PubMed/NCBI

147 

Sun H, Hu S and Zhang Z, Lun J, Liao W and Zhang Z: Expression of exosomal microRNAs during chondrogenic differentiation of human bone mesenchymal stem cells. J Cell Biochem. 120:171–181. 2019. View Article : Google Scholar

148 

Lu L, Wang J, Fan A, Wang P, Chen R, Lu L and Yin F: Synovial mesenchymal stem cell-derived extracellular vesicles containing microRN555A-26a-5p ameliorate cartilage damage of osteoarthritis. J Gene Med. 23:e33792021. View Article : Google Scholar : PubMed/NCBI

149 

Li Y, Tu Q, Xie D, Chen S, Gao K, Xu X, Zhang Z and Mei X: Triamcinolone acetonide-loaded nanoparticles encapsulated by CD90+ MCSs-derived microvesicles drive anti-inflammatory properties and promote cartilage regeneration after osteoarthritis. J Nanobiotechnology. 20:1502022. View Article : Google Scholar

150 

Zhou X, Cao H, Guo J, Yuan Y and Ni G: Effects of BMSC-derived EVs on bone metabolism. Pharmaceutics. 14:10122022. View Article : Google Scholar : PubMed/NCBI

151 

Zhang Y, Qi G, Yan Y, Wang C, Wang Z, Jiang C, Jiang Z, Ma T, Zhang C and Yan Z: Exosomes derived from bone marrow mesenchymal stem cells pretreated with decellularized extracellular matrix enhance the alleviation of osteoarthritis through miR-3473b/phosphatase and tensin homolog axis. J Gene Med. 25:e35102023. View Article : Google Scholar : PubMed/NCBI

152 

Lai C, Liao B, Peng S, Fang P, Bao N and Zhang L: Synovial fibroblast-miR-214-3p-derived exosomes inhibit inflammation and degeneration of cartilage tissues of osteoarthritis rats. Mol Cell Biochem. 478:637–649. 2023. View Article : Google Scholar :

153 

Ji Y, Xiong L, Zhang G, Xu M, Qiu W, Xiu C, Kuang G and Rui Y: Synovial fluid exosome-derived miR-182-5p alleviates osteoarthritis by downregulating TNFAIP8 and promoting autophagy through LC3 signaling. Int Immunopharmacol. 125:1111772023. View Article : Google Scholar : PubMed/NCBI

154 

Wu S, Luo J, Zhang X, Wang L, Cai L and Xu J: Synovia tissue-specific exosomes participate in the dual variation of the osteoarthritis microenvironment via miR-182. Exp Cell Res. 436:1139812024. View Article : Google Scholar : PubMed/NCBI

155 

Qiu M, Xie Y, Tan G, Wang X, Huang P and Hong L: Synovial mesenchymal stem cell-derived exosomal miR-485-3p relieves cartilage damage in osteoarthritis by targeting the NRP1-mediated PI3K/Akt pathway: Exosomal miR-485-3p relieves cartilage damage. Heliyon. 10:e240422024. View Article : Google Scholar : PubMed/NCBI

156 

Zhang Y, Wang X, Chen J, Qian D, Gao P, Qin T, Jiang T, Yi J, Xu T, Huang Y, et al: Exosomes derived from platelet-rich plasma administration in site mediate cartilage protection in subtalar osteoarthritis. J Nanobiotechnology. 20:562022. View Article : Google Scholar : PubMed/NCBI

157 

Lou C, Jiang H, Lin Z, Xia T, Wang W, Lin C, Zhang Z, Fu H, Iqbal S, Liu H, et al: MiR-146b-5p enriched bioinspired exosomes derived from fucoidan-directed induction mesenchymal stem cells protect chondrocytes in osteoarthritis by targeting TRAF6. J Nanobiotechnology. 21:4862023. View Article : Google Scholar : PubMed/NCBI

158 

Meng S, Tang C, Deng M, Yuan J, Fan Y, Gao S, Feng Y, Yang J and Chen C: Tropoelastin-pretreated exosomes from adipose-derived stem cells improve the synthesis of cartilage matrix and alleviate osteoarthritis. J Funct Biomater. 14:2032023. View Article : Google Scholar : PubMed/NCBI

159 

Wang S, Jiang W, Lv S, Sun Z, Si L, Hu J, Yang Y, Qiu D, Liu X, Zhu S, et al: Human umbilical cord mesenchymal stem cells-derived exosomes exert anti-inflammatory effects on osteoarthritis chondrocytes. Aging (Albany NY). 15:9544–9560. 2023. View Article : Google Scholar : PubMed/NCBI

160 

Chen J, Ni X, Yang J, Yang H, Liu X, Chen M, Sun C and Wang Y: Cartilage stem/progenitor cells-derived exosomes facilitate knee cartilage repair in a subacute osteoarthritis rat model. J Cell Mol Med. 28:e183272024. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2025
Volume 55 Issue 3

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen X, Tian B, Wang Y, Zheng J and Kang X: Potential and challenges of utilizing exosomes in osteoarthritis therapy (Review). Int J Mol Med 55: 43, 2025.
APA
Chen, X., Tian, B., Wang, Y., Zheng, J., & Kang, X. (2025). Potential and challenges of utilizing exosomes in osteoarthritis therapy (Review). International Journal of Molecular Medicine, 55, 43. https://doi.org/10.3892/ijmm.2025.5484
MLA
Chen, X., Tian, B., Wang, Y., Zheng, J., Kang, X."Potential and challenges of utilizing exosomes in osteoarthritis therapy (Review)". International Journal of Molecular Medicine 55.3 (2025): 43.
Chicago
Chen, X., Tian, B., Wang, Y., Zheng, J., Kang, X."Potential and challenges of utilizing exosomes in osteoarthritis therapy (Review)". International Journal of Molecular Medicine 55, no. 3 (2025): 43. https://doi.org/10.3892/ijmm.2025.5484