Back to Journals » International Journal of Nanomedicine » Volume 20
Nanomaterial-Based Drug Delivery Systems Targeting Functional Cells for Osteoarthritis Treatment: Mechanisms, Challenges and Future Prospects
Authors Kong H , He Q, Han J , Zhang XA
Received 22 January 2025
Accepted for publication 11 April 2025
Published 25 April 2025 Volume 2025:20 Pages 5291—5320
DOI https://doi.org/10.2147/IJN.S518935
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Dr Kamakhya Misra
Hui Kong,1,2 Qijun He,2 Juanjuan Han,2 Xin-An Zhang1,2
1College of Exercise and Health, Shanghai University of Sport, Shanghai, People’s Republic of China; 2College of Exercise and Health, Shenyang Sport University, Shenyang, People’s Republic of China
Correspondence: Xin-An Zhang, Shanghai University of Sport, 200 heng’Ren Road, Yangpu District, Shanghai, 200438, People’s Republic of China, Email [email protected]
Abstract: Osteoarthritis (OA) represents a chronic joint disease characterized by articular cartilage degeneration, synovial inflammation, and subchondral bone erosions. Functional cells in OA mainly include macrophages, synoviocytes, chondrocytes, and mesenchymal stem cells. These cells can secrete cytokines and non-coding RNAs and exosomes and interact with each other to coregulate the progression of OA. Some nanomaterial-based drug delivery systems (DDSs) surface ligands can alleviate OA by targeting receptors on the surface of functional cells. Meanwhile, other nanomaterial-based DDSs, whose surfaces are masked by the cell membranes or extracellular vesicles of these functional cells, treat OA by targeting and attacking the diseased site. When ligand-modified nanomaterials target specific functional cells to treat OA, the functional cells are attacked. Functional cells become attackers, similar to arrows, when their cell membranes or extracellular vesicles are modified into nanomaterials to deliver drugs for OA treatment. An increasing number of studies have been conducted on nanomaterial-based DDS-targeted functional cells for the treatment of OA, but none has summarized the corresponding research progress and mechanism of action. In this review, the related references on the treatment of osteoarthritis with nanomaterial-based DDSs targeting functional cells have been included, and how a variety of functional cells can be engineered into nanomaterial-based DDSs serving as targets or arrows to treat OA has been summarised for the first time, providing a new idea and method for the targeted treatment of OA.
Keywords: nanomaterial-based drug delivery systems, osteoarthritis, macrophages, chondrocytes, synoviocytes, mesenchymal stem cells
Graphical Abstract:
Introduction
Osteoarthritis (OA) is the most common degenerative joint disease, and its pathologic changes include synovial inflammation, cartilage destruction, subchondral bone sclerosis, and so on.1 The major risk factors for OA include age, gender, joint trauma, and genetic factors,2 and its pathogenesis remains unclear. Functional cells, such as macrophages, synoviocytes, chondrocytes, and mesenchymal stem cells (MSCs), play an imperative role in the pathogenesis of OA.3–5 These cells can secrete cytokines, growth factors, noncoding RNAs and exosomes, and interfere with each other to collectively regulate the progression of OA.6,7 Given the complexity of OA and the limitations of therapeutic approaches, providing effective treatments to patients with OA remains a challenge.
The treatment methods for OA mainly comprise medications, surgery, exercise therapy, and lifestyle modification.8 Pharmacological treatment remains the most commonly used clinical tool for OA, and its administration includes systemic administration and intra-articular local injection.9 Nonsteroidal antiinflammatory drugs (NSAIDs) and analgesics are commonly administered orally and can provide some relief from symptoms and pain. However, the long-term use of these drugs can cause systemic adverse effects and gastrointestinal damage.10 By contrast, local intra-articular drug delivery can inject drugs directly into the lesion site, which increases the initial drug concentration in the joint cavity.11 However, drugs injected in this manner are readily phagocytosed by macrophages or cleared by the lymphatic blood system, and the amount of drug that is actually transported to the inflammation site and penetrates the cartilage is low.
To improve the effectiveness of drug delivery for OA treatment, scholars have adopted nanomaterial-based drug delivery systems (DDSs), including nanoparticles (NPs), hydrogels, liposomes, etc. These systems effectively improve the retention time of delivered drugs in the joints and demonstrates good biocompatibility.12,13 The DDS for OA has achieved abundant research results, and various types of nanomaterials have been developed to meet drug delivery requirements.14 Given the degenerative cartilage tissue damage and uneven distribution of inflammation caused by OA, targeted drug delivery to inflamed tissues or lesions can increase drug efficacy and considerably reduce off-target toxicity.15 Some nanomaterial-based DDSs can target and modulate functional cells through ligand modification to alleviate OA,16,17 and other nanomaterial-based DDSs, whose surfaces are masked by the cell membranes or extracellular vesicles (EVs) of these functional cells, treat OA by targeting and attacking the lesion site.18,19 When ligand-modified nanomaterials target specific functional cells to treat OA, the functional cells are attacked. These cells become attackers, similar to arrows, when their cell membranes or EVs are modified into nanomaterials to deliver drugs for OA treatment.
The research on nanomaterial-based DDS-targeted functional cell therapy for OA has been increasing yearly, but no one has generalized and summarized the research progress. This review focuses on the role of functional cells in the treatment of OA with engineered nanomaterial-based DDSs and discusses the prospects and challenges of these nanomaterial-based DDSs associated with OA functional cells in the treatment of OA, which provides a new idea for the development of nanomaterial-based DDSs targeting OA functional cells.
Pathophysiology and Therapeutic Targets of OA
Pathophysiology of OA
OA refers to a common chronic degenerative joint disease with a complex pathophysiologic mechanism closely related to synovial inflammation, cartilage degeneration, subchondral bone sclerosis, bone redundancy formation, and mechanical stress imbalance of joint structures.20
An increasing number of studies have revealed that the pathogenesis of OA may initiate with synovial inflammation. The activation of immune cells and release of inflammatory mediators can be observed in the synovium of patients with OA.21 Activation of immune cells, such as macrophages and T cells, causes their secretion of inflammatory factors, such as interleukin-1 (IL-1) and tumor necrosis factor-alpha (TNF-α). These inflammatory factors stimulate synovial cell proliferation and promote synovial neovascularization, which result in an increased synovial inflammation.22 Synovial inflammation leads to increased synovial exudate and fluid accumulation in the joint cavity, which leads to joint swelling and pain. In addition, synovitis results in the release of more degradative enzymes and inflammatory factors, which further damage articular cartilage and bone tissue.23
Articular cartilage comprises chondrocytes and matrix. Its main components include collagen fibers and proteoglycans, which help maintain the elasticity of cartilage and cushion pressure.24 In the pathological process of OA, synovitis-derived inflammatory factors lead to chondrocyte dysfunction and the overexpressions of matrix metalloproteinases (MMPs).25 In addition, the abnormal perception of mechanical stress by chondrocytes can lead to an imbalance in intracellular signaling pathways and the overexpressions of inflammatory factors and catabolic enzymes. This condition leads to the degradation of collagen and proteoglycan in the cartilage and ultimately destroys the structural integrity of the cartilage.26
Damaged to the articular cartilage results in a high stress in the subchondral bone. Accordingly, the subchondral bone remodels with a high bone density.27 This remodeling can cause the microfractures of the subchondral bone to stimulate the differentiation of MSCs into osteoblasts and promote osteophyte formation.28 Osteophyte is an essential pathological feature of OA, and its formation is associated with several factors. On the one hand, mechanical stress and inflammatory factors can stimulate the activity of osteoblasts and promote bone formation. On the other hand, growth factors derived from chondrocytes and synoviocytes, such as transforming growth factor-beta (TGF-β), can promote the formation of osteophyte.29 These changes not only influence the mechanical stability of joints but also further aggravate the degradation of cartilage through interaction with the cartilage.
In addition, epigenetic regulatory mechanisms, such as DNA methylation, histone modification, and noncoding RNA regulation, affect the progression of OA.30,31 Therefore, the pathological process of OA results from the interaction of many factors, involving cartilage degeneration, synovitis, subchondral sclerosis, osteophyte formation, and so on. The in-depth study of the pathological mechanism of OA will provide a theoretical basis for the development of targeted treatment strategies.
Therapeutic Targets for OA
Currently, targeted therapeutic strategies for OA are evolving towards precision regulation to design specific OA interventions. Early studies focused on inhibiting single molecules or pathways, but the heterogeneity of OA and the complexity of the multicellular microenvironment make the search for regulatory targets at multiple levels a key issue to be addressed in OA therapy. Targeting functional cells, cytokines, enzymes and signalling pathways is an important mechanism for precision treatment of OA (Figure 1).
Functional Cells
Targeting of functional cells aims to regulate the pathological process in joints by targeting specific cell types. Chondrocytes, synoviocytes, macrophages, and MSCs are the most commonly targeted cells in OA treatment. Chondrocytes are the main constituents of articular cartilage, and their main function is the synthesis of the cartilage matrix.32 Targeting of chondrocytes can inhibit the expression of matrix-degrading enzymes, promote the regeneration of cartilage matrix, and restore joint function.33 Synoviocytes contribute to the regulation of the pathological process of synovitis. Targeting of synoviocytes can inhibit inflammation and reduce joint swelling and pain. Macrophages aggravate local inflammation and tissue destruction by releasing pro-inflammatory cytokines in OA. Targeting macrophages can regulate immune response and reduce inflammation level.34 MSCs are pluripotent stem cells possessing self-renewal and multidirectional differentiation abilities. They can differentiate into osteoblasts, chondrocytes, and adipocytes and participate in bone and cartilage repair. Targeted MSCs can secrete a variety of growth factors and cytokines, inhibit inflammation, and promote cartilage repair.35
Many technologies, such as gene therapy, small-molecule drugs, and nanomaterial-based DDSs, have been developed to target functional cells to treat OA. Introducing therapeutic genes into specific functional cells can regulate their biological behavior and repair damaged cartilage. The introduction of anti-inflammatory genes into synoviocytes or macrophages with viral or nonviral vectors can inhibit their inflammation.36 In addition, small-molecule drugs and biologics inhibit the release of inflammatory factors or the overexpressions of enzymes by binding to cell-surface receptors or specific intracellular signaling pathways.37 More importantly, given their small size and modifiability, nanomaterial-based DDSs can deliver drugs or genes to functional cells. Using specific surface modifications (eg, antibodies or ligands), nanomaterial-based DDSs can selectively target cells at the site of OA lesions to enhance local therapeutic effects.38
Targeted functional cell therapy for OA shows great promise. Compared with traditional treatment methods, functional cell therapy has the advantages of strong targeting, remarkable efficacy, and limited side effects. With the continuous progress of technology, the isolation, culture, and expansion technology of functional cells will be improved, the gene modification technology will be safer and more effective, and the combined treatment program will become more diversified. The development of these technologies will provide more effective means for the treatment of OA. However, a strategy for achieving efficient cell targeting and effective drug delivery remains a major challenge. Further clinical trials are needed to validate the safety, efficacy, and long-term effectiveness of these approaches.
Cytokines
Targeted cytokine therapy for OA is a therapeutic strategy for the targeted modulation of immune responses and inflammatory mediators. Cytokines play a key role in the pathogenesis of OA. These substances are small-molecule proteins secreted by cells and are involved in inflammatory responses, immune regulation, and tissue repair.39 Given that OA is a degenerative joint disease associated with chronic inflammation, targeting and inhibiting pro-inflammatory cytokines or enhancing anti-inflammatory cytokine activity has become an essential means for OA treatment.40
During the pathological process of OA, cells in the articular cartilage, synovium, bone, and other joint structures produce large amounts of pro-inflammatory cytokines, such as TNF-α, IL-1β, and IL-6, due to abnormal stress or injury. These cytokines promote cartilage matrix degradation, synovial inflammation, and bone destruction, which lead to increased damage to joint structures.41 Meanwhile, anti-inflammatory cytokines, such as IL-10 and IL-4, have an inhibitory effect on inflammation and can reduce tissue damage and promote repair.42 The principle of targeted cytokine therapy is the inhibition of the production and activity of pro-inflammatory factors and promoted production or activity of anti-inflammatory cytokines. On the one hand, the use of inhibitors, blocking agents, or monoclonal antibodies against TNF-α, IL-1, and IL-6 can inhibit their activity and reduce inflammatory responses and cartilage degradation.43 On the other hand, cartilage degeneration can be alleviated by exogenous administration of factors, such as IL-10 or IL-4.44 An important challenge in this type of therapy is the effective delivery of anti-inflammatory cytokines to the site of arthropathy and the maintenance of their local activity.
The research on targeted cytokine therapy for OA remains in its infancy. Although numerous drugs targeting pro-inflammatory or anti-inflammatory cytokines exhibit a great potential in the treatment of OA, their efficacy and safety in clinical application still need further verification. In addition, how to combine nanomaterial-based DDSs to deliver these bioactive molecules accurately to the lesion area and enhance their stability and durability is an important research direction in the future.
Signaling Pathways and Enzymes
Targeting of OA-related pathways or enzymes is an effective therapeutic strategy. The occurrence and development of OA involve not only a number of complex signaling pathways, such as Wnt/β-catenin, nuclear factor-kappa B (NF-κB), janus kinase (JAK), and transforming growth factor beta (TGF-β),45,46 but also cartilage matrix-degrading enzymes and inflammatory mediators, including MMPs, phospholipase A2, a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS), and cyclooxygenase-2 (COX-2).47 We can use small-molecule inhibitors, monoclonal antibodies, gene therapy, and other methods to target OA-related pathways or enzymes. The JAK inhibitor tofacitinib regulates the JAK1/TNF-α pathway through Mir-149-5p, which reduces chondrocyte hypertrophy and inhibits OA inflammation.48 Tissue inhibitor of metalloproteinase-1, an inhibitor of MMPs, may inhibit cartilage degradation and alleviate OA.49 Rofecoxib, an inhibitor of COX-2, has been widely used in clinical treatment and has displayed good effects of inflammation inhibition and pain relief.50 In addition, gene editing technology may play a crucial role. For example, RNA interference or CRISPR-Cas9 technology can be used to selectively knock out or inhibit the FGF18 gene, regulate related signaling pathways and alleviate OA.51
Targeting signal pathway or enzyme therapy for OA is an innovative treatment with great potential. The pathological process of OA can be delayed by interfering with key pathological signal pathways or enzymes. Although therapeutic strategies targeting signal pathways or enzymes have important effects on basic research, they face certain challenges in clinics. The disease phenotype of patients may be heterogeneous, and targeting a single signal pathway or enzyme may not completely prevent the progress of OA.24 In addition, long-term inhibition of some signal pathways may cause some side effects, such as immunosuppression and cartilage calcification.52 Therefore, in the future, the dynamic regulation of each signal pathway and its role at different stages of the disease must be deeply studied to develop more accurate and individualized treatment programs.
The Current Treatment Options for OA
Currently, the main treatment options for OA are aimed at relieving symptoms and slowing disease progression. Although traditional therapies are more commonly used in the clinic, the different treatments have some limitations that limit improvements in efficacy.53 Traditional pharmacological treatments focus on NSAIDs, which reduce pain and inflammation by inhibiting cyclooxygenase (COX) activity, but long-term use can cause gastrointestinal damage and even cardiovascular risks.54 To avoid these adverse effects, treatment options for OA have gradually shifted to local drug delivery. For example, intra-articular injections of glucocorticoids rapidly suppress synovial inflammatory exudates and their short-term efficacy is evident, but repeated injections may accelerate joint degeneration.55 Chondroprotective agents such as chondroitin sulphate and hyaluronic acid (HA) may slow disease progression by improving joint lubrication, providing a practical basis for the therapeutic concept of targeting the cartilage microenvironment.56
In addition, non-pharmacological interventions play an important role in the treatment of OA. Exercise rehabilitation improves joint stability by increasing muscle strength, and weight management directly reduces mechanical loading, and the two work synergistically to slow the progression of early OA.57,58 However, patient compliance can be poor and the effectiveness of this approach is not clear in patients with moderate to advanced OA. When conservative treatment fails, surgery is the last option. However, arthroscopic debridement does not promote cartilage regeneration despite short-term removal of inflamed synovium and debris.59 While arthroplasty significantly restores knee function, it is associated with the risk of infection and prosthesis longevity.60
All of these traditional treatments have limitations. Currently, achieving precise drug delivery and long-lasting modulation is a key issue that needs to be urgently addressed. For example, systemic drug delivery results in poor drug utilisation due to poor joint targeting, while local injection has the problem of rapid clearance and repeated manipulation.61 In addition, the dynamic complexity of the inflammatory microenvironment makes it difficult to block OA progression by single-pathway inhibition, and multi-target synergistic intervention is required.62 In recent years, emerging biologics such as anti-cytokine antibodies and gene therapy have the potential for targeted intervention, but they are costly and have complex technical requirements.63 Therefore, the rapid development of nanomaterial-based drug delivery systems is expected to solve the problems of existing therapies and provide a new approach for precise and personalised treatment of OA by precisely regulating drug release, improving lesion targeting and reducing systemic toxicity.64
Research Progress on Nanomaterial-Based DDSs in OA
Nanomaterial-based DDSs are widely used in the treatment of OA. Nanotechnology has great potential in the treatment of OA, mainly in terms of drug targeting, bioavailability, and duration of action.65 Compared with traditional drug delivery methods, nanomaterial-based DDSs can deliver drugs to the lesion site more accurately, increase the effective concentration of drugs, and control the drug release time.66 The lesion site of OA is mainly in the joints, which involve synoviocytes, macrophages, and chondrocytes.67 Traditional drugs are widely distributed throughout the body, and their accurate delivery to these diseased cells presents difficulty. Nanomaterial-based DDSs have unique size and surface charge, which can accumulate in diseased joints. In addition, ligands on the surface of nanomaterial-based DDSs can bind to receptors on the surface of diseased cells and play an active targeting role. Loading ligands that can specifically recognize and bind to joint tissues or cells on nanomaterials has become the main means to achieving the active targeted delivery of nanomaterial-based DDSs.68,69 Many biological targeting ligands, such as peptides, nucleic acids, small-molecular compounds and macromolecular compounds, show good stability, high affinity and easy modification.
Polypeptides have low immunogenicity, high affinity and easy biological coupling. Therefore, polypeptide ligands are being increasingly applied in the field of nanotherapy.70 Collagen is the main component of cartilage matrix, with type II collagen accounting for 90–95% of the collagen network. Therefore, targeting ligands for type II collagen can considerably enhance the close combination between nanocarriers and cartilage. Rothefuh et al71 screened the isolated bovine cartilage through phage display technology, and WYRGRL peptide showed a strong specificity for type II collagen binding. WYRGRL peptides were modified on the surface of nanomaterials through covalent bonds. The aggregation of these nanomaterials containing targeted peptides in the extracellular matrix was substantially higher than that in nanomaterials with interfering peptides. In addition, as an important component of cartilage tissue, chondrocytes are located in the deep layer of cartilage tissues, and their main function is the production and maintenance of the extracellular matrix. If the specific targeting of chondrocytes can be achieved, nanomaterial-based DDSs can penetrate the cartilage matrix and directly reach its deep tissues for effective treatment. Pi et al72 identified a chondrocyte affinity peptide (CAP) with the sequence of DWRVIIPPRPSA using phage display technology. The affinity peptide has a high specificity for chondrocytes and shows no species difference. Zhao et al73 modified the copolymer compound with CAP to realize miRNA delivery. The results showed that CAP modification can enhance the ability of chondrocytes to absorb polymers and attains good targeting efficiency and cartilage permeability.
Nucleic acid aptamers consist of single-stranded DNA or RNA. They are synthesized artificially by virtue of strict recognition ability and affinity between nucleotides. These small-molecule biomolecules possess a unique tertiary structure and can specifically bind to various targeting targets, such as peptides, small-molecular particles or whole cells.74,75 Given the easy synthesis and modification of aptamers and their high affinity and specificity for target molecules, nanomaterials modified by aptamers are widely applied in the targeted therapy of OA. Chen et al76 screened a nucleic acid aptamer CX3, which can specifically bind to fibroblast-like synovial cells. On this basis, CX3-modified liposomes were further constructed, and targeted delivery of dasatinib was used to remove aging synovial cells at the OA site. In vitro cell co-incubation experiments revealed that CX3-modified liposomes have a high specificity for fibroblast-like synovial cells, and they can effectively promote the delivery of nanocarriers to synovial cells. However, the development cost of nucleic acid aptamers is high, and their structural stability is insufficient. Therefore, the related application research remains in the basic stage, and only a few materials have entered clinical studies.
In addition, small-molecular and macromolecular compounds are common biological targeting ligands. Chondroitin sulfate (ChS) is a natural anionic glycosaminoglycan and is also a chronic drug for the treatment of OA. This compound can inhibit the apoptosis of damaged cartilage tissue and repair damaged cartilage.77 Chitosan (CS), as a small-molecule ligand, can bind to type II collagen, cluster of differentiation 36 (CD36), and other receptors on chondrocytes to achieve targeted localization.78,79 Bishnoi et al80 used CS-coupled liposomes to prepare conjugated polymer CS solid-lipid NPs (SLN). After intravenous administration, the experimental group of CS SLN exhibited a considerably higher absorption rate of drugs at the articular cartilage compared with that in the liposome group of unmodified CS. This finding confirmed that CS can target the articular cartilage. The surface of activated M1 macrophages overexpresses the receptors of folic acid (FA) and cluster of differentiation 44 (CD44), which display low expressions in normal cells. Therefore, FA and CD44 can be modified onto the surface of nanomaterials as their corresponding specific macromolecular ligands to construct DDSs that can specifically target and activate macrophages.81,82 Yang et al83 used FA-modified HA to wrap carbon monoxide-releasing molecules to construct a nanogel. The FA-modified HA nanogel was marked with CY 5.5, and a strong fluorescence signal was observed in the phagocytosis experiment of activated macrophages. Therefore, small and large molecules used for targeted modification have strong binding capability. However, these ligands, especially monoclonal antibodies, affect their activity due to storage conditions and modification preparation. Thus, the synthesis technology must be further improved in the future to reduce their adverse effects.
Functional Cells Serve as Targets of Nanomaterial-Based DDSs in the Treatment of OA
Targeting Macrophages
Macrophages play a key role in the pathogenesis of OA and directly influence joint degeneration by modulating the inflammatory response. The early stages of OA are usually associated with articular cartilage damage, which activates synovial macrophages.67 Once activated, M1 macrophages release large amounts of pro-inflammatory cytokines, such as IL-6, IL-1β and TNF-α. These cytokines not only exacerbate cartilage destruction but also stimulate synovial membrane proliferation, which results in persistent chronic inflammation in the joint cavity.84 However, in addition to their destructive role, macrophages can have beneficial effects. Notably, M2 macrophages promote tissue repair and reduce inflammation.85
Therefore, symptoms of OA can be alleviated by modulating the macrophage phenotype, reducing the expressions of inflammatory factors and scavenging reactive oxygen species (ROS) and NO in the body.86 Activated macrophages have several overexpressed specific receptors on their surface, such as FR2, scavenger receptor class A (SR-A) and CD44.87,88 Some nanomaterial-based DDSs can selectively deliver drugs to OA-lesion areas through passive targeting effects and actively target macrophages via nanomaterial-based DDSs surface ligands linked to specific receptors (Figure 2). Table 1 shows the nanomaterial-based DDSs targeting macrophages for the treatment of OA.
![]() |
Table 1 Nanomaterial-Based DDSs Targeting Macrophages for the Treatment of OA |
Nanomaterial-based DDSs employ various mechanisms to target macrophages for the treatment of OA. Ligands on the surface of nanomaterial-based DDSs can bind to specific receptors on the macrophage surface and enhance targeting recognition. Antibodies are common ligands on the surface of nanomaterial-based DDSs and have many applications in the targeting of macrophages for the treatment of OA.17,89,94,95 Kou et al89 prepared opsonized NPs (IgG/Bb@BRPL) by combining IgG, an oxidative stress-responsive bilirubin grafted polylysine biomaterial (BR-PLL), and an anti-inflammatory agent berberine (Bb). IgG/Bb@BRPL specifically targets M1 macrophages in the OA joint cavity by binding IgG to macrophage surface receptors. Bb and BR-PLL can then inhibit the inflammation of M1 macrophages, promote their polarization into M2 macrophages, and ultimately play a therapeutic role in OA. In addition, peptides can be used as ligands on the surface of NPs to target macrophages for the treatment of OA. Qi et al16 prepared MMP9-targeting peptide-modified mesoporous Prussian blue NPs (MPB NPs) for targeted delivery to synovial macrophages. By loading rapamycin (RAPA), the RAPA@MPB-MMP9 NPs inhibited macrophage pyroptosis and mammalian target of rapamycin (mTOR) complex 1 pathway activation, which attenuated inflammation. Small molecules are also common ligands on the surface of NPs that can target macrophages. She et al88 observed that dextran sulfate-triamcinolone acetonide conjugate NPs can selectively target SR-A on activated macrophages and mediate the internalization of NPs, which considerably inhibited the viability of activated M1 macrophages and ultimately reduced the expressions of inflammatory factors.
Nanomaterial-based DDSs can also be surface modified to increase their specific biocompatibility and targeting. Shi et al90 revealed that polyethylenimine (PEI)-functionalized diselenide-bridged mesoporous silica NPs (MSN-PEI) can target the joint lumen and aggregated at the site of OA inflammation upon entry into the body due to the presence of MSN-PEI surface charges interacting with cells. Subsequently, MSN-PEI inhibited inflammation and suppressed macrophage activation by reducing the binding of cfDNA to toll-like receptor (TLR9). Yan et al12 reported that after the “two‐in‐one” nanocarrier (NAHA‐CaP/siCA9 NPs) were passively targeted to the inflammatory sites in the joint cavity, the NP-loaded CA9 siRNA (siCA9) regulated the intracellular and extracellular pH balance, inhibited the inflammatory response of M1 macrophages, and promoted the repolarization of macrophages toward the M2 phenotype by binding to the CA9 mRNA overexpressed by OA macrophages.
Nanomaterial-based DDSs can be engineered to respond to the inflammatory microenvironment. They can undergo structural changes or release drugs in response to acidity, oxidative stress, or specific enzymes. This responsive targeting mechanism allows nanomaterial-based DDSs to aggregate and release therapeutic drugs specifically at the inflammation site, which increases the local drug concentration and enhances the therapeutic effect. Li et al85 indicated that after ROS‐responsive biodegradable NPs were introduced into the body via intravenous injection, the thioketal bonds of the NPs were broken in M1 macrophages with high ROS levels, which led to the dissociation of the NPs, release of the loaded drug, and a therapeutic effect. Similarly, Zhen et al102 showed that NPs (mPEG-TK-GLX@PVA-MMA) were ROS responsive and slowly released GLX351322 (GLX), which inhibited inflammation, ROS production, and ferroptosis in OA macrophages. Kang et al92 revealed that under acidic conditions, acid-activatable curcumin polymer microspheres rapidly dissociated due to the rapid hydrophobic/hydrophilic transition of protonated amine groups and responsively released curcumin, which exerted antioxidant, anti-inflammatory and anti-arthritic effects.
Therefore, nanomaterial-based DDSs can be used to treat OA through the precise delivery of drugs or genetic materials into OA macrophages through multiple targeting mechanisms. In addition, the retention time and specificity of nanomaterial-based DDSs in joints can be improved by optimizing their size, shape and surface functionalization, thus further enhancing their efficacy. Although nanomaterial-based DDSs still needs to be improved, their application in the treatment of OA targeting macrophages is very promising.
Targeting Synoviocytes
Under normal physiological conditions, synoviocytes are responsible for maintaining a healthy environment in the joint cavity. In the development of OA, synoviocytes proliferate and release inflammatory factors, which in turn promote the production of MMPs and accelerate cartilage degradation.110 Moreover, synovial hyperplasia leads to tissue hypertrophy, which restricts joint motion and exacerbates pain. Although synoviocytes may be involved in the repair mechanism in the early stages, their dysfunction and apoptosis increase with disease progression, which ultimately leads to joint dysfunction.111 Synoviocytes are both drivers of pathological changes and important indicators of disease progression in OA. Therefore, specific regulation and intervention of synoviocytes is the key to the treatment of OA. We can develop targeted drugs or treatments by identifying specific molecular markers or receptors related to synoviocytes, which inhibit the abnormal proliferation and inflammation of synoviocytes.
Nanomaterial-based DDSs can target synoviocytes to treat OA through surface modification and interaction with cell surface receptors. HA in CS/HA NPs can target synoviocytes through electrostatic interaction and binding with CD44 receptor on the surface of synoviocytes.69 Similarly, Deng et al112 and Zhou et al113 observed that HA in HA/CS-cytokine response modifier A and CS/HA/plasmid-DNA encoding IL-1 receptor antagonist gene can target CD44 receptor on the surface of synoviocytes, which inhibits synovitis and cartilage degradation. In addition, huntingtin-associated protein 1 peptide can be used as a ligand to bind to specific receptors on the surface of synoviocytes. Thus, the nanocomposite 4-arm-poly(ethylene glycol) (PEG)-maleimide/poly (lactic-co-glycolic) acid (PLGA) NP microgels can target synoviocytes. The microgels can remain in the joint cavity for a long time without causing articular cartilage degeneration or synovial thickening and thus have a good effect on relieving synovitis.68
Nanomaterial-based DDSs can also be used as drug delivery carriers to deliver various therapeutic drugs or therapeutic genes to the synovial tissue and play a therapeutic role. PLGA NPs can encapsulate diacetylsalicylic acid (DIA) and reduce inflammation by releasing DIA continuously.114 Similarly, PLGA NPs can encapsulate p16INK4a siRNA to form p16INK4a siRNA-loaded PLGA NPs (p16 si_NP). p16 si_NP mostly enters synoviocytes and some chondrocytes, which ultimately inhibits synovial inflammation, cartilage degradation, and pain in mice.115 Table 2 shows the nanomaterial-based DDSs targeting synoviocytes for the treatment of OA.
![]() |
Table 2 Nanomaterial-Based DDSs Targeting Synoviocytes for the Treatment of OA |
Nanomaterial-based DDSs target synoviocytes to inhibit synovitis, slow down cartilage degeneration, and ultimately treat OA. Ren et al116 reported that ceria NPs targeted synoviocytes and alleviated synoviocyte senescence by scavenging ROS and inactivating the NF-κB pathway. Histidine functionalized generation 5 polyamidoamine dendrimer delivered miR-224-5p into synoviocytes and protected them from degradation. Subsequently, they exerted anti-inflammatory, anti-apoptotic, antineovascularization and autophagy-promoting effects.122 Sanse Powder essential oil nanoemulsion targets synoviocytes, inhibits transient receptor potential ankyrin 1 (TRPA1) through the AMP-activated protein kinase (AMPK)-mTOR signaling pathway, and alleviates OA synovitis.117 Therefore, targeting synoviocytes to modulate multiple cytokines and signaling pathways to inhibit synovial inflammation and slow down cartilage degeneration may be a potential therapeutic strategy for OA (Figure 3).
Targeting MSCs
MSCs play an important role in OA. On the one hand, MSCs can undergo self-renewal and multidirectional differentiation and differentiate into chondrocytes, osteoblasts, etc., which promote the repair of damaged joint tissues.123 However, during the pathogenesis of OA, changes in the joint microenvironment, release of inflammatory factors, and increased oxidative stress may affect the function of MSCs. On the other hand, MSCs can also regulate the local immune response and inflammatory state of joints by secreting various bioactive factors, such as growth and anti-inflammatory factors.124 Nanomaterial-based DDSs targeting MSCs shows great potential in the treatment of OA. Nanomaterial-based DDSs not only enhance the viability of the stem cells but also provide sustained drug release, which further enhances the therapeutic effect. Table 3 shows the nanomaterial-based DDSs targeting MSCs for the treatment of OA.
![]() |
Table 3 Nanomaterial-Based DDSs Targeting Mesenchymal Stem Cells for the Treatment of OA |
A variety of nanomaterial-based DDSs have demonstrated unique advantages in targeting MSCs for the treatment of OA. These nanomaterial-based DDSs include PLGA NPs, polydopamine (PDA)-modified NPs, and CS (CHI)-bonded NPs with kartogenin (KGN).139 PLGA NPs can be used as KGN carrier and prepared through emulsification–solvent evaporation method. The PDA modified on its surface can scavenge ROS and reduce oxidative stress.133 PDA can also improve the hydrophilicity and biocompatibility of NPs and increase their stability and circulation time in vivo. The NPs formed by the combination of CHI and KGN can promote the differentiation of MSCs into chondrocytes and enhance their ability for cartilage regeneration.135 In addition, magnetic NPs, such as iron oxide NPs, can deliver KGN through surface modification and external magnetic field, which can improve the concentration and residence time of drugs in joints.129 These nanomaterial-based DDSs can target MSCs through magnetic field, electrostatic attraction, pH response, and mutual recognition between surface ligands and MSC surface receptors and play a therapeutic role (Figure 4).
The mechanism of nanomaterial-based DDSs targeting MSCs in the treatment of OA mainly includes the following aspects. First, nanomaterial-based DDSs can accurately deliver therapeutic drugs to MSCs and regulate their biological functions, such as proliferation, differentiation, and migration. Nanomaterial-based DDSs can also deliver KGN to MSCs, induce MSCs to differentiate into chondrocytes, and repair damaged cartilage tissues.131 Second, nanomaterial-based DDSs can improve the microenvironment of MSCs, reduce inflammation, and inhibit apoptosis. Antioxidants carried by nanomaterial-based DDSs can eliminate ROS, reduce the damage of oxidative stress to MSCs, and maintain the normal function of MSCs.138 In addition, nanomaterial-based DDSs can reduce inflammation and promote the repair and regeneration of joint tissue by regulating the immune function of MSCs.
Therefore, nanomaterial-based DDSs targeting MSCs is a promising treatment strategy for OA and has great potential for clinical application. By thoroughly studying the characteristics and mechanism of nanomaterial-based DDSs and optimising the treatment plan, more effective treatment methods can be provided for OA patients.
Targeting Chondrocytes
Chondrocytes are the only cell type found in articular cartilage. In the OA microenvironment, inflammatory factors, ROS, and MMPs can induce abnormal chondrocyte function, which further disrupts cartilage homeostasis and accelerates cartilage degradation.140 The damaged chondrocytes in turn secrete inflammatory mediators that further exacerbate the local inflammatory response, which creates a vicious cycle that accelerates the progression of OA.141 Therefore, selective chondrocyte-targeted modulation may be a promising therapeutic approach for OA. Table 4 provides the nanomaterial-based DDSs targeting chondrocytes for the treatment of OA. Figure 5 shows the mechanism of action of nanomaterial-based DDSs targeting chondrocytes for the treatment of OA.
![]() |
Table 4 Nanomaterial-Based DDSs Targeting Chondrocytes for the Treatment of OA |
In the design and development of nanomaterial-based DDSs, targeting is the key to improving therapeutic efficacy. Through surface modification and ligand attachment, nanomaterial-based DDSs can specifically recognize chondrocytes. Researchers often use specific antibodies or targeting peptides to increase the affinity of nanomaterial-based DDSs for chondrocytes, a process that not only improves drug targeting but also reduces damage to surrounding healthy tissues. Liang et al15 designed a melatonin-loaded nano-delivery system (MT@PLGA-COLBP) whose surface cartilage-targeting peptide (COLBP) can specifically bind to type II collagen on the surface of chondrocytes, which allows the NPs to accumulate and target chondrocytes in the joints. Subsequently, MT@PLGA-COLBP released melatonin, which exerted a protective effect on chondrocytes by inhibiting the TLR2/4-myeloid differentiation primary response 88 (MyD88)-NF-κB signaling pathway and scavenging ROS. Similarly, Xiong et al66 observed that formononetin (FMN)-PEG (denoted as PCFMN), consisting of PEG, cartilage targeting peptide (CollBP), and FMN, can be used for the treatment of OA. CollBP bound to collagen II, which made PCFMN cartilage-targeted. PEG increased the solubility of the drug. FMN showed anti-inflammatory and antioxidant effects, which protected chondrocytes from oxidative stress damage and activated the Nrf2-dependent antioxidant pathway. Chen et al169 also developed a WYRGRL-modified HA-based cartilage-targeted nanomicelles (C-HA-DOs). The system consisted of HA, WYRGRL peptide, and dodecylamine. The WYRGRL peptide can specifically bind to collagen II on the surface of chondrocytes to target them. After the NPs entered the cells, Pio@C-HA-DOs protected chondrocytes from oxidative stress damage by decreasing the intracellular ROS levels and restoring the mitochondrial membrane potential. In addition, the physicochemical properties of nanomaterial-based DDSs, such as size, shape, and surface charge, influence their distribution and targeting ability in vivo.147,149 Some nanomaterial-based DDSs can also respond to physiological signals in vivo, such as pH and redox potential, to realize intelligent drug release and further improve treatment precision.146,182
Nanomaterial-based DDSs can slowly release drugs after entering chondrocytes through endocytosis, which in turn regulates the physiological activities and biological function of chondrocytes. Nanomaterial-based DDSs can effectively deliver anti-inflammatory drugs and promote the regeneration of repair factors, which alleviates symptoms and promotes cartilage repair in OA. PLGA NPs can encapsulate and continuously release drugs, such as rapamycin, which effectively alleviates the symptoms of OA.139 Some nanomaterial-based DDSs can promote the proliferation and differentiation of chondrocytes, which improves the repair ability of cartilage tissue. CS NPs can improve chondrocyte activity, promote cartilage matrix synthesis, and reduce chondrocyte apoptosis.142 In addition, nanomaterial-based DDSs can inhibit the inflammatory response and oxidative stress in chondrocytes, which reduces the progression of OA.152 Therefore, Nanomaterial-based DDSs targeting chondrocytes also have great prospects for application in OA treatment.
Functional Cells Serve as Arrows of Nanomaterial-Based DDSs in the Treatment of OA
Although nanomaterial-based DDSs have achieved great research progress in targeting functional cells to treat OA, some shortcomings remain. Exogenous nanomaterial-based DDSs cannot avoid phagocytosis by reticuloendothelial cells and have poor long-term stability and safety.209 However, endogenous bionanomimetic membranes display many advantages in nanomaterial-based DDSs, such as low immunogenicity, capability to interact with natural autologous components, long duration of action, and low toxicity and side effects.210 EVs can also communicate between cells and transport cytokines, small-molecule metabolites, or enzymes to different cells.211 Therefore, during treatment of OA using nanomaterial-based DDSs, in addition to targeting functional cells, such as macrophages, synoviocytes, MSCs, and chondrocytes, cell membranes or EVs can be used as arrows to target primitive cells (Table 5). Figure 6 briefly illustrates the arrows of nanomaterial-based DDSs for the treatment of OA.
![]() |
Table 5 Functional Cells Serve as Arrows in the Treatment of OA by Nanomaterial-Based DDSs |
Nanomaterial-Based DDSs Apply Cell Membranes as Arrows
Nanomaterials modified by the cell membrane retain antigens and the cell membrane structure and thus possess the unique functions of protocells, such as active targeting, long-term blood circulation, and immune escape.221 These characteristics render this type of materials with a great potential for the treatment of OA. A variety of biomembrane nanocarriers have been developed, and they mainly include macrophage, MSC, and chondrocyte membranes.18,213
During inflammation of the body, a large number of immune cells are activated. In the development of OA, macrophages accumulate in the synovium and joint cavity, where inflammation occurs and induces polarization reaction.222 Therefore, the biomimetic carriers of macrophage membrane have attracted increasing attention in the field of drug delivery, and the polarization of M2 macrophages shows a great potential in the treatment of OA. Ma et al223 used mouse macrophage membrane to cover nanogelatin and ChS and crosslinked them to form a nanogel surface to construct artificial M2 macrophages. The results show that, compared to the nanogel without a membrane coverage, the artificial M2 macrophages had evident adhesion and accumulation on the surface of inflammatory cartilage and synovium and an anti-inflammatory effect. Zhou et al18 used PEI to condense cell-penetrating peptide Kafak and shRNA-LEPR, modified them with HA, and coated M2 macrophage membrane to develop an M2H @ RPK nanotherapeutic system. M2H @ RPK had the highest internalization degree in synoviocytes and can deliver shRNA-LEPR efficiently. Therefore, polarized biomimetic nanocarriers of macrophage membrane can effectively target inflammatory tissues and cells in joints using specific antibodies and chemotaxis on macrophage membrane. In addition, it can also inhibit OA inflammation and delay the progression of OA by promoting macrophage repolarization.
The MSC membrane also serves as one of the carriers commonly used in nanodelivery systems. MSCs have multidirectional differentiation potential and immunomodulatory function, and their cell membranes can retain these characteristics.215 The application of MSC membrane in nanomaterial-based DDSs can improve their biocompatibility and safety and reduce immune rejection. Zhang et al217 created MSCs in bone marrow highly expressing CXC chemokine receptor 4 (CXCR4) through genetic engineering technology, which enhanced their ability to migrate to the injured site. Then, the cell membranes of these cells were separated, purified, and coated on fluorescent NPs to prepare NPs with an active targeting capability. These NPs can increase the uptake and penetration depth of damaged chondrocytes and protect the damaged cartilage. However, the stability and functional integrity of the MSC membrane may be challenged during storage and transportation and thus need further improvement.
Chondrocyte membrane has the physiological function of chondrocytes and can achieve targeted drug delivery. Coating chondrocyte membrane on nanomaterial-based DDSs can improve their uptake and accumulation in chondrocytes. A kind of PLGA NPs coated with chondrocyte membrane can effectively penetrate the cartilage tissue, deliver drugs to chondrocytes, inhibit chondrocyte apoptosis, and promote the synthesis of cartilage matrix.210 Apoptotic chondrocyte membrane can be recognized and absorbed by macrophages. Coating apoptotic chondrocyte membrane on nanomaterial-based DDSs can enhance the accumulation and retention of nanomaterial-based DDSs at inflammatory sites and improve the therapeutic effect of drugs. The hollow mesoporous cerium oxide nanospheres coated with apoptotic chondrocyte membrane can effectively simulate chondrocyte apoptosis bodies in joints, which enables the targeted delivery of the anti-inflammatory drug dexamethasone and subsequent controlled release triggered by the acidic environment of OA. These nanomaterial-based DDSs can eventually eliminate ROS, relieve inflammation, reduce mitochondrial damage, and inhibit chondrocyte apoptosis.218
However, despite the remarkable progress of cell membranes as carriers of nanomaterial-based DDSs in the treatment of OA, several challenges remain. Future studies need to continuously improve the stability and biocompatibility of cell membranes and optimize their modification and functionalization strategies to provide effective means for the treatment of OA.
Nanomaterial-Based DDSs Apply EVs as Arrows
EVs are mainly composed of apoptotic vesicles, microparticles, microvesicles, and exosomes.224 They can facilitate information transfer between cells by carrying biomolecules, such as proteins, lipids, RNA, etc., and act as carriers to transport bioactive molecules (eg, drugs and signaling molecules) to the target cells.211 EVs can inherit some endogenous protein ligands from the source cells and actively target specific loci.
MSC-EVs can help nanomaterial-based DDSs to recognize the site of inflammation due to their homing capability. D’Atri et al214 removed the cytoplasm of MSCs and transformed them into nanosized vesicles. The accumulation of nanovesicles in inflammatory chondrocytes exceeded that in healthy cells. These nanovesicles can target inflammatory cells and show good anti-inflammatory effect, and thus, they are ideal targeted drug-delivery carriers. Colombini et al225 extracted the exosomes of adipose-derived MSCs to deliver miRNA to the cartilage layer. The penetration test of chondrocyte explants revealed the highest content of exosomes extracted from the collagen-rich cartilage matrix layer. EVs contain a variety of biomolecules that have high targeting and biological safety and can selectively act on recipient cells or deliver biological information to regulate cell functions.
In addition, the common exosomes in EVs, with a diameter of 40–160 nm, mainly originate from the inner buds of cells and surrounded by lipid bilayers.226,227 Given their immune escape, anti-inflammation, and good capability to target chondrocytes, MSC exosomes are also often used as carriers of nanomaterial-based DDSs to target inflammatory sites. Li et al19 developed CD90 MSC-derived microvesicles-encapsulated NPs (CD90@NPs) and observed that CD90@NPs can bind to damaged primary chondrocytes. Then, they encapsulated a model glucocorticoid, triamcinolone acetonide, in CD90@NP to form T-CD90@NPs. The injection of T-CD90@NP into the articular cavity can inhibit inflammation and apoptosis and promote cartilage regeneration and repair. Yang et al219 prepared magnetic polysaccharide microcarriers (MPM) composed of natural polysaccharides, Fe₃O₄@MgSiO₃ and an anti-inflammatory drug, diclofenac sodium (DS), and loaded them with exosomes. The MPM showed a good biocompatibility, promoted chondrocyte proliferation, migration, and differentiation, recruited stem cells, and induced macrophage polarization toward M2 phenotypes. MPM-loaded DS and Exo synergistically protected chondrocytes in vitro and effectively attenuated cartilage degradation and promoted chondrogenesis in OA rats in vivo. This finding was comparable to the effect of MSCs. Cao et al220 prepared a hydrogel microsphere (GM@PDA@S-EXO) enriched with exosomes of SOD3. GM@PDA@S-EXO effectively delivered SOD3 to cartilages, improved the antioxidant capacity of chondrocytes, reduced the level of mitochondrial ROS, and attenuated the progression of OA.
At present, the new method of therapeutic drug delivery by EVs has a great application prospect in the treatment of OA. As a new model of stem-cell therapy, EVs have opened a new avenue to reduce the treatment risk of OA. However, standardized EV separation technology and quality control means remain lacking, and thus, EV preparation and purification technology must be further explored to ensure their quality and therapeutic effect.
Conclusion and Prospects
With the gradual application of nanomaterial-based DDSs in the treatment of OA, an increasing number of studies have shown that functional cells play an important role as targets or arrows. Nanomaterial-based DDSs targeting functional cells represents a breakthrough frontier in OA therapeutics. The unique advantages of nanomaterials, such as tunable biodistribution, enhanced cellular uptake and stimuli-responsive drug release, make it achieve unprecedented accuracy in modulating chondrocytes, macrophages, synoviocytes and MCSs. This review describes the mechanism of action and research progress of nanomaterial-based DDS-targeted functional cell therapy for OA and discusses how functional cells can be used as the arrow of nanomaterial-based DDSs in the treatment of OA. Nanomaterial-based DDSs use cell membrane and EVs as arrows, which can improve the drug targeting and biocompatibility and reduce side effects.212,220 Therefore, nanomaterial-based DDSs are expected to become a new means for OA treatment.
Although nanomaterial-based DDSs have obvious curative effects in the treatment of OA, there are still some challenges in clinical application. For example, inconsistent targeting efficiency in heterogeneous OA microenvironment and unresolved long-term biocompatibility concerns. This is mainly due to the clinical complexity of disease progression in patients with OA, and the inability to precisely control the release of drugs in nanomaterial-based DDSs according to the disease conditions of different patients.209 In addition, the toxicity and elimination of nanomaterial-based DDSs are key challenges limiting clinical translation. Although liposomal and polymeric NPs can be cleared by the reticuloendothelial system, when their particle size is too small, they can easily penetrate vascular endothelium and enter parenchymal organs, resulting in off-target toxicity.161,165 Notably, nanomaterial-based DDSs mainly rely on renal excretion, hepatic metabolism and phagocytosis by the reticuloendothelial system for in vivo elimination, but the elimination efficiency varies significantly among different materials.228 For example, dendritic polymers are difficult to filter through glomeruli due to their dense structure and may be retained in the liver, whereas iron oxide NPs can be phagocytosed by macrophages and then excreted via the hepatobiliary system.229–231 Therefore, there are still many issues that need to be addressed before nanomaterial-based DDSs can be better applied in OA therapy.
In the future, the research on nanomaterial-based DDS-targeted functional cell therapy for OA will continue to deepen. First, with the continuous development of nanotechnology, more novel nanodelivery systems will be developed to provide additional options for OA treatment. Smart NPs can automatically adjust drug release based on changes in the body environment to improve their therapeutic effect.232 Multifunctional NPs can simultaneously target multiple therapeutic targets to realize synergistic therapy.233 Second, with the in-depth understanding of OA pathophysiology, more new therapeutic targets will be discovered. Nanomaterial-based DDSs can treat OA by regulating gut flora and inhibiting inflammation. Third, combination therapy is a future development direction.234 Combining nanomaterial-based DDS-targeted functional cell therapy with traditional therapeutic methods, such as physical and drug therapies, can improve the therapeutic outcomes and reduce side effects. Fourth, integrate nanotechnology with gene-editing tools or design artificial intelligence-driven platforms to develop multi-mode treatments. In future research, we need to strengthen multidisciplinary cooperation and integrate the knowledge and technology of nanotechnology, biology, medicine, and other fields to jointly promote the research and application of nanomaterial-based DDS-targeted functional cell therapy for OA.
Data Sharing Statement
Data sharing is not applicable to this article as no new data was created or analyzed in this study.
Acknowledgment
This work was supported by the National Natural Science Foundation of China (Grant No. 32371184), Liaoning Province Applied Basic Research Program (No. 2023JH2/101300072), the Science and Technology Innovation Program of the General Administration of Sport of China (No. 22KJCX040) and Basic Scientific Research Project of the Higher Education Institutions of Liaoning Province (No. JYTZD2023131). The Figures were Created in BioRender (https://www.biorender.com/).
Disclosure
The authors report no conflicts of interest in this work.
References
1. Tang S, Zhang C, Oo WM. et al. Osteoarthritis. Nat Rev Dis Primers. 2025;11:10. doi:10.1038/s41572-025-00594-6
2. Courties A, Kouki I, Soliman N, Mathieu S, Sellam J. Osteoarthritis year in review 2024: epidemiology and therapy. Osteoarthritis Cartilage. 2024;32:1397–1404. doi:10.1016/j.joca.2024.07.014
3. Tian R, Su S, Yu Y, et al. Revolutionizing osteoarthritis treatment: how mesenchymal stem cells hold the key. Biomed Pharmacother. 2024;173:116458. doi:10.1016/j.biopha.2024.116458
4. Chang J-W, Tang C-H. The role of macrophage polarization in rheumatoid arthritis and osteoarthritis: pathogenesis and therapeutic strategies. Int Immunopharmacol. 2024;142:113056. doi:10.1016/j.intimp.2024.113056
5. Ruan Q, Wang C, Zhang Y, Sun J. Ruscogenin attenuates cartilage destruction in osteoarthritis through suppressing chondrocyte ferroptosis via Nrf2/SLC7A11/GPX4 signaling pathway. Chem Biol Interact. 2024;388:110835. doi:10.1016/j.cbi.2023.110835
6. Yu H, Huang Y, Yang L. Research progress in the use of mesenchymal stem cells and their derived exosomes in the treatment of osteoarthritis. Ageing Res Rev. 2022;80:101684. doi:10.1016/j.arr.2022.101684
7. Liu B, Xian Y, Chen X, et al. Inflammatory fibroblast-like synoviocyte-derived exosomes aggravate osteoarthritis via enhancing macrophage glycolysis. Adv Sci. 2024;11:e2307338. doi:10.1002/advs.202307338
8. Haber T, Hall M. When should we treat knee osteoarthritis and with which treatments? Lancet Rheumatol. 2024;6:e195–e196. doi:10.1016/S2665-9913(24)00006-7
9. Gezer HH, Ostor A. What is new in pharmacological treatment for osteoarthritis? Best Pract Res Clin Rheumatol. 2023;37:101841. doi:10.1016/j.berh.2023.101841
10. Kjeken I, Bordvik DH, Osteras N, et al. Efficacy and safety of non-pharmacological, pharmacological and surgical treatments for hand osteoarthritis in 2024: a systematic review. RMD Open. 2025;11(1):e004963. doi:10.1136/rmdopen-2024-004963
11. Lei T, Wang Y, Li M, Hua L. Clinical efficacy of multiple intra-articular injection for hip osteoarthritis. Bone Joint J. 2024;106-B:532–539. doi:10.1302/0301-620X.106B6.BJJ-2023-1272.R1
12. Yan Y, Lu A, Dou Y, et al. Nanomedicines reprogram synovial macrophages by scavenging nitric oxide and silencing CA9 in progressive osteoarthritis. Adv Sci. 2023;10:e2207490. doi:10.1002/advs.202207490
13. Zhu Y, Cao L, Yuan M, et al. Microgel encapsulated mesoporous silica nanoparticles for releasing Wnt16 to synergistically treat temporomandibular joint osteoarthritis. Adv Sci. 2024;11(41):e2404396. doi:10.1002/advs.202404396
14. Peng Y, Wang Y, Bai R, et al. Nanomaterials: recent advances in knee osteoarthritis treatment. Adv Healthc Mater. 2024;13(32):e2400615. doi:10.1002/adhm.202400615
15. Liang H, Yan Y, Sun W, et al. Preparation of melatonin-loaded nanoparticles with targeting and sustained release function and their application in osteoarthritis. Int J Mol Sci. 2023;24.
16. Qi W, Jin L, Huang S, et al. Modulating synovial macrophage pyroptosis and mitophagy interactions to mitigate osteoarthritis progression using functionalized nanoparticles. Acta Biomater. 2024;181:425–439. doi:10.1016/j.actbio.2024.05.014
17. Huang H, Zheng S, Wu J, et al. Opsonization inveigles macrophages engulfing carrier-free bilirubin/JPH203 nanoparticles to suppress inflammation for osteoarthritis therapy. Adv Sci. 2024;11:e2400713. doi:10.1002/advs.202400713
18. Zhou K, Yang C, Shi K, et al. Activated macrophage membrane-coated nanoparticles relieve osteoarthritis-induced synovitis and joint damage. Biomaterials. 2023;295:122036. doi:10.1016/j.biomaterials.2023.122036
19. Li Y, Tu Q, Xie D, et al. Triamcinolone acetonide-loaded nanoparticles encapsulated by CD90+ MCSs-derived microvesicles drive anti-inflammatory properties and promote cartilage regeneration after osteoarthritis. J Nanobiotechnol. 2022;20:150. doi:10.1186/s12951-022-01367-z
20. Martel-Pelletier J, Barr AJ, Cicuttini FM, et al. Osteoarthritis. Nat Rev Dis Primers. 2016;2:16072. doi:10.1038/nrdp.2016.72
21. Hügle T, Geurts J. What drives osteoarthritis?-synovial versus subchondral bone pathology. Rheumatology. 2017;56:1461–1471. doi:10.1093/rheumatology/kew389
22. Rosshirt N, Trauth R, Platzer H, et al. Proinflammatory T cell polarization is already present in patients with early knee osteoarthritis. Arthritis Res Ther. 2021;23:37. doi:10.1186/s13075-020-02410-w
23. Atukorala I, Kwoh CK, Guermazi A, et al. Synovitis in knee osteoarthritis: a precursor of disease? Ann Rheum Dis. 2016;75:390–395. doi:10.1136/annrheumdis-2014-205894
24. Cho Y, Jeong S, Kim H, et al. Disease-modifying therapeutic strategies in osteoarthritis: current status and future directions. Exp mol Med. 2021;53:1689–1696. doi:10.1038/s12276-021-00710-y
25. Robinson WH, Lepus CM, Wang Q, et al. Low-grade inflammation as a key mediator of the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 2016;12:580–592. doi:10.1038/nrrheum.2016.136
26. Wang S, Li W, Zhang P, et al. Mechanical overloading induces GPX4-regulated chondrocyte ferroptosis in osteoarthritis via Piezo1 channel facilitated calcium influx. J Adv Res. 2022;41:63–75. doi:10.1016/j.jare.2022.01.004
27. Hu W, Chen Y, Dou C, Dong S. Microenvironment in subchondral bone: predominant regulator for the treatment of osteoarthritis. Ann Rheum Dis. 2021;80:413–422. doi:10.1136/annrheumdis-2020-218089
28. Cardoneanu A, Macovei LA, Burlui AM, et al. Temporomandibular joint osteoarthritis: pathogenic mechanisms involving the cartilage and subchondral bone, and potential therapeutic strategies for joint regeneration. Int J Mol Sci. 2022;24(1):171. doi:10.3390/ijms24010171
29. Xu Z, Chen T, Luo J, et al. Cartilaginous metabolomic study reveals potential mechanisms of osteophyte formation in osteoarthritis. J Proteome Res. 2017;16(4):1425–1435. doi:10.1021/acs.jproteome.6b00676
30. Tong L, Yu H, Huang X, et al. Current understanding of osteoarthritis pathogenesis and relevant new approaches. Bone Res. 2022;10:60. doi:10.1038/s41413-022-00226-9
31. Rice SJ, Beier F, Young DA, Loughlin J. Interplay between genetics and epigenetics in osteoarthritis. Nat Rev Rheumatol. 2020;16:268–281. doi:10.1038/s41584-020-0407-3
32. Charlier E, Deroyer C, Ciregia F, et al. Chondrocyte dedifferentiation and osteoarthritis (OA). Biochem Pharmacol. 2019;165:49–65. doi:10.1016/j.bcp.2019.02.036
33. Fu W, Vasylyev D, Bi Y, et al. Nav1.7 as a chondrocyte regulator and therapeutic target for osteoarthritis. Nature. 2024;625:557–565. doi:10.1038/s41586-023-06888-7
34. Yang J, Li S, Li Z, et al. Targeting YAP1-regulated glycolysis in fibroblast-like synoviocytes impairs macrophage infiltration to ameliorate diabetic osteoarthritis progression. Adv Sci. 2024;11:e2304617. doi:10.1002/advs.202304617
35. Lei J, Jiang X, Li W, et al. Exosomes from antler stem cells alleviate mesenchymal stem cell senescence and osteoarthritis. Protein Cell. 2022;13:220–226. doi:10.1007/s13238-021-00860-9
36. Bonato A, Fisch P, Ponta S, et al. Engineering inflammation-resistant cartilage: bridging gene therapy and tissue engineering. Adv Healthc Mater. 2023;12:e2202271. doi:10.1002/adhm.202202271
37. Shi Y, Hu X, Cheng J, et al. A small molecule promotes cartilage extracellular matrix generation and inhibits osteoarthritis development. Nat Commun. 2019;10:1914. doi:10.1038/s41467-019-09839-x
38. Chen H, Ye T, Hu F, et al. Urchin-like ceria nanoparticles for enhanced gene therapy of osteoarthritis. Sci Adv. 2023;9:eadf0988. doi:10.1126/sciadv.adf0988
39. Molnar V, Matišić V, Kodvanj I, et al. Cytokines and chemokines involved in osteoarthritis pathogenesis. Int J Mol Sci. 2021;22(17):9208. doi:10.3390/ijms22179208
40. Liu S, Deng Z, Chen K, et al. Cartilage tissue engineering: from proinflammatory and anti‑inflammatory cytokines to osteoarthritis treatments. mol Med Rep. 2022;25. doi:10.3892/mmr.2022.12615
41. Boehme KA, Rolauffs B. Onset and progression of human osteoarthritis-can growth factors, inflammatory cytokines, or differential miRNA expression concomitantly induce proliferation, ECM degradation, and inflammation in articular cartilage? Int J Mol Sci. 2018;19:2282. doi:10.3390/ijms19082282
42. Ko C-Y, Lin -Y-Y, Achudhan D, et al. Omentin-1 ameliorates the progress of osteoarthritis by promoting IL-4-dependent anti-inflammatory responses and M2 macrophage polarization. Int J Biol Sci. 2023;19:5275–5289. doi:10.7150/ijbs.86701
43. Zheng W, Zhou T, Zhang Y, et al. Simplified α2-macroglobulin as a TNF-α inhibitor for inflammation alleviation in osteoarthritis and myocardial infarction therapy. Biomaterials. 2023;301:122247. doi:10.1016/j.biomaterials.2023.122247
44. Guo D, Kan S, Zhang L, et al. IL-10 enhances cell-to-cell communication in chondrocytes via STAT3 signaling pathway. Cell Signal. 2023;105:110605. doi:10.1016/j.cellsig.2023.110605
45. Shang X, Böker KO, Taheri S, et al. The interaction between microRNAs and the Wnt/β-catenin signaling pathway in osteoarthritis. Int J Mol Sci. 2021;22(18):9887. doi:10.3390/ijms22189887
46. Choi M-C, Jo J, Park J, Kang HK, Park Y. NF-κB signaling pathways in osteoarthritic cartilage destruction. Cells. 2019;8(7):734. doi:10.3390/cells8070734
47. Li T, Peng J, Li Q, et al. The mechanism and role of ADAMTS protein family in osteoarthritis. Biomolecules. 2022;12.
48. Chiu Y-S, Bamodu OA, Fong I-H, et al. The JAK inhibitor Tofacitinib inhibits structural damage in osteoarthritis by modulating JAK1/TNF-alpha/IL-6 signaling through Mir-149-5p. Bone. 2021;151:116024. doi:10.1016/j.bone.2021.116024
49. Rabie MA, Sayed RH, Venkatesan JK, et al. Intra-articular injection of rAAV-hFGF-2 ameliorates monosodium iodoacetate-induced osteoarthritis in rats via inhibiting TLR-4 signaling and activating TIMP-1. Toxicol Appl Pharmacol. 2023;459:116361. doi:10.1016/j.taap.2022.116361
50. Ehrich EW, Schnitzer TJ, McIlwain H, et al. Effect of specific COX-2 inhibition in osteoarthritis of the knee: a 6 week double blind, placebo controlled pilot study of rofecoxib. Rofecoxib Osteoarthritis Pilot Study Group. J Rheumatol. 1999;26:2438–2447.
51. Chen M, Lu Y, Liu Y, et al. Injectable microgels with hybrid exosomes of chondrocyte-targeted FGF18 gene-editing and self-renewable lubrication for osteoarthritis therapy. Adv Mater. 2024;36:e2312559. doi:10.1002/adma.202312559
52. Efferth T, Oesch F. The immunosuppressive activity of artemisinin-type drugs towards inflammatory and autoimmune diseases. Med Res Rev. 2021;41:3023–3061. doi:10.1002/med.21842
53. Perruccio AV, Young JJ, Wilfong JM, et al. Osteoarthritis year in review 2023: epidemiology & therapy. Osteoarthritis Cartilage. 2024;32:159–165. doi:10.1016/j.joca.2023.11.012
54. Richard MJ, Driban JB, McAlindon TE. Pharmaceutical treatment of osteoarthritis. Osteoarthritis Cartilage. 2023;31:458–466. doi:10.1016/j.joca.2022.11.005
55. Oo WM, Liu X, Hunter DJ. Pharmacodynamics, efficacy, safety and administration of intra-articular therapies for knee osteoarthritis. Expert Opin Drug Metab Toxicol. 2019;15:1021–1032. doi:10.1080/17425255.2019.1691997
56. Paget LDA, Mokkenstorm MJ, Tol JL, Kerkhoffs GMMJ, Reurink G. What is the efficacy of intra-articular injections in the treatment of ankle osteoarthritis? A systematic review. Clin Orthop Relat Res. 2023;481:1813–1824. doi:10.1097/CORR.0000000000002624
57. Conley B, Bunzli S, Bullen J, et al. Core recommendations for osteoarthritis care: a systematic review of clinical practice guidelines. Arthritis Care Res. 2023;75:1897–1907. doi:10.1002/acr.25101
58. Moseng T, Vliet Vlieland TPM, Battista S, et al. EULAR recommendations for the non-pharmacological core management of Hip and knee osteoarthritis: 2023 update. Ann Rheum Dis. 2024;83:730–740. doi:10.1136/ard-2023-225041
59. Cotter EJ, Weissman AC, Yazdi AA, Muth SA, Cole BJ. Arthroscopic debridement of mild and moderate knee osteoarthritis results in clinical improvement at short-term follow-up: a systematic review. Arthroscopy. 2025;41:377–389. doi:10.1016/j.arthro.2024.03.016
60. Orange GM, Hince DA, Travers MJ, et al. Physical function following total knee arthroplasty for osteoarthritis: a longitudinal systematic review with meta-analysis. J Orthop Sports Phys Ther. 2025;55(1):1–11. doi:10.2519/jospt.2024.12570
61. Jones IA, Togashi R, Wilson ML, Heckmann N, Vangsness CT. Intra-articular treatment options for knee osteoarthritis. Nat Rev Rheumatol. 2019;15:77–90. doi:10.1038/s41584-018-0123-4
62. Malek N, Mrugala M, Makuch W, et al. A multi-target approach for pain treatment: dual inhibition of fatty acid amide hydrolase and TRPV1 in a rat model of osteoarthritis. Pain. 2015;156:890–903. doi:10.1097/j.pain.0000000000000132
63. Grol MW. The evolving landscape of gene therapy strategies for the treatment of osteoarthritis. Osteoarthritis Cartilage. 2024;32:372–384. doi:10.1016/j.joca.2023.12.009
64. Yi X, Leng P, Wang S, Liu L, Xie B. Functional nanomaterials for the treatment of osteoarthritis. Int J Nanomed. 2024;19:6731–6756. doi:10.2147/IJN.S465243
65. Zhou D, Zhou F, Sheng S, et al. Intra-articular nanodrug delivery strategies for treating osteoarthritis. Drug Discov Today. 2023;28(3):103482. doi:10.1016/j.drudis.2022.103482
66. Xiong W, Lan Q, Liang X, et al. Cartilage-targeting poly(ethylene glycol) (PEG)-formononetin (FMN) nanodrug for the treatment of osteoarthritis. J Nanobiotechnol. 2021;19:197. doi:10.1186/s12951-021-00945-x
67. Zhang H, Lin C, Zeng C, et al. Synovial macrophage M1 polarisation exacerbates experimental osteoarthritis partially through R-spondin-2. Ann Rheum Dis. 2018;77:1524–1534. doi:10.1136/annrheumdis-2018-213450
68. Mancipe Castro LM, Sequeira A, García AJ, Guldberg RE. Articular cartilage- and synoviocyte-binding poly(ethylene glycol) nanocomposite microgels as intra-articular drug delivery vehicles for the treatment of osteoarthritis. ACS Biomater Sci Eng. 2020;6:5084–5095. doi:10.1021/acsbiomaterials.0c00960
69. Qiu B, Xu X-F, Deng R-H, et al. Hyaluronic acid-chitosan nanoparticles encoding CrmA attenuate interleukin-1β induced inflammation in synoviocytes in vitro. Int J Mol Med. 2019;43:1076–1084. doi:10.3892/ijmm.2018.3997
70. Stapleton M, Sawamoto K, Alméciga-Díaz CJ, et al. Development of bone targeting drugs. Int J Mol Sci. 2017;18(7):1345. doi:10.3390/ijms18071345
71. Rothenfluh DA, Bermudez H, O’Neil CP, Hubbell JA. Biofunctional polymer nanoparticles for intra-articular targeting and retention in cartilage. Nat Mater. 2008;7:248–254.
72. Pi Y, Zhang X, Shi J, et al. Targeted delivery of non-viral vectors to cartilage in vivo using a chondrocyte-homing peptide identified by phage display. Biomaterials. 2011;32:6324–6332.
73. Zhao Y, Deng X, Tan S, et al. Co-polymer carrier with dual advantages of cartilage-penetrating and targeting improves delivery and efficacy of MicroRNA treatment of osteoarthritis. Adv Healthc Mater. 2023;12:e2202143. doi:10.1002/adhm.202202143
74. Schofield P, Taylor AI, Rihon J, et al. Characterization of an HNA aptamer suggests a non-canonical G-quadruplex motif. Nucleic Acids Res. 2023;51:7736–7748. doi:10.1093/nar/gkad592
75. Zhu G, Chen X. Aptamer-based targeted therapy. Adv Drug Deliv Rev. 2018;134:65–78. doi:10.1016/j.addr.2018.08.005
76. Chen X, Zhang L, Shao X, et al. Specific clearance of senescent synoviocytes suppresses the development of osteoarthritis based on aptamer-functionalized targeted drug delivery system. Adv Funct Mater. 2022;32:2109460.
77. Corradetti B, Taraballi F, Minardi S, et al. Chondroitin sulfate immobilized on a biomimetic scaffold modulates inflammation while driving chondrogenesis. Stem Cells Transl Med. 2016;5:670–682. doi:10.5966/sctm.2015-0233
78. Sobal G, Dorotka R, Menzel J, Sinzinger H. Uptake studies with chondrotropic 99mTc-chondroitin sulfate in articular cartilage. Implications for imaging osteoarthritis in the knee. Nucl Med Biol. 2013;40:1013–1017. doi:10.1016/j.nucmedbio.2013.07.007
79. Sobal G, Velusamy K, Kosik S, et al. Preclinical evaluation of (99m)Tc labeled chondroitin sulfate for monitoring of cartilage degeneration in osteoarthritis. Nucl Med Biol. 2016;43:339–346. doi:10.1016/j.nucmedbio.2016.02.009
80. Bishnoi M, Jain A, Hurkat P, Jain SK. Aceclofenac-loaded chondroitin sulfate conjugated SLNs for effective management of osteoarthritis. J Drug Target. 2014;22:805–812. doi:10.3109/1061186X.2014.928714
81. Choi KY, Han HS, Lee ES, et al. Hyaluronic acid-based activatable nanomaterials for stimuli-responsive imaging and therapeutics: beyond CD44-mediated drug delivery. Adv Mater. 2019;31:e1803549. doi:10.1002/adma.201803549
82. Lu K-Y, Lin P-Y, Chuang E-Y, et al. H2O2-depleting and O2-generating selenium nanoparticles for fluorescence imaging and photodynamic treatment of proinflammatory-activated macrophages. ACS Appl Mater Interfaces. 2017;9:5158–5172. doi:10.1021/acsami.6b15515
83. Yang G, Fan M, Zhu J, et al. A multifunctional anti-inflammatory drug that can specifically target activated macrophages, massively deplete intracellular H2O2, and produce large amounts CO for a highly efficient treatment of osteoarthritis. Biomaterials. 2020;255:120155. doi:10.1016/j.biomaterials.2020.120155
84. Warmink K, Vinod P, Korthagen NM, Weinans H, Rios JL. Macrophage-driven inflammation in metabolic osteoarthritis: implications for biomarker and therapy development. Int J Mol Sci. 2023;24(7):6112. doi:10.3390/ijms24076112
85. Li H, Yuan Y, Zhang L, et al. Reprogramming macrophage polarization, depleting ROS by astaxanthin and thioketal-containing polymers delivering rapamycin for osteoarthritis treatment. Adv Sci. 2024;11(9):e2305363. doi:10.1002/advs.202305363
86. Zhou F, Mei J, Han X, et al. Kinsenoside attenuates osteoarthritis by repolarizing macrophages through inactivating NF-κB/MAPK signaling and protecting chondrocytes. Acta Pharm Sin B. 2019;9:973–985. doi:10.1016/j.apsb.2019.01.015
87. Zhan M, Sun H, Wang Z, et al. Nanoparticle-mediated multiple modulation of bone microenvironment to tackle osteoarthritis. ACS Nano. 2024;18:10625–10641. doi:10.1021/acsnano.4c00909
88. She P, Bian S, Cheng Y, et al. Dextran sulfate-triamcinolone acetonide conjugate nanoparticles for targeted treatment of osteoarthritis. Int J Biol Macromol. 2020;158:1082–1089. doi:10.1016/j.ijbiomac.2020.05.013
89. Kou L, Huang H, Tang Y, et al. Opsonized nanoparticles target and regulate macrophage polarization for osteoarthritis therapy: a trapping strategy. J Control Release. 2022;347:237–255. doi:10.1016/j.jconrel.2022.04.037
90. Shi T, Zhao J, Long K, et al. Cationic mesoporous silica nanoparticles alleviate osteoarthritis by targeting multiple inflammatory mediators. Biomaterials. 2023;303:122366. doi:10.1016/j.biomaterials.2023.122366
91. Nah Y, Sobha S, Saravanakumar G, et al. Nitric oxide-scavenging hyaluronic acid nanoparticles for osteoarthritis treatment. Biomater Sci. 2024;12(6):1477–1489. doi:10.1039/D3BM01918G
92. Kang C, Jung E, Hyeon H, Seon S, Lee D. Acid-activatable polymeric curcumin nanoparticles as therapeutic agents for osteoarthritis. Nanomedicine. 2020;23:102104. doi:10.1016/j.nano.2019.102104
93. Li X, Wang X, Liu Q, et al. ROS-responsive boronate-stabilized polyphenol-poloxamer 188 assembled dexamethasone nanodrug for macrophage repolarization in osteoarthritis treatment. Adv Healthc Mater. 2021;10:e2100883. doi:10.1002/adhm.202100883
94. Lv Z, Wang P, Li W, et al. Bifunctional TRPV1 targeted magnetothermal switch to attenuate osteoarthritis progression. Research. 2024;7:0316. doi:10.34133/research.0316
95. Zhou F, Mei J, Yang S, et al. Modified ZIF-8 nanoparticles attenuate osteoarthritis by reprogramming the metabolic pathway of synovial macrophages. ACS Appl Mater Interfaces. 2020;12:2009–2022. doi:10.1021/acsami.9b16327
96. Deng Z, Yang C, Xiang T, et al. Gold nanoparticles exhibit anti-osteoarthritic effects via modulating interaction of the “microbiota-gut-joint” axis. J Nanobiotechnol. 2024;22:157. doi:10.1186/s12951-024-02447-y
97. Tong M-J, Song M-X, Liu Z, et al. A bionic thermosensitive sustainable delivery system for reversing the progression of osteoarthritis by remodeling the joint homeostasis. Adv Healthc Mater. 2024;13:e2303792. doi:10.1002/adhm.202303792
98. Yu Y, Kim S-M, Park K, et al. Therapeutic nanodiamonds containing icariin ameliorate the progression of osteoarthritis in rats. Int J Mol Sci. 2023;24(21):15977. doi:10.3390/ijms242115977
99. Chen X, Liu Y, Wen Y, et al. A photothermal-triggered nitric oxide nanogenerator combined with siRNA for precise therapy of osteoarthritis by suppressing macrophage inflammation. Nanoscale. 2019;11:6693–6709. doi:10.1039/C8NR10013F
100. Qin Z, Li X, Wang P, et al. Ultrasmall Prussian blue nanozyme attenuates osteoarthritis by scavenging reactive oxygen species and regulating macrophage phenotype. Nano Lett. 2024;24:11697–11705. doi:10.1021/acs.nanolett.4c03314
101. Pontes-Quero GM, Benito-Garzón L, Pérez Cano J, Aguilar MR, Vázquez-Lasa B. Modulation of inflammatory mediators by polymeric nanoparticles loaded with anti-inflammatory drugs. Pharmaceutics. 2021;13(2):290. doi:10.3390/pharmaceutics13020290
102. Zhen J, Wan T, Sun G, Chen X, Zhang S. A ROS-responsive microsphere capsule encapsulated with NADPH oxidase 4 inhibitor ameliorates macrophage inflammation and ferroptosis. Heliyon. 2024;10:e23589. doi:10.1016/j.heliyon.2023.e23589
103. Lei X, Tan G, Wang Y, et al. Mitochondrial calcium nanoregulators reverse the macrophage proinflammatory phenotype through restoring mitochondrial calcium homeostasis for the treatment of osteoarthritis. Int J Nanomed. 2023;18:1469–1489. doi:10.2147/IJN.S402170
104. Bartlett RL, Sharma S, Panitch A. Cell-penetrating peptides released from thermosensitive nanoparticles suppress pro-inflammatory cytokine response by specifically targeting inflamed cartilage explants. Nanomedicine. 2013;9:419–427. doi:10.1016/j.nano.2012.09.003
105. Wei H, Qin J, Huang Q, et al. Epigallocatechin-3-gallate (EGCG) based metal-polyphenol nanoformulations alleviates chondrocytes inflammation by modulating synovial macrophages polarization. Biomed Pharmacother. 2023;161:114366. doi:10.1016/j.biopha.2023.114366
106. Mota AH, Direito R, Carrasco MP, et al. Combination of hyaluronic acid and PLGA particles as hybrid systems for viscosupplementation in osteoarthritis. Int J Pharm. 2019;559:13–22. doi:10.1016/j.ijpharm.2019.01.017
107. Kang M-L, Kim J-E, Im G-I. Thermoresponsive nanospheres with independent dual drug release profiles for the treatment of osteoarthritis. Acta Biomater. 2016;39:65–78. doi:10.1016/j.actbio.2016.05.005
108. Wu X, Li P, Cheng J, et al. ROS-sensitive nanoparticles co-delivering dexamethasone and CDMP-1 for the treatment of osteoarthritis through chondrogenic differentiation induction and inflammation inhibition. Front Bioeng Biotechnol. 2021;9:608150. doi:10.3389/fbioe.2021.608150
109. Liu S, Zhang C, Zhou Y, et al. MRI-visible mesoporous polydopamine nanoparticles with enhanced antioxidant capacity for osteoarthritis therapy. Biomaterials. 2023;295:122030. doi:10.1016/j.biomaterials.2023.122030
110. Mathiessen A, Conaghan PG. Synovitis in osteoarthritis: current understanding with therapeutic implications. Arthritis Res Ther. 2017;19:18. doi:10.1186/s13075-017-1229-9
111. Han D, Fang Y, Tan X, et al. The emerging role of fibroblast-like synoviocytes-mediated synovitis in osteoarthritis: an update. J Cell mol Med. 2020;24:9518–9532. doi:10.1111/jcmm.15669
112. Deng R-H, Qiu B, Zhou P-H. Chitosan/hyaluronic acid/plasmid-DNA nanoparticles encoding interleukin-1 receptor antagonist attenuate inflammation in synoviocytes induced by interleukin-1 beta. J Mater Sci Mater Med. 2018;29:155. doi:10.1007/s10856-018-6160-3
113. Zhou P-H, Qiu B, Deng R-H, et al. Chondroprotective effects of hyaluronic acid-Chitosan nanoparticles containing plasmid DNA encoding cytokine response modifier A in a rat knee osteoarthritis model. Cell Physiol Biochem. 2018;47(3):1207–1216. doi:10.1159/000490217
114. Jung JH, Kim SE, Kim H-J, et al. A comparative pilot study of oral diacerein and locally treated diacerein-loaded nanoparticles in a model of osteoarthritis. Int J Pharm. 2020;581:119249. doi:10.1016/j.ijpharm.2020.119249
115. Park H, Lee H-R, Shin HJ, et al. p16INK4a-siRNA nanoparticles attenuate cartilage degeneration in osteoarthritis by inhibiting inflammation in fibroblast-like synoviocytes. Biomater Sci. 2022;10:3223–3235. doi:10.1039/D1BM01941D
116. Ren X, Zhuang H, Jiang F, Zhang Y, Zhou P. Ceria nanoparticles alleviated osteoarthritis through attenuating senescence and senescence-associated secretory phenotype in synoviocytes. Int J Mol Sci. 2023;24.
117. Li M, Zhang L, Liu Z, et al. Sanse powder essential oil nanoemulsion negatively regulates TRPA1 by AMPK/mTOR signaling in synovitis: knee osteoarthritis rat model and fibroblast-like synoviocyte isolates. Mediators Inflamm. 2021;2021:4736670. doi:10.1155/2021/4736670
118. Zheng Y, Xiao L, Yu C, et al. Enhanced antiarthritic efficacy by nanoparticles of (-)-epigallocatechin gallate-glucosamine-casein. J Agric Food Chem. 2019;67:6476–6486. doi:10.1021/acs.jafc.9b02075
119. Li H, Guo H, Lei C, et al. Nanotherapy in joints: increasing endogenous hyaluronan production by delivering hyaluronan synthase 2. Adv Mater. 2019;31:e1904535. doi:10.1002/adma.201904535
120. Maudens P, Seemayer CA, Pfefferlé F, Jordan O, Allémann E. Nanocrystals of a potent p38 MAPK inhibitor embedded in microparticles: therapeutic effects in inflammatory and mechanistic murine models of osteoarthritis. J Control Release. 2018;276:102–112. doi:10.1016/j.jconrel.2018.03.007
121. Lu H, Dai Y, Lv L, Zhao H. Chitosan-graft-polyethylenimine/DNA nanoparticles as novel non-viral gene delivery vectors targeting osteoarthritis. PLoS One. 2014;9:e84703. doi:10.1371/journal.pone.0084703
122. Chen H, Chen F, Hu F, et al. MicroRNA-224-5p nanoparticles balance homeostasis via inhibiting cartilage degeneration and synovial inflammation for synergistic alleviation of osteoarthritis. Acta Biomater. 2023;167:401–415. doi:10.1016/j.actbio.2023.06.010
123. Giorgino R, Albano D, Fusco S, et al. Knee osteoarthritis: epidemiology, pathogenesis, and mesenchymal stem cells: what else is new? An update. Int J Mol Sci. 2023;24(7):6405. doi:10.3390/ijms24076405
124. Cao H, Chen M, Cui X, et al. Cell-free osteoarthritis treatment with sustained-release of chondrocyte-targeting exosomes from umbilical cord-derived mesenchymal stem cells to rejuvenate aging chondrocytes. ACS Nano. 2023;17:13358–13376. doi:10.1021/acsnano.3c01612
125. Zhai Q, Chen X, Fei D, et al. Nanorepairers rescue inflammation-induced mitochondrial dysfunction in mesenchymal stem cells. Adv Sci. 2022;9:e2103839. doi:10.1002/advs.202103839
126. Lu Y, Chen J, Li L, et al. Hierarchical functional nanoparticles boost osteoarthritis therapy by utilizing joint-resident mesenchymal stem cells. J Nanobiotechnol. 2022;20:89. doi:10.1186/s12951-022-01297-w
127. Wu C, Huang Z, Chen J, et al. Efficiently directing differentiation and homing of mesenchymal stem cells to boost cartilage repair in osteoarthritis via a nanoparticle and peptide dual-engineering strategy. Biomaterials. 2025;312:122720. doi:10.1016/j.biomaterials.2024.122720
128. Çelik E, Bayram C, Denkbaş EB. Chondrogenesis of human mesenchymal stem cells by microRNA loaded triple polysaccharide nanoparticle system. Mater Sci Eng C Mater Biol Appl. 2019;102:756–763. doi:10.1016/j.msec.2019.05.006
129. Nasiri N, Hosseini S, Reihani-Sabet F, Baghaban Eslaminejad M. Targeted mesenchymal stem cell therapy equipped with a cell-tissue nanomatchmaker attenuates osteoarthritis progression. Sci Rep. 2022;12:4015. doi:10.1038/s41598-022-07969-9
130. Almeida B, Wang Y, Shukla A. Effects of nanoparticle properties on kartogenin delivery and interactions with mesenchymal stem cells. Ann Biomed Eng. 2020;48:2090–2102. doi:10.1007/s10439-019-02430-x
131. Zeng W-N, Zhang Y, Wang D, et al. Intra-articular injection of kartogenin-enhanced bone marrow-derived mesenchymal stem cells in the treatment of knee osteoarthritis in a rat model. Am J Sports Med. 2021;49:2795–2809. doi:10.1177/03635465211023183
132. Hu Q, Ding B, Yan X, et al. Polyethylene glycol modified PAMAM dendrimer delivery of kartogenin to induce chondrogenic differentiation of mesenchymal stem cells. Nanomedicine. 2017;13:2189–2198. doi:10.1016/j.nano.2017.05.011
133. Zong L, Wang Q, Sun H, et al. Intra-articular injection of PLGA/Polydopamine core-shell nanoparticle attenuates osteoarthritis progression. ACS Appl Mater Interfaces. 2024;16:21450–21462. doi:10.1021/acsami.3c18464
134. Jiang Z, Zhang Z, Li S, Lin S, Yuan H. Magnetically guided intracartilaginous delivery of kartogenin improves stem cell-targeted degenerative arthritis therapy. Int J Nanomed. 2022;17:5511–5524. doi:10.2147/IJN.S381815
135. Kang ML, Ko J-Y, Kim JE, Im G-I. Intra-articular delivery of kartogenin-conjugated chitosan nano/microparticles for cartilage regeneration. Biomaterials. 2014;35:9984–9994. doi:10.1016/j.biomaterials.2014.08.042
136. Valenti MT, Mottes M, Biotti A, et al. Clodronate as a therapeutic strategy against osteoarthritis. Int J Mol Sci. 2017;18(12):2696. doi:10.3390/ijms18122696
137. Deng C, Zhou Q, Zhang M, et al. Bioceramic scaffolds with antioxidative functions for ROS scavenging and osteochondral regeneration. Adv Sci. 2022;9:e2105727. doi:10.1002/advs.202105727
138. Wang P, Zhao Z, Li Z, et al. Attenuation of osteoarthritis progression via locoregional delivery of Klotho-expressing plasmid DNA and Tanshinon IIA through a stem cell-homing hydrogel. J Nanobiotechnol. 2024;22:325. doi:10.1186/s12951-024-02608-z
139. Ma J-C, Luo T, Feng B, et al. Exploring the translational potential of PLGA nanoparticles for intra-articular rapamycin delivery in osteoarthritis therapy. J Nanobiotechnol. 2023;21:361. doi:10.1186/s12951-023-02118-4
140. Mehana E-SE, Khafaga AF, El-Blehi SS. The role of matrix metalloproteinases in osteoarthritis pathogenesis: an updated review. Life Sci. 2019;234:116786. doi:10.1016/j.lfs.2019.116786
141. Chow YY, Chin K-Y. The Role of Inflammation in the Pathogenesis of Osteoarthritis. Mediators Inflamm. 2020;2020:8293921. doi:10.1155/2020/8293921
142. Wang J, Zhang L, Zhu J, et al. Hyaluronic acid modified curcumin-loaded chitosan nanoparticles inhibit chondrocyte apoptosis to attenuate osteoarthritis via upregulation of activator protein 1 and RUNX family transcription factor 2. J Biomed Nanotechnol. 2022;18(1):144–157. doi:10.1166/jbn.2022.3193
143. Guo J, Su K, Wang L, et al. Poly(p-coumaric acid) nanoparticles alleviate temporomandibular joint osteoarthritis by inhibiting chondrocyte ferroptosis. Bioact Mater. 2024;40:212–226. doi:10.1016/j.bioactmat.2024.06.007
144. Li Y, Zhu S, Luo J, et al. The protective effect of selenium nanoparticles in osteoarthritis: in vitro and in vivo studies. Drug Des Devel Ther. 2023;17:1515–1529. doi:10.2147/DDDT.S407122
145. Hu W, Yao X, Li Y, et al. Injectable hydrogel with selenium nanoparticles delivery for sustained glutathione peroxidase activation and enhanced osteoarthritis therapeutics. Mater Today Bio. 2023;23:100864. doi:10.1016/j.mtbio.2023.100864
146. Song J, Kim EH, Yang J-H, et al. Upregulated FOXM1 stimulates chondrocyte senescence in Acot12-/-Nudt7-/- double knockout mice. Theranostics. 2023;13:5207–5222. doi:10.7150/thno.89033
147. Jiang Y, Li T, Yang J, et al. Sustained intra-articular reactive oxygen species scavenging and alleviation of osteoarthritis by biocompatible amino-modified tantalum nanoparticles. Front Bioeng Biotechnol. 2023;11:1118850. doi:10.3389/fbioe.2023.1118850
148. Wang X, Cai Y, Wu C, et al. Conversion of senescent cartilage into a pro-chondrogenic microenvironment with antibody-functionalized copper sulfate nanoparticles for efficient osteoarthritis therapy. J Nanobiotechnol. 2023;21:258. doi:10.1186/s12951-023-02036-5
149. Liu Y, Yao J, Deng G, et al. Microgel encapsulated nanoparticles for intra-articular disulfiram delivery to treat osteoarthritis. mol Pharm. 2024;21(1):87–101. doi:10.1021/acs.molpharmaceut.3c00462
150. Chen Q, Jin Y, Chen T, et al. Injectable nanocomposite hydrogels with enhanced lubrication and antioxidant properties for the treatment of osteoarthritis. Mater Today Bio. 2024;25:100993. doi:10.1016/j.mtbio.2024.100993
151. Moghadam NA, Bagheri F, Eslaminejad MB. Chondroitin sulfate modified chitosan nanoparticles as an efficient and targeted gene delivery vehicle to chondrocytes. Colloids Surf B Biointerfaces. 2022;219:112786. doi:10.1016/j.colsurfb.2022.112786
152. Wu H, Wang J, Lin Y, et al. Injectable ozone-rich nanocomposite hydrogel loaded with D-Mannose for anti-inflammatory and cartilage protection in osteoarthritis treatment. Small. 2024;20:e2309597. doi:10.1002/smll.202309597
153. Kang L-J, Yoon J, Rho JG, et al. Self-assembled hyaluronic acid nanoparticles for osteoarthritis treatment. Biomaterials. 2021;275:120967. doi:10.1016/j.biomaterials.2021.120967
154. Wang J, Wang X, Cao Y, et al. Therapeutic potential of hyaluronic acid/chitosan nanoparticles for the delivery of curcuminoid in knee osteoarthritis and an in vitro evaluation in chondrocytes. Int J Mol Med. 2018;42:2604–2614. doi:10.3892/ijmm.2018.3817
155. Kondreddy V, Banerjee R, Devi BLAP, Muralidharan K, Piramanayagam S. Inhibition of the MALT1-LPCAT3 axis protects cartilage degeneration and osteoarthritis. Cell Commun Signal. 2024;22:189. doi:10.1186/s12964-024-01547-4
156. Zhang Z, Leong DJ, Xu L, et al. Curcumin slows osteoarthritis progression and relieves osteoarthritis-associated pain symptoms in a post-traumatic osteoarthritis mouse model. Arthritis Res Ther. 2016;18:128. doi:10.1186/s13075-016-1025-y
157. Shin HJ, Park H, Shin N, et al. p47phox siRNA-loaded PLGA nanoparticles suppress ROS/oxidative stress-induced chondrocyte damage in osteoarthritis. Polymers. 2020;12(2):443. doi:10.3390/polym12020443
158. Xiong L, Bao H, Li S, et al. Cerium oxide nanoparticles protect against chondrocytes and cartilage explants from oxidative stress via Nrf2/HO-1 pathway in temporomandibular joint osteoarthritis. Front Bioeng Biotechnol. 2023;11:1076240. doi:10.3389/fbioe.2023.1076240
159. Wei L, Pan Q, Teng J, Zhang H, Qin N. Intra-articular administration of PLGA resveratrol sustained-release nanoparticles attenuates the development of rat osteoarthritis. Mater Today Bio. 2024;24:100884. doi:10.1016/j.mtbio.2023.100884
160. Kim SE, Choi SJ, Park K, et al. Intra-articular injection of rebamipide-loaded nanoparticles attenuate disease progression and joint destruction in osteoarthritis rat model: a pilot study. Cartilage. 2022;13(1):19476035211069250. doi:10.1177/19476035211069250
161. Chang M-C, Chiang P-F, Kuo Y-J, et al. Hyaluronan-loaded liposomal dexamethasone-diclofenac nanoparticles for local osteoarthritis treatment. Int J Mol Sci. 2021;22.
162. Zerrillo L, Que I, Vepris O, et al. pH-responsive poly(lactide-co-glycolide) nanoparticles containing near-infrared dye for visualization and hyaluronic acid for treatment of osteoarthritis. J Control Release. 2019;309:265–276. doi:10.1016/j.jconrel.2019.07.031
163. Li W, Lv Z, Wang P, et al. Near infrared responsive gold nanorods attenuate osteoarthritis progression by targeting TRPV1. Adv Sci. 2024;11:e2307683. doi:10.1002/advs.202307683
164. Valentino A, Conte R, De Luca I, et al. Thermo-responsive gel containing hydroxytyrosol-chitosan nanoparticles (Hyt@tgel) counteracts the increase of osteoarthritis biomarkers in human chondrocytes. Antioxidants. 2022;11. doi:10.3390/antiox11061210
165. Shin HJ, Park H, Shin N, et al. p66shc siRNA nanoparticles ameliorate chondrocytic mitochondrial dysfunction in osteoarthritis. Int J Nanomed. 2020;15:2379–2390. doi:10.2147/IJN.S234198
166. Nabizadeh Z, Nasrollahzadeh M, Kruppke B, Nasrabadi D. A combination of chitosan nanoparticles loaded with celecoxib and kartogenin has anti-inflammatory and chondroprotective effects: results from an in vitro model of osteoarthritis. Heliyon. 2024;10:e31058. doi:10.1016/j.heliyon.2024.e31058
167. Gui T, Wei Y, Luo L, et al. Activating EGFR signaling attenuates osteoarthritis development following loading injury in mice. J Bone Miner Res. 2022;37:2498–2511. doi:10.1002/jbmr.4717
168. Sheng W, Li A, Yue Y, et al. A novel curcumin-loaded nanoplatform alleviates osteoarthritis by inhibiting chondrocyte ferroptosis. Macromol Rapid Commun. 2024;46:e2400495. doi:10.1002/marc.202400495
169. Chen J, Xu W, Dai T, et al. Pioglitazone-loaded cartilage-targeted nanomicelles (Pio@C-HA-DOs) for osteoarthritis treatment. Int J Nanomed. 2023;18:5871–5890. doi:10.2147/IJN.S428938
170. Xie C, Sun Q, Dong Y, et al. Calcitriol-loaded multifunctional nanospheres with superlubricity for advanced osteoarthritis treatment. ACS Nano. 2023;17:12842–12861. doi:10.1021/acsnano.3c04241
171. Zhou T, Ran J, Xu P, et al. A hyaluronic acid/platelet-rich plasma hydrogel containing MnO2 nanozymes efficiently alleviates osteoarthritis in vivo. Carbohydr Polym. 2022;292:119667. doi:10.1016/j.carbpol.2022.119667
172. Fan W, Li J, Yuan L, et al. Intra-articular injection of kartogenin-conjugated polyurethane nanoparticles attenuates the progression of osteoarthritis. Drug Deliv. 2018;25:1004–1012. doi:10.1080/10717544.2018.1461279
173. Nabizadeh Z, Nasrollahzadeh M, Shabani AA, Mirmohammadkhani M, Nasrabadi D. Evaluation of the anti-inflammatory activity of fisetin-loaded nanoparticles in an in vitro model of osteoarthritis. Sci Rep. 2023;13:15494. doi:10.1038/s41598-023-42844-1
174. Conte R, Finicelli M, Borrone A, et al. MMP-2 silencing through siRNA loaded positively-charged nanoparticles (AcPEI-NPs) counteracts chondrocyte de-differentiation. Polymers. 2023;15(5):1172. doi:10.3390/polym15051172
175. Jin Z, Zhan Y, Zheng L, et al. Cannabidiol-loaded poly lactic-co-glycolic acid nanoparticles with improved bioavailability as a potential for osteoarthritis therapeutic. Chembiochem. 2023;24(9):e202200698. doi:10.1002/cbic.202200698
176. Cullier A, Cassé F, Manivong S, et al. Functionalized nanogels with endothelin-1 and bradykinin receptor antagonist peptides decrease inflammatory and cartilage degradation markers of osteoarthritis in a horse organoid model of cartilage. Int J Mol Sci. 2022;23(16):8949. doi:10.3390/ijms23168949
177. Pi Y, Zhang X, Shao Z, et al. Intra-articular delivery of anti-Hif-2α siRNA by chondrocyte-homing nanoparticles to prevent cartilage degeneration in arthritic mice. Gene Ther. 2015;22(6):439–448. doi:10.1038/gt.2015.16
178. Jin T, Wu D, Liu X-M, et al. Intra-articular delivery of celastrol by hollow mesoporous silica nanoparticles for pH-sensitive anti-inflammatory therapy against knee osteoarthritis. J Nanobiotechnol. 2020;18:94. doi:10.1186/s12951-020-00651-0
179. Pan J, Cai Y, Zhang C, Xu S. Intra-articular delivery of geraniol encapsulated by pH/redox-responsive nanogel ameliorates osteoarthritis by regulating oxidative stress and inflammation. J Mol Histol. 2023;54:579–591. doi:10.1007/s10735-023-10163-4
180. Geiger BC, Wang S, Padera RF, Grodzinsky AJ, Hammond PT. Cartilage-penetrating nanocarriers improve delivery and efficacy of growth factor treatment of osteoarthritis. Sci Transl Med. 2018;10. doi:10.1126/scitranslmed.aat8800
181. Lin Y-W, Fang C-H, Meng F-Q, Ke C-J, Lin F-H. Hyaluronic acid loaded with cerium oxide nanoparticles as antioxidant in hydrogen peroxide induced chondrocytes injury: an in vitro osteoarthritis model. Molecules. 2020;25(19):4407. doi:10.3390/molecules25194407
182. Wang Z, Yin X, Zhuang C, et al. Injectable regenerated silk fibroin micro/nanosphere with enhanced permeability and stability for osteoarthritis therapy. Small. 2024;20(46):e2405049. doi:10.1002/smll.202405049
183. Li T, Yang J, Weng C, et al. Intra-articular injection of anti-inflammatory peptide-loaded glycol chitosan/fucoidan nanogels to inhibit inflammation and attenuate osteoarthritis progression. Int J Biol Macromol. 2021;170:469–478. doi:10.1016/j.ijbiomac.2020.12.158
184. Dwivedi P, Nayak V, Kowshik M. Role of gold nanoparticles as drug delivery vehicles for chondroitin sulfate in the treatment of osteoarthritis. Biotechnol Prog. 2015;31:1416–1422. doi:10.1002/btpr.2147
185. Liu P, Gu L, Ren L, et al. Intra-articular injection of etoricoxib-loaded PLGA-PEG-PLGA triblock copolymeric nanoparticles attenuates osteoarthritis progression. Am J Transl Res. 2019;11:6775–6789.
186. Cao F, Liu Y, Gao Y, et al. CKIP-1-loaded cartilage-affinitive nanoliposomes reverse osteoarthritis by restoring chondrocyte homeostasis. ACS Biomater Sci Eng. 2024;10:4437–4451. doi:10.1021/acsbiomaterials.4c00222
187. Zhang K, Yang J, Sun Y, et al. Thermo-sensitive dual-functional nanospheres with enhanced lubrication and drug delivery for the treatment of osteoarthritis. Chemistry. 2020;26:10564–10574. doi:10.1002/chem.202001372
188. Ji M-L, Jiang H, Wu F, et al. Precise targeting of miR-141/200c cluster in chondrocytes attenuates osteoarthritis development. Ann Rheum Dis. 2021;80:356–366. doi:10.1136/annrheumdis-2020-218469
189. Kan T, Tian Z, Sun L, et al. Quercetin-loaded zeolitic imidazolate framework-8 (ZIF-8) nanoparticles attenuate osteoarthritis by activating autophagy via the Pi3k/Akt signaling. ACS Appl Mater Interfaces. 2024;16:40444–40454. doi:10.1021/acsami.4c04649
190. Zhou Y, Liu S-Q, Peng H, et al. In vivo anti-apoptosis activity of novel berberine-loaded chitosan nanoparticles effectively ameliorates osteoarthritis. Int Immunopharmacol. 2015;28(1):34–43. doi:10.1016/j.intimp.2015.05.014
191. Deloney M, Smart K, Christiansen BA, Panitch A. Thermoresponsive, hollow, degradable core-shell nanoparticles for intra-articular delivery of anti-inflammatory peptide. J Control Release. 2020;323:47–58. doi:10.1016/j.jconrel.2020.04.007
192. Kumar S, Adjei IM, Brown SB, Liseth O, Sharma B. Manganese dioxide nanoparticles protect cartilage from inflammation-induced oxidative stress. Biomaterials. 2019;224:119467. doi:10.1016/j.biomaterials.2019.119467
193. Mei H, Sha C, Lv Q, et al. Multifunctional polymeric nanocapsules with enhanced cartilage penetration and retention for osteoarthritis treatment. J Control Release. 2024;374:466–477. doi:10.1016/j.jconrel.2024.08.031
194. Jiang Z, Wang H, Zhang Z, Pan J, Yuan H. Cartilage targeting therapy with reactive oxygen species-responsive nanocarrier for osteoarthritis. J Nanobiotechnol. 2022;20:419. doi:10.1186/s12951-022-01629-w
195. Zhao X, Huang H, Jiang X, et al. Supramolecular nanoparticle loaded with bilirubin enhances cartilage protection and alleviates osteoarthritis via modulating oxidative stress and inflammatory responses. Colloids Surf B Biointerfaces. 2024;245:114243. doi:10.1016/j.colsurfb.2024.114243
196. Ouyang Z, Tan T, Liu C, et al. Targeted delivery of hesperetin to cartilage attenuates osteoarthritis by bimodal imaging with Gd2(CO3)3@PDA nanoparticles via TLR-2/NF-κB/Akt signaling. Biomaterials. 2019;205:50–63. doi:10.1016/j.biomaterials.2019.03.018
197. Yan H, Hu Y, Akk A, et al. Induction of WNT16 via peptide-mRNA nanoparticle-based delivery maintains cartilage homeostasis. Pharmaceutics. 2020;12(1):73. doi:10.3390/pharmaceutics12010073
198. Wu J, Qin Z, Jiang X, et al. ROS-responsive PPGF nanofiber membrane as a drug delivery system for long-term drug release in attenuation of osteoarthritis. NPJ Regen Med. 2022;7:66. doi:10.1038/s41536-022-00254-3
199. Yu H, Ren P, Pan X, et al. Intracellular delivery of itaconate by metal-organic framework-anchored hydrogel microspheres for osteoarthritis therapy. Pharmaceutics. 2023;15:724. doi:10.3390/pharmaceutics15030724
200. Hu H, Yang J, Zhong Y, et al. Polydopamine-Pd nanozymes as potent ROS scavengers in combination with near-infrared irradiation for osteoarthritis treatment. iScience. 2023;26:106605. doi:10.1016/j.isci.2023.106605
201. Yan H, Duan X, Pan H, et al. Suppression of NF-κB activity via nanoparticle-based siRNA delivery alters early cartilage responses to injury. Proc Natl Acad Sci U S A. 2016;113:E6199–E6208. doi:10.1073/pnas.1608245113
202. He M, Qin Z, Liang X, et al. A pH-responsive mesoporous silica nanoparticles-based drug delivery system with controlled release of andrographolide for OA treatment. Regen Biomater. 2021;8:rbab020. doi:10.1093/rb/rbab020
203. Karim A, Qaisar R, Suresh S, Jagal J, Rawas-Qalaji M. Nanoparticle-delivered quercetin exhibits enhanced efficacy in eliminating iron-overloaded senescent chondrocytes. Nanomedicine. 2024;19(26):2159–2170. doi:10.1080/17435889.2024.2393074
204. Duan X, Cai L, Pham CTN, et al. Amelioration of posttraumatic osteoarthritis in mice using intraarticular silencing of periostin via nanoparticle-based small interfering RNA. Arthritis Rheumatol. 2021;73:2249–2260. doi:10.1002/art.41794
205. Ebada HMK, Nasra MMA, Nassra RA, Abdallah OY. Chondroitin sulfate-functionalized lipid nanoreservoirs: a novel cartilage-targeting approach for intra-articular delivery of cassic acid for osteoarthritis treatment. Drug Deliv. 2022;29:652–663. doi:10.1080/10717544.2022.2041130
206. Tajik E, Vaezi Z, Tabarsa M, Hekmat A, Naderi-Manesh H. Grafting of sinapic acid onto glucosamine nanoparticle as a potential therapeutic drug with enhanced anti-inflammatory activities in osteoarthritis treatment. Int J Biol Macromol. 2023;253:127454. doi:10.1016/j.ijbiomac.2023.127454
207. Gong X, Wang F, Huang Y, et al. Magnetic-targeting of polyethylenimine-wrapped iron oxide nanoparticle labeled chondrocytes in a rabbit articular cartilage defect model. RSC Adv. 2018;8:7633–7640. doi:10.1039/C7RA12039G
208. Gao X, Ma Y, Zhang G, et al. Targeted elimination of intracellular reactive oxygen species using nanoparticle-like chitosan- superoxide dismutase conjugate for treatment of monoiodoacetate-induced osteoarthritis. Int J Pharm. 2020;590:119947. doi:10.1016/j.ijpharm.2020.119947
209. Pontes AP, Welting TJM, Rip J, Creemers LB. Polymeric nanoparticles for drug delivery in osteoarthritis. Pharmaceutics. 2022;14:2639. doi:10.3390/pharmaceutics14122639
210. Deng R, Zhao R, Zhang Z, et al. Chondrocyte membrane-coated nanoparticles promote drug retention and halt cartilage damage in rat and canine osteoarthritis. Sci Transl Med. 2024;16:eadh9751. doi:10.1126/scitranslmed.adh9751
211. Hu S, Hu Y, Yan W. Extracellular vesicle-mediated interorgan communication in metabolic diseases. Trends Endocrinol Metab. 2023;34:571–582. doi:10.1016/j.tem.2023.06.002
212. Zhang L, Chen X, Cai P, et al. Reprogramming mitochondrial metabolism in synovial macrophages of early osteoarthritis by a camouflaged meta-defensome. Adv Mater. 2022;34:e2202715. doi:10.1002/adma.202202715
213. Zhang X, Chen J, Jiang Q, et al. Highly biosafe biomimetic stem cell membrane-disguised nanovehicles for cartilage regeneration. J Mater Chem B. 2020;8:8884–8893. doi:10.1039/D0TB01686A
214. D’Atri D, Zerrillo L, Garcia J, et al. Nanoghosts: mesenchymal stem cells derived nanoparticles as a unique approach for cartilage regeneration. J Control Release. 2021;337:472–481. doi:10.1016/j.jconrel.2021.05.015
215. Cai Y, Wu C, Ou Q, et al. Enhanced osteoarthritis therapy by nanoengineered mesenchymal stem cells using biomimetic CuS nanoparticles loaded with plasmid DNA encoding TGF-β1. Bioact Mater. 2023;19:444–457. doi:10.1016/j.bioactmat.2022.04.021
216. Deng C, Li Z, Lu L, et al. Sophisticated magneto-mechanical actuation promotes in situ stem cell assembly and chondrogenesis for treating osteoarthritis. ACS Nano. 2023;17:21690–21707. doi:10.1021/acsnano.3c06909
217. Zhang X, Sun Y, Chen W, et al. Nanoparticle functionalization with genetically-engineered mesenchymal stem cell membrane for targeted drug delivery and enhanced cartilage protection. Biomater Adv. 2022;136:212802. doi:10.1016/j.bioadv.2022.212802
218. Chen G, Cui L, Luo P, et al. A reactive oxygen species “sweeper” based on hollow mesopore cerium oxide nanospheres for targeted and anti-inflammatory management of osteoarthritis. ACS Appl Mater Interfaces. 2024;16:34705–34719. doi:10.1021/acsami.4c06231
219. Yang L, Li W, Zhao Y, Shang L. Magnetic polysaccharide mesenchymal stem cells exosomes delivery microcarriers for synergistic therapy of osteoarthritis. ACS Nano. 2024.
220. Cao H, Li W, Zhang H, et al. Bio-nanoparticles loaded with synovial-derived exosomes ameliorate osteoarthritis progression by modifying the oxidative microenvironment. J Nanobiotechnol. 2024;22:271. doi:10.1186/s12951-024-02538-w
221. Duan H, Liu Y, Gao Z, Huang W. Recent advances in drug delivery systems for targeting cancer stem cells. Acta Pharm Sin B. 2021;11:55–70. doi:10.1016/j.apsb.2020.09.016
222. Manferdini C, Paolella F, Gabusi E, et al. Adipose stromal cells mediated switching of the pro-inflammatory profile of M1-like macrophages is facilitated by PGE2: in vitro evaluation. Osteoarthritis Cartilage. 2017;25:1161–1171. doi:10.1016/j.joca.2017.01.011
223. Ma Y, Yang H, Zong X, et al. Artificial M2 macrophages for disease-modifying osteoarthritis therapeutics. Biomaterials. 2021;274:120865. doi:10.1016/j.biomaterials.2021.120865
224. van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev mol Cell Biol. 2018;19:213–228. doi:10.1038/nrm.2017.125
225. Colombini A, Ragni E, Mortati L, et al. Adipose-derived mesenchymal stromal cells treated with interleukin 1 beta produced chondro-protective vesicles able to fast penetrate in cartilage. Cells. 2021;10(5):1180. doi:10.3390/cells10051180
226. Barteneva NS, Maltsev N, Vorobjev IA. Microvesicles and intercellular communication in the context of parasitism. Front Cell Infect Microbiol. 2013;3:49. doi:10.3389/fcimb.2013.00049
227. Paolicelli RC, Bergamini G, Rajendran L. Cell-to-cell communication by extracellular vesicles: focus on microglia. Neuroscience. 2019;405:148–157. doi:10.1016/j.neuroscience.2018.04.003
228. Pape E, Pinzano A, Henrionnet C, et al. Rat synovial tissue and blood rapamycin pharmacokinetics after intra-articular injection of free solution or nanoparticles vs free rapamycin intravenous shot. Int J Pharm. 2022;624:122026. doi:10.1016/j.ijpharm.2022.122026
229. Mei X, Villamagna IJ, Nguyen T, et al. Polymer particles for the intra-articular delivery of drugs to treat osteoarthritis. Biomed Mater. 2021;16(4):042006. doi:10.1088/1748-605X/abee62
230. Jin G-Z. Current nanoparticle-based technologies for osteoarthritis therapy. Nanomaterials. 2020;10(12):2368. doi:10.3390/nano10122368
231. Manivong S, Cullier A, Audigié F, et al. New trends for osteoarthritis: biomaterials, models and modeling. Drug Discov Today. 2023;28:103488. doi:10.1016/j.drudis.2023.103488
232. Naeem M, Hoque MZ, Ovais M, Basheer C, Ahmad I. Stimulus-responsive smart nanoparticles-based CRISPR-cas delivery for therapeutic genome editing. Int J Mol Sci. 2021;22(20):11300. doi:10.3390/ijms222011300
233. Habibi N, Quevedo DF, Gregory JV, Lahann J. Emerging methods in therapeutics using multifunctional nanoparticles. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2020;12:e1625. doi:10.1002/wnan.1625
234. Shen Q, Huang W, Qiu Y, et al. Bergapten exerts a chondroprotective effect in temporomandibular joint osteoarthritis by combining intestinal flora alteration and reactive oxygen species reduction. Biomed Pharmacother. 2023;167:115525. doi:10.1016/j.biopha.2023.115525
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 4.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.
Recommended articles


The Level of Histone Deacetylase 4 is Associated with Aging Cartilage Degeneration and Chondrocyte Hypertrophy
Dong Z, Ma Z, Yang M, Cong L, Zhao R, Cheng L, Sun J, Wang Y, Yang R, Wei X, Li P
Journal of Inflammation Research 2022, 15:3547-3560
Published Date: 16 June 2022

A Tissue-Tended Mycophenolate-Modified Nanoparticle Alleviates Systemic Lupus Erythematosus in MRL/Lpr Mouse Model Mainly by Promoting Local M2-Like Macrophagocytes Polarization
Jiang B, Zhang Y, Li Y, Chen Y, Sha S, Zhao L, Li D, Wen J, Lan J, Lou Y, Su H, Zhang C, Zhu J, Tao J
International Journal of Nanomedicine 2022, 17:3251-3267
Published Date: 28 July 2022

Recent Advances in Reactive Oxygen Species (ROS)-Responsive Polyfunctional Nanosystems 3.0 for the Treatment of Osteoarthritis
Ding DF, Xue Y, Wu XC, Zhu ZH, Ding JY, Song YJ, Xu XL, Xu JG
Journal of Inflammation Research 2022, 15:5009-5026
Published Date: 31 August 2022

Effects of Immune Cells and Cytokines on Different Cells in OA
Luo P, Yuan Q, Wan X, Yang M, Xu P
Journal of Inflammation Research 2023, 16:2329-2343
Published Date: 30 May 2023

Current and Novel Therapeutics for Articular Cartilage Repair and Regeneration
Cong B, Sun T, Zhao Y, Chen M
Therapeutics and Clinical Risk Management 2023, 19:485-502
Published Date: 20 June 2023