International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Degenerative joint diseases include osteoarthritis (OA), intervertebral disc (IVD) degeneration (IVDD) and rheumatoid arthritis (RA). The gradual structural and functional deterioration of articular cartilage, subchondral bone and surrounding joint tissues are common features, which lead to joint pain, stiffness and functional impairment (1-3). It is reported that degenerative joint diseases affect >250 million individuals worldwide (4). In OA, progressive degradation of the cartilage matrix occurs, with notable reductions in the main components of the extracellular matrix (ECM), such as type II collagen and proteoglycans; in later stages, osteophytes (which are outgrowths of bone at joint margins that develop in response to cartilage loss) often form, and joint deformities may develop (2,5). In IVDD, the nucleus pulposus (NP) cells and annulus fibrosus cells progressively lose their viability and function; over time, the intervertebral discs become dehydrated, collapse and deform, causing various clinical issues such as lower back pain (6). Furthermore, in RA, an autoimmune disease, chronic inflammation and synovial hyperplasia erode the articular cartilage and subchondral bone, typically manifesting as marked joint swelling, morning stiffness and cartilage destruction (7,8). Regardless of the specific form, inflammation and oxidative stress both serve roles in the pathogenesis of these diseases.
Oxidative stress is the excessive accumulation of reactive oxygen species (ROS) and reactive nitrogen species that surpass the scavenging capacity of endogenous antioxidant systems (such as superoxide dismutase, catalase, glutathione peroxidase and peroxiredoxins). Oxidative stress causes cellular damage and can lead to apoptosis or necrosis (9,10). In tissues such as articular cartilage and intervertebral discs, oxidative stress induces the expression of matrix-degrading enzymes [such as matrix metalloproteinases (MMPs; MMP-1, -3 and -13) and aggrecanases (such as a disintegrin and metalloproteinase with thrombospondin motifs-4 and -5)], which accelerates the degradation of the ECM (2,11,12). Moreover, oxidative stress can activate associated signaling pathways (such as the NLRP3 inflammasome), exacerbating local inflammation (13,14). With aging or disease progression, the capacity of joint cells to defend against free radical damage declines. This results in an increased likelihood that oxidative damage will accumulate, which further exacerbates joint or disc degeneration (2,15).
Mitochondria, the organelles responsible for oxidative phosphorylation and energy supply in cells, are the primary sites of endogenous ROS production (1,16). During oxidative phosphorylation in the mitochondrial electron transport chain, superoxide anions (O2-) are generated and then converted to H2O2, OH·, singlet oxygen and peroxynitrite by antioxidant systems such as superoxide dismutases (SODs; such as SOD2) (17). If the levels of the O2- exceeds this regulatory capacity, damage on the mitochondrial proteins, membrane lipids and DNA may occur (16). Previous studies reveal that mitochondrial dysfunction and excess mitochondrial ROS production are involved in numerous degenerative diseases, including OA, RA and IVDD (5,8,18). Therefore, protecting mitochondria and improving redox homeostasis is a notable research focus and may be a strategy in the prevention and treatment of degenerative joint diseases.
Mitochondrial oxidative stress results in tissue damage in degenerative joint diseases; however, therapeutic strategies aimed at restoring the mitochondrial redox balance are scarce. Although conventional antioxidants (such as vitamin C, vitamin E and N-acetylcysteine) and anti-inflammatory drugs (for example, non-steroidal anti-inflammatory drugs such as ibuprofen and indomethacin, or corticosteroids such as prednisone) can transiently alleviate symptoms, they cannot mitigate the mitochondria-derived ROS at the source (2). Despite growing evidence that aberrant mitochondrial ROS contribute to cartilage degradation, NP cell senescence and synovial inflammation, only a small number of therapeutic strategies target mitochondrial oxidative stress. At present, the existing antioxidant approaches rely on non-mitochondria-specific compounds, such as vitamin C, vitamin E, N-acetylcysteine, glutathione and polyphenols (for example, resveratrol), and clinical translation is limited (2). Therefore, an evaluation of the mitochondria-targeted antioxidants and quality-control mechanisms was needed in order to assess this translational gap and highlight the novel disease-modifying treatments for OA, IVDD and RA.
A systematic search of PubMed (https://pubmed.ncbi.nlm.nih.gov/), Web of Science (https://www.webofscience.com/) and the Cochrane Library (https://www.cochranelibrary.com/) was carried out for articles published between 2000 and June 2025. Search terms included combinations of ‘osteoarthritis’ OR ‘intervertebral disc degeneration’ OR ‘rheumatoid arthritis’ with ‘mitochondria’, ‘oxidative stress’, ‘mitophagy’, ‘MitoQ’, ‘SS-31’ and ‘mitochondrial antioxidant’. Boolean operators and Medical Subject Headings were used as appropriate. Reference lists of relevant reviews and clinical guidelines were also reviewed to identify additional studies.
The inclusion criteria used were as follows: i) Primary research or review articles written in English; ii) in vitro, in vivo or ex vivo models of OA, IVDD or RA; iii) interventions that directly targeted mitochondrial function or oxidative stress; and iv) quantifiable outcomes regarding the levels of ROS, cell survival, ECM integrity or functional scores.
The exclusion criteria used were as follows: i) Publications not written in English; ii) conference abstracts or editorials; iii) duplicate reports; or iv) studies with insufficient methodological detail.
Within mitochondria, 1-5% of the oxygen taken in by the cell may ‘leak’ in the form of ROS during the activity of the electron transport chain (19,20). Under normal conditions, SODs located in the mitochondria (such as SOD2, which is primarily located in the mitochondrial matrix), peroxiredoxins and glutathione peroxidases convert or remove these ROS, which keeps the levels of the ROS within a reasonable range (typically at nM to low µM concentrations, such as superoxide in the 10-100 nM range and hydrogen peroxide in the 1-10 µM range, which act as signaling molecules) and allows them to function in cell signal transduction and normal metabolic processes, such as MAPK (ERK, JNK and p38), NF-κB, PI3K-Akt, hypoxia-inducible factor-1α and Wnt/β-catenin signaling pathways (2,21) (Fig. 1). However, when mitochondrial function is impaired (due to exogenous harmful stimuli, aging or gene mutations that inactivate complexes involved in the electron transport chain) excessive levels of ROS can accumulate, resulting in lipid peroxidation, oxidative protein modifications and breaks in the DNA (16,22).
Mitochondrial dysfunction is associated with degenerative changes in joint tissues by promoting apoptosis, pyroptosis and inflammatory responses, as well as undermining cartilage matrix integrity and normal cellular functions (23,24). The excessive generation of mitochondrial ROS and the loss of the mitochondrial membrane potential can cause a release of cytochrome c. This can lead to the subsequent activation of the caspase cascade, which induces apoptosis (13,25). Furthermore, elevated ROS levels can promote the assembly of the NLRP3 inflammasome, triggering pyroptosis and increasing the inflammatory damage within joint tissues (13,26).
In addition to the aforementioned direct mechanisms of cell death, oxidative stress activates inflammatory pathways such as NF-κB and MAPK. These inflammatory pathways increase the production of proinflammatory factors (such as IL-1β and TNF-α) and MMPs, which together accelerate cartilage matrix degradation (7,27).
As mitochondrial damage worsens, cells face an energy deficit. An overabundance of ROS further impairs proteoglycans and collagen, increasing the breakdown of the ECM in the cartilage (15,28). For example, in RA, mitochondrial dysfunction in fibroblast-like synoviocytes increases both local inflammation and synovial hyperplasia (7). Moreover, the accumulation of ROS may lead to cell cycle arrest, reduced chondrocyte proliferation and the emergence of cellular senescence phenotypes (such as increased p16 and p21 expression levels), which are observed in conditions such as osteoporosis, OA and IVDD (29-31).
MitoQ is a mitochondria-targeted antioxidant formed by conjugating a ubiquinone analog with triphenylphosphonium (TPP) through an alkyl chain. It demonstrates certain protective effects in models of both OA and IVDD (2,32) (Table I).
In vitro studies, using chondrocyte models induced with oxidative stress, demonstrate that MitoQ mitigates mitochondrial damage and ECM degradation, while enhancing the expression of anti-inflammatory genes, such as IL-10 and TGF-β, as well as antioxidant genes including SOD2, catalase, glutathione peroxidase 1 and NAD(P)H:quinone oxidoreductase 1 (2,31). In vivo, MitoQ alleviates cartilage degeneration and reduces the levels of inflammatory cytokines, which slows the progression of OA (32,33).
Research in IVDD. Studies indicate that under mechanical overload or inflammatory stimulation, intervertebral disc tissue exhibits notable accumulation of ROS, elevated apoptosis and impaired ECM synthesis (34,35). MitoQ inhibits the activation of inflammatory signaling pathways such as the NLRP3 inflammasome and NF-κB, and prevents the excessive secretion of key cytokines, such as IL-1β, TNF-α and IL-6 (30,32,36). In a rat IVDD model, MitoQ administration maintains disc height and hydration; therefore, delaying degenerative changes (6,30,36).
These findings suggest that MitoQ confers protective effects on both articular cartilage and intervertebral discs, potentially due to its ability to reduce mitochondrial oxidative damage and stabilize energy metabolism.
MitoTEMPO is a mitochondria-targeted antioxidant formed by coupling the superoxide scavenger TEMPO with the TPP+ cationic group, which removes mitochondrial superoxide radicals (37,38). Multiple in vivo and in vitro studies highlight its beneficial role in counteracting oxidative stress and apoptosis in joint cells (27,37).
Under high-glucose conditions or inflammatory stimulation, ROS levels in chondrocytes increase. Previous studies report that MitoTEMPO blocks the JNK/activator protein-1 and NF-κB pathways that are activated by ROS, leading to a reduction in the expression of MMPs compared with the expression of MMPs without MitoTEMPO treatment (27,37). Furthermore, intra-articular injection of MitoTEMPO in mice with cholesterol-induced OA reduces MMP-13 gene expression levels compared with vehicle-treated OA controls. Additionally, this intervention alleviates cartilage lesions and leads to a notable reduction in cartilage degeneration (2).
Research in IVDD. MitoTEMPO also demonstrates protective effects in IVDD. In NP cells with IL-1β exposure or endplate chondrocytes with H2O2 exposure, MitoTEMPO notably reduces the pathological increase in mitochondrial ROS compared with the stressor-only groups without MitoTEMPO treatment. This suppresses the activation of the caspase cascade and pyroptosis (25,39). When mitochondrial ROS are suppressed, PTEN-induced kinase 1 (PINK1)/Parkin-mediated mitophagy is maintained, reducing the accumulation of damaged mitochondria in cells (25,39). This process improves the function of NP cells and the homeostasis of the ECM, exerting an interventional effect on IVDD.
Research in RA. Although the research on MitoTEMPO in RA is limited, studies in synovial cell models suggest that exogenous mitochondrial ROS scavenging can attenuate the inflammatory cascade and ROS bursts (7,40). Therefore, MitoTEMPO exhibits broad-spectrum antioxidant protection by targeting mitochondria in various degenerative joint diseases.
SkQ1 is a mitochondria-targeted antioxidant derived from plastoquinone linked to TPP (8). In a RA model, low doses of SkQ1 (on the nM scale) suppresses the progression of arthritis and reduces the pathological damage to cartilage and the synovium (8). Its mechanism is associated with inducing neutrophil apoptosis, alleviating oxidative stress and improving the joint microenvironment. Although direct evidence in OA and IVDD is scarce, the robust mitochondrial antioxidant properties of SkQ1 and its efficacy in autoimmune arthritis models suggest it may hold potential value for degenerative joint diseases as well.
SS-31 (also known as Elamipretide) is a mitochondria-targeting short peptide that binds to cardiolipin on the inner mitochondrial membrane. This helps to stabilize the electron transport chain and reduce the generation of ROS (41). Studies demonstrate that SS-31 improves the mitochondrial membrane potential, inhibits inflammation and promotes the synthesis of the ECM in both intervertebral disc cells and chondrocytes (6,42).
Additionally, SS-31 reduces mitochondrial lipid peroxidation, a key factor in oxidative damage, and partially reverses stress-induced cellular senescence phenotypes (43). Its ability to ameliorate mitochondrial function also contributes to mitigating inflammatory signaling and promoting the stability of the ECM, which is critical for preventing tissue degeneration (44).
Apart from the direct use of mitochondria-targeted antioxidants, recent studies increasingly focus on mitochondrial dynamics, mitophagy and mitochondrial biogenesis as indirect methods for reducing mitochondrial ROS accumulation, which may offer a therapeutic potential in degenerative joint diseases (Fig. 2) (45,46).
The PINK1/Parkin pathway is an essential cellular mechanism for clearing damaged mitochondria. When the mitochondrial membrane potential decreases, PINK1 accumulates on the outer mitochondrial membrane, recruiting Parkin to the mitochondrial surface. Parkin then ubiquitinates several outer mitochondrial membrane substrates, including mitofusins 1/2, voltage-dependent anion channel 1 and translocase of outer mitochondrial membrane 20, which facilitates the recognition and clearance of damaged mitochondria through autophagy. Enhancing PINK1/Parkin-mediated mitophagy can reduce the release of mitochondrial ROS, which protects cells from damage (47). Previous studies demonstrate that inhibition of leucine-rich repeat kinase 2 can restore Parkin-mediated mitophagy and alleviate IVDD (48,49). Taurine, a naturally occurring amino acid derivative, promotes mitophagy by activating the PINK1/Parkin pathway, which ameliorates IVDD (50). Additionally, knocking down the expression of early growth response protein 1 activates PINK1-Parkin-dependent mitophagy, which suppresses NP cell senescence and mitochondrial damage, and slows disc degeneration (51). Furthermore, a recent study reveals that cellular repressor of E1A-stimulated genes 1 can alleviate NP cell pyroptosis through the PINK1/Parkin-associated mitophagy pathway, which improves IVDD (52).
An imbalance in mitochondrial fission and fusion also leads to the excessive generation of ROS (36). Under pathological stimuli such as mechanical compression, excessive mitochondrial fission (mediated by dynamin-related protein 1) separates damaged mitochondrial segments; however, impaired fusion processes (mediated by mitofusin 1/2 and optic atrophy 1) result in the accumulation of fragmented and dysfunctional mitochondria, causing a persistent elevation of ROS (36). Antioxidants such as MitoQ not only reduce ROS levels but also attenuate excessive fission and promote fusion of functionally intact mitochondria (36,53). Therefore, the modulation of proteins involved in mitochondrial dynamics or the combined use with mitochondrial antioxidants may be a future therapeutic direction for joint degeneration.
Nrf2, a central transcription factor for cellular antioxidant responses, induces the expression of antioxidant genes including heme oxygenase-1 (HO-1), NAD(P)H quinone dehydrogenase 1 and SOD2(32). When mitochondrial ROS levels increase, oxidative modification of Keap1 leads to the stabilization and nuclear translocation of Nrf2, which then activates antioxidant gene expression to alleviate oxidative stress and maintain cellular homeostasis (54). A recent study demonstrates that both pharmacological and genetic activation of the Nrf2 pathway serves a crucial role in mitigating inflammatory responses and apoptosis in joint cartilage and IVD tissues (49). For example, Kartogenin-loaded hydrogel promotes IVD repair by protecting mesenchymal stem cells from oxidative stress via the Nrf2/Thioredoxin-interacting protein/NLRP3 axis, which reduces inflammation and cellular apoptosis (55). Additionally, astaxanthin activates the Nrf2/HO-1 pathway, which suppresses oxidative stress and prevents cartilage endplate degeneration by inhibiting apoptosis and the degradation of the ECM (56).
SIRT family proteins serve essential roles in mitochondrial homeostasis, redox balance and the regulation of energy metabolism (25,28). For example, SIRT3, localized in mitochondria, deacetylates and activates key antioxidant enzymes such as manganese superoxide dismutase (SOD2), isocitrate dehydrogenase 2 and catalase, which enhances mitochondrial ROS detoxification (28). Upregulation of SIRT3 suppresses cellular senescence and inflammation in joint cells.
A study by Guo et al (57) reveals that dysregulation of SIRT3 and its metabolic network, including downstream effectors such as isocitrate dehydrogenase 2 and manganese SOD, serves a role in mitochondrial oxidative stress and cartilage degradation in OA. This suggests that restoring the activity of SIRT3 may serve as a therapeutic approach to delay joint degeneration (57).
AMP-activated protein kinase (AMPK)-peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) pathway. The AMPK-PGC-1α signaling axis promotes mitochondrial biogenesis and the expression of antioxidant enzymes. A study by Yang et al (58) demonstrates that exposure to advanced glycation end-products represses the AMPKα-SIRT1-PGC-1α pathway in osteoarthritic chondrocytes, with reductions in PGC-1α expression levels compared with untreated control cells, resulting in impaired mitochondrial biogenesis and antioxidant defense. However, the study by Li et al (59) demonstrates that adipokine omentin-1 can activate the AMPK-PGC-1α pathway, promoting mitochondrial biogenesis in chondrocytes and slowing OA progression. The study by Yang et al (58) demonstrates that advanced glycation end products inhibit the AMPK-SIRT1-PGC-1α pathway, which induces mitochondrial dysfunction and inflammation in chondrocytes. This suggests the antioxidant and anti-aging properties of the AMPK-SIRT1-PGC-1α pathway (58).
Additionally, the study by Guo et al (57) reveals that activation of the AMPK-PGC-1α pathway not only restores energy metabolism but also reduces the accumulation of ROS and the expression of proinflammatory cytokines, including IL-1β, IL-6 and TNF-α, in degenerative joint diseases. This demonstrates its potential role as a therapeutic axis (57).
Itaconate derivatives (such as 4-octyl itaconate). A previous study indicates that exogenous itaconate reduces inflammatory responses and inhibits the generation of mitochondrial ROS (54). Zinc-based metal-organic super containers, such as 4-octyl itaconate (4-OI)@Zn-NH-pyr, demonstrates enhanced mitochondrial targeting and ROS-scavenging activity compared with free 4-OI alone, which alleviates proinflammatory states in arthritis models (54).
Additionally, novel mitochondria-targeted nanomedicines such as Mn3O4/UIO-TPP nanozymes scavenge mitochondrial ROS and restore mitochondrial function in OA cartilage, demonstrating potential therapeutic effects in rat models with anterior cruciate ligament transection induced OA (60,61). These nanozymes represent a promising material-based extension of mitochondrial antioxidant therapy and support the feasibility of precision mitochondria-targeted intervention (62).
Mitochondria and the ER are functionally connected via mitochondria-associated membranes, which coordinate calcium signaling, lipid exchange and cellular stress responses. In degenerative joint diseases, this interplay becomes dysregulated, which contributes to oxidative injury and inflammation (63).
ER stress promotes the release of Ca2+ through inositol 1,4,5-trisphosphate receptors, while excessive uptake of Ca2+ by the mitochondria via the mitochondrial calcium uniporter leads to the overproduction of ROS, mitochondrial dysfunction and apoptosis (14). Furthermore, ER stress activates proapoptotic pathways (such as protein kinase R-like ER kinase-activating transcription factor 4-CHOP) and promotes the activation of the NLRP3 inflammasome (64).
In addition, ion channels (such as transient receptor potential cation channel, subfamily V, member 4) participate in mechano-oxidative signaling in joint cells, and their dysregulation may further exacerbate mitochondrial stress (65). Targeting ER-mitochondria interactions or associated Ca2+ channels may offer new therapeutic avenues in OA, IVDD and RA.
Mitochondrial oxidative stress not only contributes to direct tissue damage but also serves a role in modulating immune responses, which are involved in the pathogenesis of OA, IVDD and RA. Both innate immune cells, such as macrophages, dendritic cells, neutrophils and natural killer cells, and adaptive immune cells, including T cells and B cells, respond to and are regulated by mitochondrial metabolism and ROS signaling, forming an interconnected immunometabolic axis (66,67).
Innate immune cells, such as macrophages and neutrophils, are sensitive to mitochondrial ROS. In RA and IVDD, macrophages polarize toward a proinflammatory (M1-like) phenotype under oxidative stress conditions, leading to an enhanced production of TNF-α, IL-1β and IL-6, which further exacerbates tissue inflammation and matrix degradation (68). Mitochondrial ROS act as secondary messengers to activate the NLRP3 inflammasome, a key inflammatory complex that promotes caspase-1 activation and IL-1β/IL-18 maturation, which accelerates pyroptosis in disc and synovial tissues (69).
Neutrophils also exhibit enhanced respiratory bursts and NETosis in the presence of mitochondrial ROS, which contributes to synovial inflammation and cartilage degradation in RA (70). Toll-like receptors (TLRs), especially TLR2 and 4, recognize damage-associated molecular patterns (such as mitochondrial DNA and cardiolipin) that are released from stressed mitochondria. This activates the myeloid differentiation primary response protein 88-dependent signaling pathway and NF-κB-induced transcription of proinflammatory cytokines, including TNF-α, IL-1β and IL-6 (71,72).
T cell differentiation and function are dependent on metabolism. Under elevated ROS conditions, mitochondrial dysfunction promotes a T helper 17 phenotype in CD4+ T cells, promoting proinflammatory responses in RA and OA synovium (73,74). Regulatory T cells, which rely on oxidative phosphorylation for their function, are suppressed in oxidative microenvironments, which further amplifies inflammation (73).
B cells are also influenced by mitochondrial metabolism, with mitochondrial ROS promoting autoantibody production and immune complex deposition in RA (75). Mitochondria-targeted antioxidants such as MitoQ and SkQ1 dampen the activation of T cells and the secretion of cytokines, suggesting that the modulation of mitochondrial ROS may re-establish immune homeostasis in degenerative joint diseases (76).
Cytokines, such as IL-1β, TNF-α and IL-6, are not only induced by mitochondrial ROS but also feedback to further disrupt mitochondrial integrity and function. These cytokines impair mitochondrial membrane potential, increase the generation of ROS and inhibit mitophagy, forming a positive feedback loop of oxidative inflammation (77). However, anti-inflammatory cytokines (such as IL-10) may promote mitophagy and mitochondrial quality control by activating pathways such as SIRT3 or AMPK-PGC-1α (78).
Chemokines, such as chemokine (C-C motif) ligand 2 and chemokine (C-X-C motif) ligand 8, recruit immune cells into inflamed joints and are also modulated by mitochondrial oxidative stress (70). Therefore, therapeutic strategies targeting mitochondrial pathways may reduce tissue damage and immune cell infiltration.
Although mitochondria-targeted antioxidants such as MitoQ, MitoTEMPO and SkQ1 reveal notable protective effects on joints in animal models (including rodents such as mice and rats, mid-sized models such as rabbits, and large animals such as dogs and sheep), the pathological processes in animal models are more acute or controlled compared with clinical populations. Therefore, these animal models do not fully replicate the chronic, long-term degenerative processes observed in humans (79). Additionally, there are notable differences in pharmacokinetics and drug sensitivity in different species (80). Therefore, translating results from animal studies into clinical applications necessitates further validation through extensive clinical trials (8,18,32).
The majority of studies on mitochondria-targeted antioxidants rely on intra-articular injections or local administration (79,81). Although several mitochondria-targeted antioxidants, such as MitoQ, SkQ1 (Visomitin) and to an extent MitoTEMPO, can be administered orally, systemic delivery raises concerns regarding bioavailability, mitochondrial targeting efficiency, and systemic safety profiles (2,32). Local injections also have risks of tissue trauma and infection. Advancements in drug delivery systems, such as hydrogels and nanocarriers combined with mitochondria-targeting ligands, may overcome these limitations. However, achieving efficient, sustained and localized drug release at the joint is still challenging at present (3,6,82).
While drugs such as MitoQ and SkQ1 demonstrate good tolerability at low doses [for example, SkQ1 at 0.25-1.25 nmol/kg/day (0.13-0.70 µg/kg/day) in rat models and MitoQ at ~20 mg/day in human trials] (8), long-term administration (defined as continuous dosing for 3-6 months or longer in preclinical and clinical studies) requires vigilant monitoring to avoid causing redox imbalances in normal tissues. Additionally, moderate levels of ROS serve critical physiological roles in signaling and tissue repair in degenerative joint diseases. Excessive clearance of ROS may disrupt essential cellular signaling pathways (1,15). Therefore, calibrating the dose and duration of mitochondria-targeted antioxidants is crucial for clinical application.
Numerous studies increasingly advocate for combining mitochondria-targeted antioxidants (such as MitoQ, SkQ1 and MitoTEMPO) with autophagy regulators (such as apigenin and rapamycin), inflammasome inhibitors (such as MCC950 and CTSB inhibitors), growth factors (such as TGF-β and IGF-1) or conventional anti-inflammatory medications (such as non-steroidal anti-inflammatory drugs and disease-modifying anti-rheumatic drugs) in order to achieve synergistic therapeutic effects on degenerative joint diseases (7,62). For example, combining MitoTEMPO with autophagy activators, such as salidroside, honokiol or urolithin A, simultaneously reduces excessive ROS and increases the clearance rate of damaged mitochondria, which potentially inhibits apoptosis in cartilage or intervertebral discs (83).
In addition, cellular and organelle-level strategies enhance mitochondrial restoration. One such approach is mitochondrial transplantation, in which healthy exogenous mitochondria are delivered into dysfunctional cells to rescue energy production and reduce oxidative stress (84). Recent studies demonstrate that mitochondrial transplantation improves joint tissue homeostasis and attenuates degeneration in in vitro chondrocyte and NP cell models, as well as in vivo mice with destabilization of the medial meniscus-induced OA and rat with needle puncture-induced IVDD models (85,86). This strategy may be biologically complementary to chemical antioxidant therapies.
Furthermore, future therapeutic paradigms may involve mitochondrial genome editing to correct mitochondrial DNA mutations that are associated with joint degeneration. Emerging tools such as mitochondria-targeted zinc finger nucleases and mitochondria-targeted transcription activator-like effector nucleases demonstrate potential in restoring mitochondrial integrity and function in tissues with OA (87,88). Although these technologies are still mostly experimental, they could potentially be used in precision mitochondrial medicine for degenerative joint diseases in the future (87).
Mitochondrial dysfunction and an excessive generation of ROS are central pathological mechanisms underlying degenerative joint diseases, including OA, IVDD and RA. The present study demonstrated the role of mitochondrial oxidative damage in promoting disease progression, from the degradation of the cartilage ECM in OA and NP cell death in IVDD, to chronic synovitis and immune activation in RA. Mitochondria-targeted antioxidants, such as MitoQ, MitoTEMPO, SkQ1 and SS-31, demonstrate potential in mitigating oxidative stress and slowing disease deterioration. These findings provided theoretical support for a paradigm shift from symptom-based management toward mitochondria-centered interventions.
At present, despite promising preclinical data, the evidence is mostly limited to in vitro and small-animal studies, such as mice and rats, with scarce validation in human tissues or large-scale clinical settings. Additionally, the heterogeneity of degenerative joint diseases further complicates the interpretation of therapeutic efficacy. In particular, differences in mitochondrial activity, ROS levels and drug responsiveness among OA, IVDD and RA are yet to be fully characterized. Furthermore, the lack of multicenter randomized clinical trials evaluating dosing, safety profiles and delivery methods for mitochondria-targeted antioxidants is a notable limitation. Additionally, the majority of studies overlook long-term outcomes and potential off-target effects, especially in elderly or comorbid populations.
Future studies should investigate the temporal and spatial regulation of mitochondrial ROS in specific joint tissues and immune cell subpopulations. In particular, the association between mitochondrial dysfunction and epigenetic regulation, cytokine signaling or metabolic reprogramming warrants further investigation. Elucidating how mitochondrial stress interacts with cartilage calcification, subchondral bone remodeling and synovial hyperplasia may reveal additional therapeutic checkpoints. The use of advanced models such as patient-derived organoids, single-cell mitochondrial profiling and spatial transcriptomics may aid mechanistic investigations. Additionally, mitochondrial-nuclear crosstalk or sex-specific mitochondrial responses should also be investigated in the future.
Clinical translation should prioritize the development of delivery systems, including hydrogels, nanoparticles and scaffold-based platforms that enable a localized, sustained release of mitochondria-targeted antioxidants (such as MitoQ, MitoTEMPO or SkQ1). Additionally, their combination with autophagy enhancers (such as rapamycin, spermidine or resveratrol) or regenerative factors [such as bone morphogenetic proteins (BMP)-2 and -7] or TGF-β) at degenerative sites should be investigated. Furthermore, integrating mitochondria-targeted antioxidants with mitophagy enhancers (such as rapamycin, urolithin A and salidroside), immune modulators (such as IL-17A antibodies, Treg enhancers and fumaric acid derivatives) or regenerative therapies (such as stem cells or exosome-based systems) may also offer a therapeutic effect. Such combination regimens may synergistically halt or reverse disease progression. Furthermore, precision medicine frameworks leveraging genetic, metabolic or imaging biomarkers may allow for personalized dosing and treatment selection, which may also enhance therapeutic outcomes.
Mitochondria-centered therapies involve multi-disciplinary teams from orthopedics, immunology, geriatrics and bioengineering. However, a number of controversies remain unresolved. For example, contradictory findings regarding the role of ROS as only deleterious vs. the role of ROS as signaling mediators in joint homeostasis. Moreover, the possibility that prolonged ROS suppression might impair physiological defense mechanisms or tissue remodeling should be investigated. Interdisciplinary dialogue and standardized methodological approaches will likely be required to address these issues and guide the rational design of future therapies.
Not applicable.
Funding: No funding was received.
Not applicable.
YH designed the study and drafted the manuscript. XY summarized the comments from the reviewers, drafted the responses and contributed to manuscript revision and language editing. TZ contributed to the literature review and assisted in data interpretation. YG revised the manuscript. JS designed the present study, revised the manuscript and supervised the project. All authors read and approved the final version of the manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Martin JA, Martini A, Molinari A, Morgan W, Ramalingam W, Buckwalter JA and McKinley TO: Mitochondrial electron transport and glycolysis are coupled in articular cartilage. Osteoarthritis Cartilage. 20:323–329. 2012.PubMed/NCBI View Article : Google Scholar | |
|
Bolduc JA, Collins JA and Loeser RF: Reactive oxygen species, aging and articular cartilage homeostasis. Free Radic Biol Med. 132:73–82. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Chen Q, Qian Q, Xu H, Zhou H, Chen L, Shao N, Zhang K, Chen T, Tian H, Zhang Z, et al: Mitochondrial-targeted metal-phenolic nanoparticles to attenuate intervertebral disc degeneration: alleviating oxidative stress and mitochondrial dysfunction. ACS Nano. 18:8885–8905. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Hunter DJ and Bierma-Zeinstra S: Osteoarthritis. Lancet. 393:1745–1759. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Nasto LA, Robinson AR, Ngo K, Clauson CL, Dong Q, St Croix C, Sowa G, Pola E, Robbins PD, Kang J, et al: Mitochondrial-derived reactive oxygen species (ROS) play a causal role in aging-related intervertebral disc degeneration. J Orthop Res. 31:1150–1157. 2013.PubMed/NCBI View Article : Google Scholar | |
|
Wang Y, Deng M, Wu Y, Zheng C, Zhang F, Guo C, Zhang B, Hu C, Kong Q and Wang Y: A multifunctional mitochondria-protective gene delivery platform promote intervertebral disc regeneration. Biomaterials. 317(123067)2025.PubMed/NCBI View Article : Google Scholar | |
|
Al-Azab M, Qaed E, Ouyang X, Elkhider A, Walana W, Li H, Li W, Tang Y, Adlat S, Wei J, et al: TL1A/TNFR2-mediated mitochondrial dysfunction of fibroblast-like synoviocytes increases inflammatory response in patients with rheumatoid arthritis via reactive oxygen species generation. FEBS J. 287:3088–3104. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Andreev-Andrievskiy AA, Kolosova NG, Stefanova NA, Lovat MV, Egorov MV, Manskikh VN, Zinovkin RA, Galkin II, Prikhodko AS, Skulachev MV and Lukashev AN: Efficacy of mitochondrial antioxidant plastoquinonyl-decyl-triphenylphosphonium bromide (SkQ1) in the rat model of autoimmune arthritis. Oxid Med Cell Longev. 2016(8703645)2016.PubMed/NCBI View Article : Google Scholar | |
|
Saeidnia S and Abdollahi M: Toxicological and pharmacological concerns on oxidative stress and related diseases. Toxicol Appl Pharmacol. 273:442–455. 2013.PubMed/NCBI View Article : Google Scholar | |
|
Burton GJ and Jauniaux E: Oxidative stress. Best Pract Res Clin Obstet Gynaecol. 25:287–299. 2011.PubMed/NCBI View Article : Google Scholar | |
|
Cao G, Yang S, Cao J, Tan Z, Wu L, Dong F, Ding W and Zhang F: the role of oxidative stress in intervertebral disc degeneration. Oxid Med Cell Longev. 2022(2166817)2022.PubMed/NCBI View Article : Google Scholar | |
|
Wen P, Zheng B, Zhang B, Ma T, Hao L and Zhang Y: The role of ageing and oxidative stress in intervertebral disc degeneration. Front Mol Biosci. 9(1052878)2022.PubMed/NCBI View Article : Google Scholar | |
|
Peng X, Zhang C, Zhou ZM, Wang K, Gao JW, Qian ZY, Bao JP, Ji HY, Cabral VLF and Wu XT: A20 attenuates pyroptosis and apoptosis in nucleus pulposus cells via promoting mitophagy and stabilizing mitochondrial dynamics. Inflamm Res. 71:695–710. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Li W, Cao T, Luo C, Cai J, Zhou X, Xiao X and Liu S: Crosstalk between ER stress, NLRP3 inflammasome, and inflammation. Appl Microbiol Biotechnol. 104:6129–6140. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Bartell SM, Kim HN, Ambrogini E, Han L, Iyer S, Serra Ucer S, Rabinovitch P, Jilka RL, Weinstein RS, Zhao H, et al: FoxO proteins restrain osteoclastogenesis and bone resorption by attenuating H2O2 accumulation. Nat Commun. 5(3773)2014.PubMed/NCBI View Article : Google Scholar | |
|
Kim J, Xu M, Xo R, Mates A, Wilson GL, Pearsall AW IV and Grishko V: Mitochondrial DNA damage is involved in apoptosis caused by pro-inflammatory cytokines in human OA chondrocytes. Osteoarthritis Cartilage. 18:424–432. 2010.PubMed/NCBI View Article : Google Scholar | |
|
Palma FR, He C, Danes JM, Paviani V, Coelho DR, Gantner BN and Bonini MG: Mitochondrial superoxide dismutase: What the established, the intriguing, and the novel reveal about a key cellular redox switch. Antioxid Redox Signal. 32:701–714. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Farnaghi S, Prasadam I, Cai G, Friis T, Du Z, Crawford R, Mao X and Xiao Y: Protective effects of mitochondria-targeted antioxidants and statins on cholesterol-induced osteoarthritis. FASEB J. 31:356–367. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Liu Y, Fiskum G and Schubert D: Generation of reactive oxygen species by the mitochondrial electron transport chain. J Neurochem. 80:780–787. 2002.PubMed/NCBI View Article : Google Scholar | |
|
Murphy MP: How mitochondria produce reactive oxygen species. Biochem J. 417:1–13. 2008.PubMed/NCBI View Article : Google Scholar | |
|
Valcárcel-Ares MN, Riveiro-Naveira RR, Vaamonde-García C, Loureiro J, Hermida-Carballo L, Blanco FJ and López-Armada MJ: Mitochondrial dysfunction promotes and aggravates the inflammatory response in normal human synoviocytes. Rheumatology (Oxford). 53:1332–1343. 2014.PubMed/NCBI View Article : Google Scholar | |
|
Cui H, Kong Y and Zhang H: Oxidative stress, mitochondrial dysfunction, and aging. J Signal Transduct. 2012(646354)2012.PubMed/NCBI View Article : Google Scholar | |
|
Blanco FJ, López-Armada MJ and Maneiro E: Mitochondrial dysfunction in osteoarthritis. Mitochondrion. 4:715–728. 2004.PubMed/NCBI View Article : Google Scholar | |
|
Early JO, Fagan LE, Curtis AM and Kennedy OD: Mitochondria in injury, inflammation and disease of articular skeletal joints. Front Immunol. 12(695257)2021.PubMed/NCBI View Article : Google Scholar | |
|
Ma Z, Tang P, Dong W, Lu Y, Tan B, Zhou N, Hao J, Shen J and Hu Z: SIRT1 alleviates IL-1β induced nucleus pulposus cells pyroptosis via mitophagy in intervertebral disc degeneration. Int Immunopharmacol. 107(108671)2022.PubMed/NCBI View Article : Google Scholar | |
|
Hu Z, Wang Y, Gao X, Zhang Y, Liu C, Zhai Y, Chang X, Li H, Li Y, Lou J and Li C: Optineurin-mediated mitophagy as a potential therapeutic target for intervertebral disc degeneration. Front Pharmacol. 13(893307)2022.PubMed/NCBI View Article : Google Scholar | |
|
Ansari MY, Ahmad N, Voleti S, Wase SJ, Novak K and Haqqi TM: Mitochondrial dysfunction triggers a catabolic response in chondrocytes via ROS-mediated activation of the JNK/AP1 pathway. J Cell Sci. 133(jcs247353)2020.PubMed/NCBI View Article : Google Scholar | |
|
Song Y, Li S, Geng W, Luo R, Liu W, Tu J, Wang K, Kang L, Yin H, Wu X, et al: Sirtuin 3-dependent mitochondrial redox homeostasis protects against AGEs-induced intervertebral disc degeneration. Redox Biol. 19:339–353. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Wu C, Luo J, Liu Y, Fan J, Shang X, Liu R, Ye C, Yang J and Cao H: Doxorubicin suppresses chondrocyte differentiation by stimulating ROS production. Eur J Pharm Sci. 167(106013)2021.PubMed/NCBI View Article : Google Scholar | |
|
Li BL, Liu X, Gao M, Zhang F, Chen X, He Z, Wang J, Tian W, Chen D, Zhou Z and Liu S: Programmed NP cell death induced by mitochondrial ROS in a one-strike loading disc degeneration organ culture model. Oxid Med Cell Longev. 2021(5608133)2021.PubMed/NCBI View Article : Google Scholar | |
|
Shao Y, Zhang H, Guan H, Wu C, Qi W, Yang L, Yin J, Zhang H, Liu L, Lu Y, et al: PDZK1 protects against mechanical overload-induced chondrocyte senescence and osteoarthritis by targeting mitochondrial function. Bone Res. 12(41)2024.PubMed/NCBI View Article : Google Scholar | |
|
Hou L, Wang G, Zhang X, Lu F, Xu J, Guo Z, Lin J, Zheng Z, Liu H, Hou Y, et al: Mitoquinone alleviates osteoarthritis progress by activating the NRF2-Parkin axis. iScience. 26(107647)2023.PubMed/NCBI View Article : Google Scholar | |
|
Poudel SB, Ruff RR, Yildirim G, Miller RA, Harrison DE, Strong R, Kirsch T and Yakar S: Development of primary osteoarthritis during aging in genetically diverse UM-HET3 mice. Arthritis Res Ther. 26(118)2024.PubMed/NCBI View Article : Google Scholar | |
|
Tisherman R, Coelho P, Phillibert D, Wang D, Dong Q, Vo N, Kang J and Sowa G: NF-κB signaling pathway in controlling intervertebral disk cell response to inflammatory and mechanical stressors. Phys Ther. 96:704–711. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Wang F, Cai F, Shi R, Wang XH and Wu XT: Aging and age related stresses: A senescence mechanism of intervertebral disc degeneration. Osteoarthritis Cartilage. 24:398–408. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Kang L, Liu S, Li J, Tian Y, Xue Y and Liu X: The mitochondria-targeted anti-oxidant MitoQ protects against intervertebral disc degeneration by ameliorating mitochondrial dysfunction and redox imbalance. Cell Prolif. 53(e12779)2020.PubMed/NCBI View Article : Google Scholar | |
|
Laiguillon MC, Courties A, Houard X, Auclair M, Sautet A, Capeau J, Fève B, Berenbaum F and Sellam J: Characterization of diabetic osteoarthritic cartilage and role of high glucose environment on chondrocyte activation: Toward pathophysiological delineation of diabetes mellitus-related osteoarthritis. Osteoarthritis Cartilage. 23:1513–1522. 2025.PubMed/NCBI View Article : Google Scholar | |
|
Ansari MY, Ball HC, Wase SJ, Novak K and Haqqi TM: Lysosomal dysfunction in osteoarthritis and aged cartilage triggers apoptosis in chondrocytes through BAX mediated release of Cytochrome c. Osteoarthritis Cartilage. 29:100–112. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Kang L, Liu S, Li J, Tian Y, Xue Y and Liu X: Parkin and Nrf2 prevent oxidative stress-induced apoptosis in intervertebral endplate chondrocytes via inducing mitophagy and anti-oxidant defenses. Life Sci. 243(117244)2020.PubMed/NCBI View Article : Google Scholar | |
|
McGarry T, Biniecka M, Gao W, Cluxton D, Canavan M, Wade S, Wade S, Gallagher L, Orr C, Veale DJ and Fearon U: Resolution of TLR2-induced inflammation through manipulation of metabolic pathways in Rheumatoid arthritis. Sci Rep. 7(43165)2017.PubMed/NCBI View Article : Google Scholar | |
|
Szeto HH, Liu S, Soong Y, Alam N, Prusky GT and Seshan SV: Protection of mitochondria prevents high-fat diet-induced glomerulopathy and proximal tubular injury. Kidney Int. 90:997–1011. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Peng X, Wang K, Zhang C, Bao JP, Vlf C, Gao JW, Zhou ZM and Wu XT: The mitochondrial antioxidant SS-31 attenuated lipopolysaccharide-induced apoptosis and pyroptosis of nucleus pulposus cells via scavenging mitochondrial ROS and maintaining the stability of mitochondrial dynamics. Free Radic Res. 55:1080–1093. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Zhang X, Eliasberg CD and Rodeo SA: Mitochondrial dysfunction and potential mitochondrial protectant treatments in tendinopathy. Ann N Y Acad Sci. 1490:29–41. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Siekacz K, Piotrowski WJ, Iwański MA, Górski P and Białas AJ: The role of interaction between mitochondria and the extracellular matrix in the development of idiopathic pulmonary fibrosis. Oxid Med Cell Longev. 2021(9932442)2021.PubMed/NCBI View Article : Google Scholar | |
|
An F, Zhang J, Gao P, Xiao Z, Chang W, Song J, Wang Y, Ma H, Zhang R, Chen Z and Yan C: New insight of the pathogenesis in osteoarthritis: the intricate interplay of ferroptosis and autophagy mediated by mitophagy/chaperone-mediated autophagy. Front Cell Dev Biol. 11(1297024)2023.PubMed/NCBI View Article : Google Scholar | |
|
Wu J, Zhou X, Xu X and Xie J: A molecular chemical perspective: mitochondrial dynamics is not a bystander of cartilage diseases. ACS Pharmacol Transl Sci. 8:1473–1497. 2025.PubMed/NCBI View Article : Google Scholar | |
|
Xiao B, Goh JY, Xiao L, Xian H, Lim KL and Liou YC: Reactive oxygen species trigger Parkin/PINK1 pathway-dependent mitophagy by inducing mitochondrial recruitment of Parkin. J Biol Chem. 292:16697–16708. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Lin J, Zheng X, Zhang Z, Zhuge J, Shao Z, Huang C, Jin J, Chen X, Chen Y, Wu Y, et al: Inhibition of LRRK2 restores parkin-mediated mitophagy and attenuates intervertebral disc degeneration. Osteoarthritis Cartilage. 29:579–591. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Zeng Z, Zhou X and Wang Y, Cao H, Guo J, Wang P, Yang Y and Wang Y: Mitophagy-A new target of bone disease. Biomolecules. 12(1420)2022.PubMed/NCBI View Article : Google Scholar | |
|
Lin S, Li T, Zhang B and Wang P: Taurine rescues intervertebral disc degeneration by activating mitophagy through the PINK1/Parkin pathway. Biochem Biophys Res Commun. 739(150587)2024.PubMed/NCBI View Article : Google Scholar | |
|
Wu ZL, Wang KP, Chen YJ, Song W, Liu Y, Zhou KS, Mao P, Ma Z and Zhang HH: Knocking down EGR1 inhibits nucleus pulposus cell senescence and mitochondrial damage through activation of PINK1-Parkin dependent mitophagy, thereby delaying intervertebral disc degeneration. Free Radic Biol Med. 224:9–22. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Zhang Y, Xing D, Liu Y, Sha S, Xiao Y, Liu Z, Yin Q, Gao Z and Liu W: CREG1 attenuates intervertebral disc degeneration by alleviating nucleus pulposus cell pyroptosis via the PINK1/Parkin-related mitophagy pathway. Int Immunopharmacol. 147(113974)2025.PubMed/NCBI View Article : Google Scholar | |
|
Cheung C, Tu S, Feng Y, Wan C, Ai H and Chen Z: Mitochondrial quality control dysfunction in osteoarthritis: Mechanisms, therapeutic strategies & future prospects. Arch Gerontol Geriatr. 125(105522)2024.PubMed/NCBI View Article : Google Scholar | |
|
Luchkova A, Mata A and Cadenas S: Nrf2 as a regulator of energy metabolism and mitochondrial function. FEBS Lett. 598:2092–2105. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Wang F, Guo K, Nan L, Wang S, Lu J, Wang Q, Ba Z, Huang Y and Wu D: Kartogenin-loaded hydrogel promotes intervertebral disc repair via protecting MSCs against reactive oxygen species microenvironment by Nrf2/TXNIP/NLRP3 axis. Free Radic Biol Med. 204:128–150. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Yang G, Liu X, Jing X, Wang J, Wang H, Chen F, Wang W, Shao Y and Cui X: Astaxanthin suppresses oxidative stress and calcification in vertebral cartilage endplate via activating Nrf-2/HO-1 signaling pathway. Int Immunopharmacol. 119(110159)2023.PubMed/NCBI View Article : Google Scholar | |
|
Guo P, Alhaskawi A, Adel Abdo Moqbel S and Pan Z: Recent development of mitochondrial metabolism and dysfunction in osteoarthritis. Front Pharmacol. 16(1538662)2025.PubMed/NCBI View Article : Google Scholar | |
|
Yang Q, Shi Y, Jin T, Duan B and Wu S: Advanced glycation end products induced mitochondrial dysfunction of chondrocytes through repression of AMPKα-SIRT1-PGC-1α pathway. Pharmacology. 107:298–307. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Li Z, Zhang Y, Tian F, Wang Z, Song H, Chen H and Wu B: Omentin-1 promotes mitochondrial biogenesis via PGC1α-AMPK pathway in chondrocytes. Arch Physiol Biochem. 129:291–297. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Zhang S, Cai J, Yao Y, Huang L, Zheng L and Zhao J: Mitochondrial-targeting Mn3O4/UIO-TPP nanozyme scavenge ROS to restore mitochondrial function for osteoarthritis therapy. Regen Biomater. 10(rbad078)2023.PubMed/NCBI View Article : Google Scholar | |
|
Wang SH, Xu XL and Chen W: How do organelle-targeting nanotherapeutics treat inflammatory diseases? A comprehensive review of the literature. Int J Nanomedicine. 20:7133–7152. 2025.PubMed/NCBI View Article : Google Scholar | |
|
Chen X, Li C, Cao X, Jia X, Chen X, Wang Z, Xu W, Dai F and Zhang S: Mitochondria-targeted supramolecular coordination container encapsulated with exogenous itaconate for synergistic therapy of joint inflammation. Theranostics. 12:3251–3272. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Kan S, Duan M, Liu Y, Wang C and Xie J: Role of mitochondria in physiology of chondrocytes and diseases of osteoarthritis and rheumatoid arthritis. Cartilage. 13 (2_suppl):1102S–1121S. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Liu X, Chen Y, Wang H, Wei Y, Yuan Y, Zhou Q, Fang F, Shi S, Jiang X, Dong Y and Li X: Microglia-derived IL-1β promoted neuronal apoptosis through ER stress-mediated signaling pathway PERK/eIF2α/ATF4/CHOP upon arsenic exposure. J Hazard Mater. 417(125997)2021.PubMed/NCBI View Article : Google Scholar | |
|
Yan Z, He Z, Jiang H and Zhang Y, Xu Y and Zhang Y: TRPV4-mediated mitochondrial dysfunction induces pyroptosis and cartilage degradation in osteoarthritis via the Drp1-HK2 axis. Int Immunopharmacol. 123(110651)2023.PubMed/NCBI View Article : Google Scholar | |
|
Li P, Zhou M, Wang J, Tian J, Zhang L, Wei Y, Yang F, Xu Y and Wang G: Important role of mitochondrial dysfunction in immune triggering and inflammatory response in rheumatoid arthritis. J Inflamm Res. 17:11631–11657. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Hu K, Song M, Song T, Jia X and Song Y: Osteoimmunology in osteoarthritis: Unraveling the interplay of immunity, inflammation, and joint degeneration. J Inflamm Res. 18:4121–4142. 2025.PubMed/NCBI View Article : Google Scholar | |
|
Dou Y, Zhang Y, Liu Y, Sun X, Liu X, Li B and Yang Q: Role of macrophage in intervertebral disc degeneration. Bone Res. 13(15)2025.PubMed/NCBI View Article : Google Scholar | |
|
Wang S, Wang H, Feng C, Li C, Li Z, He J and Tu C: The regulatory role and therapeutic application of pyroptosis in musculoskeletal diseases. Cell Death Discov. 8(492)2022.PubMed/NCBI View Article : Google Scholar | |
|
Wright HL, Lyon M, Chapman EA, Moots RJ and Edwards SW: Rheumatoid arthritis synovial fluid neutrophils drive inflammation through production of chemokines, reactive oxygen species, and neutrophil extracellular traps. Front Immunol. 11(584116)2021.PubMed/NCBI View Article : Google Scholar | |
|
Wu B, Ni H, Li J, Zhuang X, Zhang J, Qi Z, Chen Q, Wen Z, Shi H, Luo X and Jin B: The impact of circulating mitochondrial DNA on cardiomyocyte apoptosis and myocardial injury after TLR4 activation in experimental autoimmune myocarditis. Cell Physiol Biochem. 42:713–728. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Mukherjee S, Patra R, Behzadi P, Masotti A, Paolini A and Sarshar M: Toll-like receptor-guided therapeutic intervention of human cancers: Molecular and immunological perspectives. Front Immunol. 14(1244345)2023.PubMed/NCBI View Article : Google Scholar | |
|
Chávez MD and Tse HM: Targeting mitochondrial-derived reactive oxygen species in T cell-mediated autoimmune diseases. Front Immunol. 12(703972)2021.PubMed/NCBI View Article : Google Scholar | |
|
Masoumi M, Alesaeidi S, Khorramdelazad H, Behzadi M, Baharlou R, Alizadeh-Fanalou S and Karami J: Role of T cells in the pathogenesis of rheumatoid arthritis: Focus on immunometabolism dysfunctions. Inflammation. 46:88–102. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Weyand CM, Wu B, Huang T, Hu Z and Goronzy JJ: Mitochondria as disease-relevant organelles in rheumatoid arthritis. Clin Exp Immunol. 211:208–223. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Jing W, Liu C, Su C, Liu L, Chen P, Li X, Zhang X, Yuan B, Wang H and Du X: Role of reactive oxygen species and mitochondrial damage in rheumatoid arthritis and targeted drugs. Front Immunol. 14(1107670)2023.PubMed/NCBI View Article : Google Scholar | |
|
Michalak KP and Michalak AZ: Understanding chronic inflammation: couplings between cytokines, ROS, NO, Cai2+, HIF-1α, Nrf2 and autophagy. Front Immunol. 16(1558263)2025.PubMed/NCBI View Article : Google Scholar | |
|
Sung JY, Kim SG, Park SY, Kim JR and Choi HC: Telomere stabilization by metformin mitigates the progression of atherosclerosis via the AMPK-dependent p-PGC-1α pathway. Exp Mol Med. 56:1967–1979. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Zhi ZY and Wang PC: The mitochondrial targeting drug SkQ1 attenuates the progression of post-traumatic osteoarthritis through suppression of mitochondrial oxidative stress. Curr Mol Pharmacol. 17(e18761429383749)2024.PubMed/NCBI View Article : Google Scholar | |
|
Xiong Z, Liao Y, Zhang Z, Wan Z, Liang S and Guo J: Molecular insights into oxidative-stress-mediated cardiomyopathy and potential therapeutic strategies. Biomolecules. 15(670)2025.PubMed/NCBI View Article : Google Scholar | |
|
Wu J, Xu J, Zhang M, Zhong J, Gao W and Wu M: Chondrocyte mitochondrial quality control: A novel insight into osteoarthritis and cartilage regeneration. Adv Wound Care (New Rochelle): Apr 18, 2025 (Epub ahead of print). | |
|
Wang Z, Yin F, Xu J, Zhang T, Wang G, Mao M, Wang Z, Sun W, Han J, Yang M, et al: CYT997(Lexibulin) induces apoptosis and autophagy through the activation of mutually reinforced ER stress and ROS in osteosarcoma. J Exp Clin Cancer Res. 38(44)2019.PubMed/NCBI View Article : Google Scholar | |
|
Saberi M, Zhang X and Mobasheri A: Targeting mitochondrial dysfunction with small molecules in intervertebral disc aging and degeneration. Geroscience. 43:517–537. 2021.PubMed/NCBI View Article : Google Scholar | |
|
McCully JD, Levitsky S, del Nido PJ and Cowan DB: Mitochondrial transplantation for therapeutic use. Clin Trans Med. 5(e16)2016.PubMed/NCBI View Article : Google Scholar | |
|
Luo H, Lai Y, Tang W, Wang G, Shen J and Liu H: Mitochondrial transplantation: A promising strategy for treating degenerative joint diseases. J Transl Med. 22(941)2024.PubMed/NCBI View Article : Google Scholar | |
|
Lee AR, Woo JS, Lee SY, Na HS, Cho KH, Lee YS, Lee JS, Kim SA, Park SH, Kim SJ and Cho ML: Mitochondrial transplantation ameliorates the development and progression of osteoarthritis. Immune Netw. 22(e14)2022.PubMed/NCBI View Article : Google Scholar | |
|
Zhong G, Madry H and Cucchiarini M: Mitochondrial genome editing to treat human osteoarthritis-A narrative review. Int J Mol Sci. 23(1467)2022.PubMed/NCBI View Article : Google Scholar | |
|
Yang X, Jiang J, Li Z, Liang J and Xiang Y: Strategies for mitochondrial gene editing. Comput Struct Biotechnol J. 19:3319–3329. 2021.PubMed/NCBI View Article : Google Scholar |