Skip to main content

From dysfunction to healing: advances in mitochondrial therapy for Osteoarthritis

Abstract

Osteoarthritis (OA) is a chronic degenerative joint condition characterised by cartilage deterioration and changes in bone morphology, resulting in pain and impaired joint mobility. Investigation into the pathophysiological mechanisms underlying OA has highlighted the significance of mitochondrial dysfunction in its progression. Mitochondria, which are cellular organelles, play a crucial role in regulating energy metabolism, generating reactive oxygen species, and facilitating essential biological processes including apoptosis. In recent years, the utilisation of exogenous drugs and MT to improve mitochondrial function in chondrocytes has shown great promise in OA treatment. Numerous studies have investigated the potential of stem cells and extracellular vesicles in mitochondrial transfer. This review aims to explore the underlying mechanisms of mitochondrial dysfunction in OA and assess the progress in utilising mitochondrial transfer as a therapeutic approach for this disease.

Introduction

Osteoarthritis (OA), a common long-term joint condition, is identified by the breakdown of cartilage, changes in the bone beneath the cartilage, the development of bony outgrowths, joint synovial inflammation, and decreased joint mobility. The condition is one of the most common causes of pain and disability, especially among the elderly [1]. It is estimated that around 10% of males and 18% of females aged ≥ 60 years are affected by OA, resulting in significant burdens on healthcare systems [3]. Unfortunately, there is currently a lack of disease-modifying therapies for OA, and the main treatment strategies still focus on pain relief and improving joint function. Consequently, the treatment of OA remains a noteworthy, unaddressed medical issue and an active field of investigation.

Mitochondria (MT) are cellular organelles responsible for the metabolism and generation of energy in eukaryotic cells. They are essential for maintaining the energy balance within cells [4]. MT take up one-fifth of the area in a eukaryotic cell [5]. Adenosine triphosphate (ATP) generated through the process of oxidative phosphorylation (OXPHOS) is essential for cellular maintenance and regeneration. In addition, OXPHOS also serves as the primary source of reactive oxygen species (ROS) in most tissues [6]. Perturbations in mitochondrial function and metabolism have been linked to many degenerative diseases, including cancer, neurodegenerative disorders, and ischaemic cardiomyopathy [7]. Thus, it is crucial to fully understand the mechanisms behind mitochondrial dysfunction to develop effective treatments for these diseases [9].

Mitochondrial dysfunction has the potential to influence multiple pathways implicated in joint degradation, encompassing hypoxia-induced signaling mechanisms within synovial epithelial cells, impaired biosynthetic processes in chondrocytes, and altered growth responses [5]. Chondrocytes are essential for maintaining the balance between the production and breakdown of the ECM in articular cartilage. Damage to chondrocytes manifests primarily through elevated levels of matrix metalloproteinase-3 (MMP-3) and MMP-13, nitric oxide (NO), and inflammatory cytokines, resulting in an imbalance between ECM catabolism and anabolism [12]. This imbalance leads to reduced levels of aggrecan and collagen II, ultimately culminating in the development of OA. Recent studies have elucidated the significant role of mitochondrial dysfunction and perturbed energy metabolism in the aetiology of OA [14]. Chondrocytes isolated from individuals with OA exhibit reduced mitochondrial membrane potential, decreased ATP synthesis, and increased ROS production, increasing oxidative stress and apoptosis, and promoting cartilage degradation [15]. More recently, a mitochondrial DNA (mtDNA) variation (m.16519 C) has also been suggested to be strongly associated with rapid progression of knee OA [18]. As a result of these findings, mitochondrial dysregulation is increasingly recognised as one of the major contributing factors to OA. Therefore, more effective therapeutic strategies targeting mitochondrial metabolism are needed.

With the advancements in stem cell therapy and biomaterials, optimising mitochondrial function provides a new therapeutic approach for treating OA [19]. Additionally, the field of gene therapy is experiencing significant growth, suggesting that biological interventions aimed at modifying OA will be a major treatment approach in the future [21]. Given that chondrocytes are the most important cells in articular cartilage, this article provides a comprehensive review of the underlying mechanism of mitochondrial dysfunction in OA chondrocytes and offers a summary of drugs that restore mitochondrial function in chondrocytes for the treatment of OA. Furthermore, we have evaluated the advancements made in mitochondrial transfer for treating OA, potentially guiding the future of mitochondrial studies aimed at addressing this condition.

Mitochondrial Dysfunction in OA

There is evidence that mitochondrial dysfunction precedes cartilage degradation and contributes to the death of chondrocytes [22]. Multiple factors have been identified as causes of mitochondrial impairment in OA, such as inflammation, ageing, infection, lack of nutrients, and genetic mutations [5]. Oxidative stress is a critical determinant in the induction of mtDNA damage, impairment of mitochondrial respiratory function, and activation of MT-mediated apoptotic pathways. Inflammatory cytokines, such as interleukin-1β (IL-1β)and tumour necrosis factor-alpha (TNF-α) have been documented to diminish mitochondrial activity and ATP production, impair mitochondrial respiration, and contribute to mitochondrial dysfunction in chondrocytes. In addition, gene mutations have also been implicated in mitochondrial dysfunction associated with OA. Aberrant expression of Parkin and P62, which are key mediators of mitophagy, has been observed in OA [25]. Other mechanisms, such as altered mitochondrial biogenesis, have been implicated in the pathophysiology of mitochondrial dysfunction in OA. Dysregulation of the PGC-1α/NRF-1 signalling axis, which serves as a critical regulator of mitochondrial biogenesis, has been observed in OA chondrocytes, resulting in a reduction of mitochondrial mass and function [26]. In summary, various factors contribute to mitochondrial dysfunction in OA.

Structural and locational alterations of mitochondrial components, induced by various factors, can precipitate mitochondrial dysfunction, inflicting significant damage on cells. The resultant mitochondrial dysfunction can trigger extensive cell death, propagating damage across tissues and organs in a cascading manner, akin to a domino effect, and culminating in life-threatening disorders. In OA, mitochondrial dysfunction primarily manifests through decreased ATP production, increased oxidative stress, disrupted mitochondrial dynamics and metabolism, alterations in morphology and function, and impaired calcium homeostasis. These mitochondrial impairments ultimately result in cartilage degeneration (Fig. 1) [5].

Fig. 1
figure 1

Mechanism of mitochondrial dysfunction in osteoarthritis (OA)

Decreased ATP Production

Cellular energy primarily comes from two processes: OXPHOS when oxygen is available and glycolysis when oxygen is not present. Chondrocytes, situated in an environment with relatively low oxygen levels, fulfil a substantial portion of their energy requirements through glycolysis, while OXPHOS accounts for only 25% of the overall ATP production in chondrocytes [29]. Although MT are not the principal energy source for chondrocytes, they perform a vital function in supporting and maintaining chondrocyte glycolysis. In their research, Rajpurohit et al. [30] utilised 2,4-dinitrophenol (2,4-DNP) to isolate electron transport from ATP synthesis, leading to a decrease in ATP generation in chondrocytes without a rise in lactate levels. In another study, researchers reduced ATP production in bovine cartilage after administering the mitochondrial oxidative respiratory chain inhibitor rotenone or the mitochondrial free radical scavenger MitoQ [31]. This phenomenon indicates that chondrocytes still rely on mitochondrial OXPHOS for energy to some extent. In fact, in a hypoxic environment, mitochondrial aerobic respiration is weak, but the oxidants produced promote anaerobic glycolysis and increase ATP production [32]. In OA chondrocytes, the mitochondrial membrane potential is lost and the activity of complexes I, II, and III in the electron transport chain is reduced, resulting in abnormal mitochondrial OXPHOS, ultimately leading to reduced ATP production [33].

Increased Oxidative Stress

A major cause of mtDNA damage, impairment of mitochondrial respiratory function, and activation of MT-mediated cell death pathways is oxidative stress [34]. MT are the primary organelles within cells responsible for producing ROS [32]. Malfunction of MT can trigger the release of mtDNA and mitochondrial ROS (mtROS), leading to activation of the inflammasome and promoting the generation of pro-inflammatory cytokines such as IL-1β and IL-18 in chondrocytes and synovial cells. The buildup of mtROS and mtDNA damage can trigger the nuclear factor-κB (NF-κB) pathway, which serves as the primary controller of inflammation [35]. Activation of NF-κB also enhances the production of MMPs, leading to degradation of the ECM and damage to cartilage. Furthermore, oxidative stress damages protein complexes of the chondrocyte mitochondrial respiratory chain [37]. Elevated ROS levels lead to mitochondrial membrane depolarization, which will further promote the continued generation of ROS. Excessive ROS production and ATP depletion inhibit cell division and disrupt the redox equilibrium [37].

Mitochondrial Dynamics Imbalance

By continually undergoing fission, fusion, and mitophagy, MT maintain a dynamic balance in the mitochondrial network [38]. Mitofusin 1 (Mfn1) and Mfn2 are responsible for merging the outer mitochondrial membrane (OMM), whereas optic atrophy 1 (OPA1) is responsible for merging the inner mitochondrial membrane (IMM). Mitochondrial fusion effectively preserves mtDNA levels and boosts mitochondrial respiration and ATP synthesis [39]. Dynamin-related protein 1 (Drp1) and dynamin 2 (Dnm2) are the main players in mitochondrial fission. Excessive mitochondrial fission in chondrocytes leads to reduced bioenergetic production, impaired calcium regulation, and disruption of redox balance. Mitophagy is a special type of autophagy that is regulated by a variety of autophagy-related proteins and can selectively degrade damaged or redundant MT to maintain mitochondrial health [41]. The balance of mitochondrial dynamics is controlled by these specific genes and proteins and changes according to the needs of the cell or external stimulation [42]. There is evidence that mitochondrial fission is increased and mitophagy and fusion are attenuated in OA chondrocytes. Restoring this balance helps restore cell function [43].

Perturbed Metabolism

Protein complexes in the IMM help move protons through the mitochondrial respiratory chain to produce ATP. Prominent examples of such complexes are NADH dehydrogenase (complex I), succinate dehydrogenase (complex II), cytochrome c (Cyt-C) reductase (complex III), and Cyt-C oxidase (complex IV) [44]. Perturbations in mitochondrial metabolism may lead to disturbances in cellular redox equilibrium and the accumulation of ROS in oxidative stress-related disorders. Recent research suggests that alterations in mitochondrial metabolism could be involved in the development of mild inflammation in OA [5]. During the development of OA, chondrocytes and synoviocytes tend to change their mitochondrial metabolism by switching from OXPHOS to glycolysis, which is mainly controlled by the AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin (mTOR) pathways. At the same time, alterations in lipid and amino acid metabolism have been observed in these cells [45]. Modulating mitochondrial metabolism helps reduce synovial inflammation and slow down the progression of early OA [47]. In addition, the imbalance of mitochondrial metabolic homeostasis in OA chondrocytes is also manifested as abnormal mitochondrial biogenesis, a process that is regulated by various transfer factors such as PGC-1a and AMPK [48]. Correcting abnormalities in mitochondrial biogenesis is also important for the treatment of OA [49].

Aberrant Mitochondrial Morphology and Function

MT are crucial for preserving cellular function. Normal MT are oval in shape with evenly distributed intimal ridges; however, the MT in OA chondrocytes are swollen and spherical in shape, and the intimal ridges are irregularly arranged [51]. These morphological changes are accompanied by a decrease in mitochondrial membrane potential and reduced ATP production, which is an intuitive manifestation of mitochondrial dysfunction [29]. Shen et al. [52] proposed that the AMPK–sirtuin 1(SIRT3) loop has a crucial impact on controlling the advancement and growth of OA, in part by adjusting the quality of chondrocyte MT. Sun-Li Hu et al. [53] also pointed out that preventing mitochondrial fragmentation and reshaping mitochondrial morphology can effectively reduce chondrocyte apoptosis and improve cartilage degradation. In addition, controlling the creation of new MT is essential to preserve their function. Studies indicate a decline in mtDNA levels and a loss of important regulators of mitochondrial biogenesis in OA, such as PGC-1α and mitochondrial transcriptional factor A (TFAM), and reversal of this event can inhibit OA progression [33].

Calcium Dysregulation

Calcium is crucial for cell function and acts as a second messenger in various signalling pathways, controlling processes such as cell growth, contraction, and gene expression. Cells possess the ability to sense alterations in intracellular calcium (Ca2+) levels, including amplitude, duration, frequency, and localisation, and respond appropriately to uphold calcium homeostasis and to mitigate cellular harm [54]. The Ca2 + influx and efflux rates between MT must be balanced. An overabundance of Ca2 + can lead to the production of ROS, mitochondrial depolarisation, impairment of mitochondrial membrane potential, and apoptosis [55]. Maintaining intracellular Ca2 + levels involves the transport of calcium into MT using the mitochondrial calcium uniporter (MCU) and the release of Ca2 + through different pathways such as the inositol-1,4,5-trisphosphate receptor (IP3R), the sodium/calcium exchanger, and the mitochondrial permeability transition pore (mPTP) [56]. Abnormal Ca2 + accumulation within the mitochondrial matrix can activate the mPTP, a substantial channel located in the IMM that responds to elevated Ca2 + levels and ROS [57]. The opening of the mPTP results in the depolarisation of the mitochondrial membrane, causing MT to swell and release calcium and Cyt-C, thereby initiating the apoptotic pathway [57]. Early studies found that calcium balance in OA cartilage tissue is dysregulated [58]. Subsequently, Huser et al. [60] directly confirmed that Ca2 + signalling is the key to the mechanical impact of OA. In addition, Zhai et al. [61] reported that the mitochondrial Ca2 + level of bone marrow–derived MSCs (BM-MSCs) derived from the subchondral bone in patients with OA was also significantly higher than in the normal group. When this Ca2 + overload was corrected, OA was improved.

MT-Targeting Drugs for OA Treatment

MT are essential to produce energy and to maintain balance within cells, and they may also help regulate cell death to prevent damage to cartilage in joints. Addressing mitochondrial dysfunction represents a hopeful approach to managing OA [62]. Consequently, the development of novel drugs and methodologies centred on repairing and/or restoring mitochondrial function is imperative for the management of OA. Table 1 lists some drugs that have shown promising effects in restoring mitochondrial dysfunction in OA. They include antioxidants, enhancers of mitochondrial biogenesis, regulators of mitochondrial dynamics, and calcium balance stabilisers.

Table 1 Treatment strategies for mitochondrial dysfunction

Antioxidants

Appropriate antioxidant strategies aimed at reducing the ROS generated by MT are crucial to protect chondrocytes from oxidative stress damage. Research has shown that compounds such as melatonin and quercetin exhibit efficacy in reducing mtROS accumulation, thereby preventing the deterioration of mitochondrial membrane potential and the release of mitochondrial Cyt-C [63]. Additionally, nanomaterials specifically targeting mtROS have also demonstrated the potential to improve mitochondrial dysfunction [72].

Mitochondrial Biogenesis Enhancers

Mitochondrial biogenesis, a process involving the self-renewal and replication of MT regulated by numerous genes, exhibits abnormalities in OA [48]. Activation of the AMPK/SIRT1/PGC-1a pathway has been shown to boost the generation of MT and to reduce oxidative stress, thereby regulating mitochondrial function and ameliorating OA [33]. Previous studies have indicated that certain compounds, such as puerarin and apple procyanidins, can stimulate the production of new MT in chondrocytes, leading to enhanced mitochondrial functionality, and may serve as promising therapeutic agents for the management of OA [65].

Regulators of Mitochondrial Dynamics

The intricate interplay between mitochondrial fission, fusion, and mitophagy is essential for the preservation of mitochondrial functionality. Perturbation of mitochondrial dynamics is observed in chondrocytes during the pathogenesis of OA [73]. The administration of the mitochondrial inhibitor Mdivi-1 has been demonstrated to attenuate mitochondrial fission-induced damage, leading to a reduction in chondrocyte apoptosis [67]. However, the complexes that promote mitochondrial fusion are less studied and deserve further attention [74]. Besides, removing damaged MT to maintain mitochondrial health has received widespread attention. β-Hydroxybutyrate and protocatechuic aldehyde have been shown to enhance chondrocyte mitophagy, promote the clearance of damaged MT, effectively improve mitochondrial function, and inhibit cartilage degradation in OA [68].

Calcium Balance Stabilisers

In the physiological state, MT regulate calcium homeostasis to ensure the health of cartilage [75]. An imbalance in calcium homeostasis, leading to mitochondrial damage and a series of changes, is considered crucial in chondrocyte apoptosis and cartilage degradation [60]. Research suggests that cyclosporin A and B-type natriuretic peptide can inhibit mPTP opening, protecting MT from Ca2 + overload–induced damage [70]. Recently, Zhai et al. [61] synthesised TMA-MSN-TPP-EGTA-PEG (METP) nanoparticles using a composite shell of silica nanoparticles, tetraethylene glycol, and triphenylphosphine. It captured the Ca2 + around the MT of mesenchymal stem cells (MSCs) and effectively treated OA [61]. Similarly, Lin [76] found that regulating calcium homeostasis helps slow down the degeneration of intervertebral discs.

Mitochondrial Transfer in OA Treatment

Pharmacological modulation of mitochondrial dysfunction is pivotal for the treatment of MT-related diseases, driving significant advancements in mitochondrial medicine and providing valuable insights. However, the development of mitochondrial-targeted therapies is confronted with substantial challenges due to the subcellular localization and complex structure of MT. Therapeutic agents must navigate numerous physiological barriers to reach the target cells and subsequently the MT. Nevertheless, even upon approaching the vicinity of the mitochondria, the highly folded and compartmentalized nature of the inner mitochondrial membrane (IMM) presents a significant obstacle for drug molecules seeking entry [77]. In addition, these drugs often face challenges in achieving effective concentration at lesion sites and within MT due to in vivo barriers and poor selectivity. In particular, the non-selective biodistribution of drugs is a primary factor contributing to suboptimal drug concentrations in targeted organs or tissues [78]. Therefore, there is an urgent need for research and development of novel therapies specifically targeting MT.

Recently, intercellular mitochondrial transfer between mammalian cells has been observed in vitro and in vivo, offering a potential universal solution for treating mitochondrial deficiency of different aetiologies [79]. This mitochondrial transfer facilitates the recovery of damaged cells, enhances OXPHOS, elevates ATP synthesis, and restores mitochondrial functionality [81]. It works in various ways, including reducing oxidative stress [82], promoting mitochondrial fusion [83], and regulating mitophagy [84], among others. As a result of these findings, researchers have begun to focus on the role that mitochondrial transfer plays in disease and explored a variety of new methods and technologies for mitochondrial transfer. Three different approaches – stem cell-based mitochondrial transfer [85], direct transfer of isolated MT [87], and transfer of extracellular vesicle (EV)-encapsulated MT [88] are discussed here as potential ways to ameliorate mitochondrial damage for the therapeutic management of OA (Fig. 2).

Fig. 2
figure 2

Mechanism of mitochondrial transfer. (1) Mitochondria (are transferred from mesenchymal stem cells to recipient cells in four ways: (a) extracellular vesicles, (b) tunnelling nanotubes, (c) gap junctions, and (d) cell fusion. (2) Transferring isolated mitochondria directly. (3) Transferring EV-encapsulated mitochondria directly

MSC-Mediated Mitochondrial Transfer

Stem cells, as the most undifferentiated cells at the apex of the cellular lineage, exhibit a remarkable ability for differentiation and self-renewal. Furthermore, they possess the capacity to differentiate into a multitude of tissues, organs, or specialised cells within the human body, thus presenting significant potential for applications in engineering and regenerative medicine. Various sources of MSC treatments have demonstrated the ability to inhibit, halt, or potentially reverse cartilage degradation in animal models [89]. In a recent double-blind, randomised phase IIb clinical trial, the authors demonstrated that patients receiving a single injection of adipose-derived MSCs (AD-MSCs) exhibited notable enhancements in their Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) scores at the 6-month mark, in contrast to the control cohort. Furthermore, there was a deceleration in joint wear progression [92]. The conclusions reached by Sadri et al. [93] were consistent with these results. However, a thorough comprehension of the mechanisms governing MSC therapy for OA is lacking.

Recent research has validated the ability of stem cells to transfer MT to neighbouring cells, resulting in the restoration of cellular respiration, the initiation of cell reprogramming, and ultimately the repair and enhancement of cellular function (Table 2). Undifferentiated MSCs exhibit decreased energy requirements in the glycolytic state, making them promising candidates for mitochondrial transfer [94]. In addition, MSCs possess an exceptional ability to home to diseased tissues, facilitating targeted mitochondrial transfer. These characteristics – combined with the immune privilege, low levels of oxidative damage, and tightly regulated redox balance of MSCs – position them as optimal donor cells for the selective delivery of healthy MT to diseased cells.

Notably, this transfer process is more common in harmful environments than in healthy cells. For example, when exposed to damaged cartilage tissue, MSCs position themselves in the area of matrix injury and extend their MT into chondrocytes located deep within microcracks. In contrast, few cells accumulate in uninjured cartilage [95]. During this process, cytokines and dysfunctional MT are released from injured cells. During this process, cytokines and mtDNA are released from injured cells, serving as indicators of potential damage. These signals stimulate MSCs to transfer their functional MT to aid in the recovery of the impaired cells [96]. On the other hand, a damaged environment will promote the production of more mitochondrial transfer channels, such as tunneling nanotubes (TNTs) [99].

The significance of mitochondrial transfer in the therapeutic efficacy of stem cells for injured tissues is increasingly acknowledged. Although MT are not the primary energy source for chondrocytes, the phenomenon of mitochondrial dysfunction and mitochondrial transfer in OA chondrocytes has attracted research attention [95]. In a co-culture system involving BM-MSCs and osteoarthritic chondrocytes, Wang et al. [51] observed that MSCs promoted the recovery of the chondrocyte mitochondrial membrane potential and increased ATP by transferring their own healthy MT, ultimately reducing the apoptotic rate of chondrocytes and enhancing the function of OA chondrocytes. This finding highlights the role of mitochondrial transfer in OA treatment.

Table 2 Applications of mitochondrial transfer in various systemic diseases

MSCs transfer MT through various mechanisms, such as TNTs, gap junction channels (GJCs), and EVs [97], as well as cell fusion (Fig. 3) [110]. TNTs are membrane-bound cellular conduits capable of directly transferring various cellular components such as endocytic vesicles, lysosomes, MT, and membrane-bound proteins from cell to cell [111]. Hsu et al. [112] utilised anti-mitochondrial antibodies and MitoTracker along with laser scanning confocal microscopy to study the transmission of MT from human BM-MSCs to mouse liver cells via TNTs. GJCs, which are formed by connexins (CXs), serve as a significant mode of intercellular communication. These pathways enable the transfer of ions and small compounds, such as Ca2+, inositol trisphosphate, cyclic nucleotides, and oligonucleotides, which help to synchronise cellular activities throughout various multicellular tissues [113]. The application of the GJC enhancer retinoic acid resulted in significant augmentation in the quantity of MT transferred from BM-MSCs to neurons. Conversely, the GJC inhibitor 18β-glycyrrhizic acid (18β-GA) reduced mitochondrial transfer. The results indicate that GJCs play a vital role in enabling the movement of MT from stem cells to neurons [81]. Specifically, connexin43 (CX43) is a critical CX protein involved in the establishment of GJCs [97]. Increasing CX43 expression with iron oxide nanoparticles has been shown to improve GJC function and to boost the mitochondrial transfer rate. Conversely, the suppression of CX43 expression eliminates this effect [114]. Moreover, recent reports have demonstrated that the upregulation of CX43 is associated with augmented formation of TNTs, while the employment of short hairpin RNA (shRNA) to suppress CX43 yields contrasting outcomes, suggesting that CX43 also assumes a crucial function in facilitating TNT formation. However, the precise regulatory mechanism remains elusive [115]. Additionally, Miro1 and Miro2, which are two types of Rho-GTPases, interact with other accessory proteins to move MT along the TNTs that connect the two cells [117]. Miro1 upregulation has been shown to enhance mitochondrial transfer by MSCs in cases of myocardial disease [118]. On the other hand, reducing Miro1 expression hinders the development of TNTs, thus blocking the transport of MT from MSCs to endothelial cells [119]. Miro2s participation in the mitochondrial transfer process is also significant [120]. Previous studies have indicated that mitochondrial migration can also occur via EVs, such as exosomes [51]. Phinney et al. [121] discovered that MSCs possess the ability to transfer their own MT into EVs, thereby facilitating the transportation of intact MT or mtDNA to macrophages. This process enhances the bioenergy of macrophages and provides additional evidence supporting the notion that EVs serve as carriers for MT. Additionally, the direct acquisition of MT through cell fusion represents the most straightforward approach [122].

Stem cell injection has been used in the clinical treatment of orthopaedic diseases [124]. However, it is imperative to acknowledge the potential risks associated with stem cell transplantation, including tumourigenicity and immunogenicity. Multiple stem cell types possess the property of tumour tropism [125]. Furthermore, stem cells exhibit a diverse array of surface antigens, such as HLA class 1 antigens, that are absent on the mitochondrial membrane. Consequently, stem cells possess a higher degree of immunogenicity compared with isolated MT [116]. Another issue that needs to be considered is that the mitochondrial transfer efficiency of stem cell therapy is low. Hence, finding a way to improve the transfer rate is the key to enhancing its efficacy.

Fig. 3
figure 3

Laser confocal imaging of mitochondrial transfer. Mesenchymal stems cell transfer mitochondria (red) to chondrocytes (green cytoplasm, blue nuclei) using different methods: (i) extracellular vesicles, (ii) gap junctions, (iii) tunnelling nanotubes, and (iv and v) cell fusion. Reproduced from a previous publication [95], with permission from the authors

Isolated Exogenous Mitochondrial Transfer

Limitations of cell-to-cell MT transfer encompass variability in cell phenotypes, low engraftment and retention rates, and inconsistent clinical outcomes. Therefore, non-contact MT transfer methods are currently being investigated. The phenomenon of intercellular mitochondrial transfer has led researchers to hypothesize that MT may possess the capability to invade cells, and various methods to transfer MT to the recipient cell artificially have been developed. In a seminal study, Tachibana et al. [127] successfully introduced healthy mitochondria into oocytes containing mutated mtDNA, demonstrating potential for the treatment of human genetic mitochondrial disorders and laying the groundwork for further research on mitochondrial transfer. Similarly, Li et al. [81] documented the internalisation of isolated MT in motor neurons following co-culturing under hypoxic conditions for 30 min. This process was concomitant with an elevation in ATP levels and the mitochondrial membrane potential and enhanced neuronal viability. Additionally, the extent of internalisation correlated directly with the concentration of co-cultured MT. Masuzawa et al. [128] validated this effect in in vivo experiments. They isolated MT from the chest muscles of New Zealand white rabbits and immediately injected them into ischaemic hearts. The findings indicated that cardiac cells internalised these MT within 2–8 h of transplantation, leading to increased oxygen consumption, synthesis of high-energy phosphates, and activation of cytokine mediators and protein pathways, ultimately shielding the heart from damage caused by ischaemia–reperfusion. Recently, direct transplantation of MT has been applied to cartilage repair in OA. Kim et al. [129] synthesised fusogenic liposomes encapsulating MT, assisting in their delivery to chondrocytes. Experiments conducted in a lab setting and within living organisms have shown that the use of fusogenic liposomes accelerates and improves mitochondrial transfer, offering a promising approach for enhancing cartilage repair. Inspired by this, researchers have designed different methods in the hope of increasing isolated mitochondrial transfer efficiency (Fig. 4).

Fig. 4
figure 4

An illustration of methods to increase the transfer rate of isolated mitochondria. (A) Promoting internalisation of isolated mitochondria by centrifugal force [130]. (B) Mitochondria are encapsulated in synthetic liposomes to enhance the delivery efficiency [129]. (C) Dextran was conjugated with TPP as carriers to increase mitochondrial delivery efficiency [131]. (D) Pep-1 peptide binds to mitochondria to enhance delivery efficiency [132]. Each panel has been reproduced from the respective publication, with permission from the authors

Although mitochondrial transfer has shown numerous benefits for cells, the technology is still in its early stages and encounters various obstacles. It is believed that damaged and dysfunctional MT may not provide benefits to host cells and could potentially cause harm [128]. Therefore, it is necessary to obtain fresh, intact, and respiratory-active MT to effectively exert therapeutic effects. Additionally, facilitating the successful entry of MT into recipient cells in adequate amounts and ensuring their complete utilisation presents a significant challenge. In addition to the methods shown in Fig. 4, technologies such as MitoCeption [134] and magnetic nanoparticles [135] have been designed to increase the efficiency of isolated mitochondrial transfer. Nevertheless, the application of these technologies in vivo necessitates additional investigation. Lastly, exogenous MT may selectively degrade after mitochondrial transfer and disappear within a week [136]. Therefore, more complex, minimally invasive methods are needed to isolate fully functional MT from cell extracts. Additionally, less invasive delivery methods need to be developed to fully exploit the beneficial effects of mitochondrial transfer.

EV-Encapsulated Mitochondrial Transfer

Recent research has identified MT-specific cargoes, including DNA, RNA, and proteins in EVs derived from various cell types, such as fibroblasts, neurons, and MSCs [137]. EVs originate from the cell membrane and can be classified based on their size as exosomes, microvesicles, and apoptotic bodies. These vesicles have the ability to transfer their cargo into the cytoplasm of recipient cells, thereby facilitating intercellular communication and modulating the physiological state of the receiving cells [138]. Functioning as pivotal agents in intercellular communication, EVs possess the capacity to selectively bind to particular cells or tissues through receptor-mediated mechanisms, subsequently releasing their contents into the corresponding target structures [139]. This attribute enables EVs to potentially serve as rescuers for recipient cells, while simultaneously preserving the homeostasis of the originating cell. As EV extraction technology has matured, the application of EVs has become increasingly widespread (Fig. 5).

Fig. 5
figure 5

Isolation methods to obtain extracellular vesicles. Reproduced from a previous publication [140], with permission from the authors

The efficacy of stem cell–derived EVs in the treatment of OA has been proven, and its mode of action is diverse. In a study using mice, the communication between EVs and methyltransferase-like 3(METTL3) results in decreased methylation of Nod-like receptor pyrin domain 3 (NLRP3) mRNA in macrophages, ultimately easing the symptoms of OA in the knee joint [141]. In addition, EVs have the ability to transport microRNAs (miRNA), which effectively inhibits chondrocyte apoptosis [142]. Furthermore, experiments conducted in living organisms and in a controlled environment demonstrate that the use of exosomes can effectively reduce the levels of MMP-13 and a disintegrin and metalloproteinase with thrombospondin motifs 5(ADAMTS-5), thus preventing the degradation of cartilage [143]. However, further investigation is required to fully understand the precise mechanisms underlying EV therapy. Recently, researchers have found that EVs transfer their own MT and mtDNA to recipient cells during the treatment of diseases [144]. In the context of ischaemic stroke, D’Souza et al. [145] revealed that microvesicles, which act as carriers of MT, have the ability to transmit functional MT to chemically impaired brain endothelial cells, thereby enhancing their chances of survival. Similarly to TNT-mediated mitochondrial transfer, this transfer process is less frequent in endothelial cells that are in a healthy state. The utilisation of exosomes derived from AD-MSCs for the treatment of acute lung injury has also been shown to effectively transfer MT, thereby alleviating airway metabolic disturbances [146]. This finding aligns with the conclusions drawn by Zhang et al. [147] in their study on Alzheimer’s disease. Thomas et al. [137] validated that MSCs can package functional MT into EVs and deliver them to chondrocytes, which holds great promise for the treatment of OA.

The utilisation of EVs as vehicles for mitochondrial transfer presents a potential solution to address certain inherent constraints associated with MSC therapy [148]. Moreover, research has indicated that EVs, upon transporting MT, exhibit minimal co-localisation with lysosomes and peroxisomes within recipient cells, thereby diminishing the degradation of exogenous MT [149]. Although this phenomenon has only been illustrated preliminarily, it offers valuable insights for future investigations in the field of mitochondrial therapy. The present obstacle pertains to the insufficiency of MT within EVs, despite their presence. It is imperative to facilitate the incorporation of a greater quantity of MT into EVs by stem cells. Preliminary investigations have substantiated that pre-treating donor cells can augment their survival rate and therapeutic efficacy [150]. In a study focused on obesity, Crewe et al. [151] discovered that elevated energy stress within adipocytes leads to an increased presence of MT enclosed within small EVs (sEVs). These sEVs subsequently migrate to the heart, augmenting its adaptability to cardiac conditions. Recently, novel methodologies have been established for the subfractionation of EV subtypes, allowing for the selective isolation of vesicles containing intact mitochondrial components such as MT, mitochondrial proteins, and mtDNA. These specialised vesicles, known as mitovehicles, appear to provide a more effective means of facilitating mitochondrial transfer [152]. Nevertheless, there have been few studies in this area, and the technology for extracting MT-rich EVs is limited. Hence, there is still a long way to go before this technique can be translated to the clinic.

Conclusion and perspectives

In the pathogenesis of OA, chondrocytes frequently experience mitochondrial dysfunction, resulting in compromised energy metabolism and subsequent cascades of oxidative stress and calcium imbalance following mitochondrial injury. These events significantly impact cell viability. The advancement of mitochondrial repair therapy has exhibited encouraging outcomes in the reversal of mitochondrial dysfunction, offering novel insights for OA treatment.

In addition to conventional pharmacological repair, mitochondrial transfer has emerged as a promising therapeutic strategy, showing significant advantages in restoring mitochondrial function. There are three established techniques for mitochondrial transfer: stem cell-mediated transfer, isolated exogenous transfer, and EV-encapsulated transfer. Each option has its own advantages and disadvantages. Researchers have conducted extensive research to better exploit the advantages of these solutions. Several studies have proposed that pre-treating stem cells could enhance therapeutic efficacy. For example, Guo et al. [20] isolated MTs from donor BM-MSCs and then transplanted them into BM-MSCs of the same batch and generation. BM-MSCs that underwent autologous mitochondrial transplantation exhibited enhanced bone defect repair capabilities. Specific pharmaceutical agents, including metformin [154], pioglitazone [155], and adiponectin [156], have the ability to activate PGC-1α and AMPK, thereby stimulating mitochondrial biogenesis and increasing the mitochondrial reserves of stem cells. Furthermore, some nanoparticles such as platinum [157], silica [158], and iron oxide [159] have been shown to upregulate CX43 expression and to increase the release of EVs, thereby promoting more effective mitochondrial transfer. On the other hand, strategies to develop advanced technologies to extract higher-quality isolated MTs or MT-rich EVs and enhance efficient mitochondrial delivery are the focus of research. Moreover, in future clinical applications, these technologies face a common problem. Cartilage ECM is characterised by low cellularity and high density, and its small pore size and high charge properties have been shown to impede the diffusion of large particles, including antibodies and other experimental biological factors [161]. This leads to the limitation of mitochondrial transfer therapy, that is, the therapeutic effect on deep-seated chondrocytes will be weaker than that of superficial cells.

Mitochondrial repair technology presents significant potential for the management of OA. The conception of mitochondrial transfer has garnered significant attention in recent years. While the mechanism of mitochondrial transfer remains poorly understood, it holds significant therapeutic promise. In future research, it is necessary to elucidate the molecular and cellular mechanisms involved in mitochondrial transfer and to develop efficient methods for the extraction and delivery of MT, in order to advance their clinical utilisation.

Data availability

Due to its nature as a review article, all references are published articles. The data underlying this article are available in the Pubmed.

Abbreviations

ATP:

adenosine triphosphate

AD-MSCs:

adipose-derived MSCs

ADAMTS 5:

a disintegrin and metalloproteinase with thrombospondin motifs 5

AMPK:

AMP activated protein kinase

BM-MSCs:

bone marrow-derived MSCs

18β-GA:

18β-glycyrrhizic acid

CX43:

connexin43

Cyt-C:

cytochrome c

Drp1:

dynamin-related protein 1

Dnm2:

dynamin 2

2,4-DNP:

2,4-dinitrophenol

EVs:

extracellular vesicles

ECM:

extracellular matrix

GJCs:

gap junction channels

IP3R:

inositol-1,4,5-trisphosphate receptor

IL-1β:

interleukin-1 beta

IMM:

inner mitochondrial membrane

METTL3:

methyltransferase-like 3

OA:

Osteoarthritis

MMPs:

matrix metalloproteinases

Mfn1/2:

mitofusins 1/2

MSC:

mesenchymal stem cell

METP:

TMA-MSN-TPP-EGTA-PEG

mtROS:

mitochondrial ROS

mtDNA:

mitochondrial DNA

mTOR:

mechanistic target of rapamycin

miRNA:

microRNA

NO:

nitric oxide

NF-κB:

factor-κB

NLRP3:

Nod-like receptor pyrin domain 3

MT:

mitochondria

OXPHOS:

oxidative phosphorylation

OMM:

outer mitochondrial membrane

ROS:

reactive oxygen species

SIRT3:

sirtuin 1

shRNA:

short hairpin RNA

TFAM:

mitochondrial transcriptional factor A

TNF-α:

tumour necrosis factor-alpha

WOMAC:

Western Ontario and McMaster Universities Osteoarthritis Index

References

  1. Luksameesate P, Tanavalee A, Taychakhoonavudh S. An economic evaluation of knee osteoarthritis treatments in Thailand. Front Pharmacol. 2022;13:926431.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Kraus VB, et al. Call for standardized definitions of osteoarthritis and risk stratification for clinical trials and clinical use. Osteoarthritis Cartilage. 2015;23(8):1233–41.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Culvenor AG, et al. Prevalence of knee osteoarthritis features on magnetic resonance imaging in asymptomatic uninjured adults: a systematic review and meta-analysis. Br J Sports Med. 2019;53(20):1268–78.

    Article  PubMed  Google Scholar 

  4. Friedman JR, Nunnari J. Mitochondrial form function Nat. 2014;505(7483):335–43.

    CAS  Google Scholar 

  5. Blanco FJ, Rego I, Ruiz-Romero C. The role of mitochondria in osteoarthritis. Nat Rev Rheumatol. 2011;7(3):161–9.

    Article  PubMed  CAS  Google Scholar 

  6. Ruiz-Romero C, et al. Mitochondrial dysregulation of osteoarthritic human articular chondrocytes analyzed by proteomics: a decrease in mitochondrial superoxide dismutase points to a redox imbalance. Mol Cell Proteom. 2009;8(1):172–89.

    Article  CAS  Google Scholar 

  7. Chen W, Zhao H, Li Y. Mitochondrial dynamics in health and disease: mechanisms and potential targets. Signal Transduct Target Ther. 2023;8(1):333.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Chang X, et al. Therapeutic strategies in ischemic cardiomyopathy: Focus on mitochondrial quality surveillance. EBioMedicine. 2022;84:104260.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Linnane AW, et al. Mitochondrial DNA mutations as an important contributor to ageing and degenerative diseases. Lancet. 1989;1(8639):642–5.

    Article  PubMed  CAS  Google Scholar 

  10. Fearon U, et al. Hypoxia, mitochondrial dysfunction and synovial invasiveness in rheumatoid arthritis. Nat Rev Rheumatol. 2016;12(7):385–97.

    Article  PubMed  CAS  Google Scholar 

  11. Qi Z, et al. The role and intervention of mitochondrial metabolism in osteoarthritis. Mol Cell Biochem. 2024;479(6):1513–24.

    Article  PubMed  CAS  Google Scholar 

  12. Molnar V et al. Cytokines and Chemokines Involved in Osteoarthritis Pathogenesis. Int J Mol Sci, 2021. 22(17).

  13. Sandell LJ, Aigner T. Articular cartilage and changes in arthritis. An introduction: cell biology of osteoarthritis. Arthritis Res. 2001;3(2):107–13.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Lee RB, Urban JP. Evidence for a negative Pasteur effect in articular cartilage. Biochem J, 1997. 321 (Pt 1)(Pt 1): pp. 95–102.

  15. Charlier E et al. Insights on Molecular Mechanisms of Chondrocytes Death in Osteoarthritis. Int J Mol Sci, 2016. 17(12).

  16. Wu X, et al. Dysregulated energy metabolism impairs chondrocyte function in osteoarthritis. Osteoarthritis Cartilage. 2023;31(5):613–26.

    Article  PubMed  CAS  Google Scholar 

  17. Chen P, et al. A plant-derived natural photosynthetic system for improving cell anabolism. Nature. 2022;612(7940):546–54.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Durán-Sotuela A, et al. A meta-analysis and a functional study support the influence of mtDNA variant m.16519 C on the risk of rapid progression of knee osteoarthritis. Ann Rheum Dis. 2023;82(7):974–84.

    Article  PubMed  Google Scholar 

  19. Chen P, et al. Desktop-stereolithography 3D printing of a radially oriented extracellular matrix/mesenchymal stem cell exosome bioink for osteochondral defect regeneration. Theranostics. 2019;9(9):2439–59.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Guo Y, et al. Mitochondria transfer enhances proliferation, migration, and osteogenic differentiation of bone marrow mesenchymal stem cell and promotes bone defect healing. Stem Cell Res Ther. 2020;11(1):245.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Zhong G, Madry H, Cucchiarini M. Mitochondrial Genome Editing to Treat Human Osteoarthritis-A Narrative Review. Int J Mol Sci, 2022. 23(3).

  22. Hu S, et al. Stabilization of HIF-1α alleviates osteoarthritis via enhancing mitophagy. Cell Death Dis. 2020;11(6):481.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Riegger J, et al. Oxidative stress as a key modulator of cell fate decision in osteoarthritis and osteoporosis: a narrative review. Cell Mol Biol Lett. 2023;28(1):76.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Coryell PR, Diekman BO, Loeser RF. Mechanisms and therapeutic implications of cellular senescence in osteoarthritis. Nat Rev Rheumatol. 2021;17(1):47–57.

    Article  PubMed  Google Scholar 

  25. D’Amico D, et al. Urolithin A improves mitochondrial health, reduces cartilage degeneration, and alleviates pain in osteoarthritis. Aging Cell. 2022;21(8):e13662.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Kim D, Song J, Jin EJ. BNIP3-Dependent Mitophagy via PGC1α Promotes Cartilage Degradation. Cells, 2021. 10(7).

  27. Qi Z, et al. The role and intervention of mitochondrial metabolism in osteoarthritis. Mol Cell Biochem; 2023.

  28. Mao X, et al. Mitochondria: Potential Targets for Osteoarthritis. Front Med (Lausanne). 2020;7:581402.

    Article  PubMed  Google Scholar 

  29. Kan S, et al. Role of Mitochondria in Physiology of Chondrocytes and Diseases of Osteoarthritis and Rheumatoid Arthritis. Cartilage. 2021;13(2suppl):s1102–21.

    Article  Google Scholar 

  30. Rajpurohit R, et al. Chondrocyte death is linked to development of a mitochondrial membrane permeability transition in the growth plate. J Cell Physiol. 1999;179(3):287–96.

    Article  PubMed  CAS  Google Scholar 

  31. Martin JA, et al. Mitochondrial electron transport and glycolysis are coupled in articular cartilage. Osteoarthritis Cartilage. 2012;20(4):323–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Wolff KJ, et al. Mechanical stress and ATP synthesis are coupled by mitochondrial oxidants in articular cartilage. J Orthop Res. 2013;31(2):191–6.

    Article  PubMed  CAS  Google Scholar 

  33. Wang Y, et al. Mitochondrial biogenesis is impaired in osteoarthritis chondrocytes but reversible via peroxisome proliferator-activated receptor γ coactivator 1α. Arthritis Rheumatol. 2015;67(8):2141–53.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Mammucari C, Rizzuto R. Signaling pathways in mitochondrial dysfunction and aging. Mech Ageing Dev. 2010;131(7–8):536–43.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Wu CL, et al. The role of macrophages in osteoarthritis and cartilage repair. Osteoarthritis Cartilage. 2020;28(5):544–54.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Yao Q, et al. Osteoarthritis: pathogenic signaling pathways and therapeutic targets. Signal Transduct Target Ther. 2023;8(1):56.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Venditti P, Di Stefano L, Di Meo S. Mitochondrial metabolism reactive oxygen species Mitochondrion. 2013;13(2):71–82.

    PubMed  CAS  Google Scholar 

  38. Chan DC. Mitochondrial Dynamics and Its Involvement in Disease. Annu Rev Pathol. 2020;15:235–59.

    Article  PubMed  CAS  Google Scholar 

  39. Pagliuso A, Cossart P, Stavru F. The ever-growing complexity of the mitochondrial fission machinery. Cell Mol Life Sci. 2018;75(3):355–74.

    Article  PubMed  CAS  Google Scholar 

  40. Westermann B. Mitochondrial fusion and fission in cell life and death. Nat Rev Mol Cell Biol. 2010;11(12):872–84.

    Article  PubMed  CAS  Google Scholar 

  41. An F, et al. New insight of the pathogenesis in osteoarthritis: the intricate interplay of ferroptosis and autophagy mediated by mitophagy/chaperone-mediated autophagy. Front Cell Dev Biol. 2023;11:1297024.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Gao S, Hu J. Mitochondrial Fusion: The Machineries In and Out. Trends Cell Biol. 2021;31(1):62–74.

    Article  PubMed  CAS  Google Scholar 

  43. Wang FS et al. Irisin Mitigates Oxidative Stress, Chondrocyte Dysfunction and Osteoarthritis Development through Regulating Mitochondrial Integrity and Autophagy. Antioxid (Basel), 2020. 9(9).

  44. Kuznetsov AV et al. The Complex Interplay between Mitochondria, ROS and Entire Cellular Metabolism. Antioxid (Basel), 2022. 11(10).

  45. Yi D, et al. AMPK Signaling in Energy Control, Cartilage Biology, and Osteoarthritis. Front Cell Dev Biol. 2021;9:696602.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Zheng L, et al. The role of metabolism in chondrocyte dysfunction and the progression of osteoarthritis. Ageing Res Rev. 2021;66:101249.

    Article  PubMed  CAS  Google Scholar 

  47. Zhang L, et al. Reprogramming Mitochondrial Metabolism in Synovial Macrophages of Early Osteoarthritis by a Camouflaged Meta-Defensome. Adv Mater. 2022;34(30):e2202715.

    Article  PubMed  Google Scholar 

  48. Wang H, et al. PGC-1α in osteoarthritic chondrocytes: From mechanism to target of action. Front Pharmacol. 2023;14:1169019.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Gao SJ, et al. Dimethyl Fumarate Attenuates Pain Behaviors in Osteoarthritis Rats via Induction of Nrf2-Mediated Mitochondrial Biogenesis. Mol Pain. 2022;18:17448069221124920.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Ajmal I, et al. Isoprenaline and salbutamol inhibit pyroptosis and promote mitochondrial biogenesis in arthritic chondrocytes by downregulating β-arrestin and GRK2. Front Pharmacol. 2022;13:996321.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Wang R, et al. Mitochondrial transfer from bone-marrow-derived mesenchymal stromal cells to chondrocytes protects against cartilage degenerative mitochondrial dysfunction in rats chondrocytes. Chin Med J (Engl). 2020;134(2):212–8.

    Article  PubMed  Google Scholar 

  52. Chen Y, et al. Mechanistic insights into AMPK-SIRT3 positive feedback loop-mediated chondrocyte mitochondrial quality control in osteoarthritis pathogenesis. Pharmacol Res. 2021;166:105497.

    Article  PubMed  CAS  Google Scholar 

  53. Hu SL, et al. TBK1-medicated DRP1 phosphorylation orchestrates mitochondrial dynamics and autophagy activation in osteoarthritis. Acta Pharmacol Sin. 2023;44(3):610–21.

    Article  PubMed  CAS  Google Scholar 

  54. East DA, Campanella M. Ca2 + in quality control: an unresolved riddle critical to autophagy and mitophagy. Autophagy. 2013;9(11):1710–9.

    Article  PubMed  CAS  Google Scholar 

  55. Yin S, et al. Transient receptor potential ankyrin 1 (trpa1) mediates il-1β-induced apoptosis in rat chondrocytes via calcium overload and mitochondrial dysfunction. J Inflamm (Lond). 2018;15:27.

    Article  PubMed  CAS  Google Scholar 

  56. Weiser A, et al. The mitochondrial calcium uniporter (MCU) activates mitochondrial respiration and enhances mobility by regulating mitochondrial redox state. Redox Biol. 2023;64:102759.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Bauer TM, Murphy E. Role of Mitochondrial Calcium and the Permeability Transition Pore in Regulating Cell Death. Circ Res. 2020;126(2):280–93.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Rizzo R, et al. Calcium, sulfur, and zinc distribution in normal and arthritic articular equine cartilage: a synchrotron radiation-induced X-ray emission (SRIXE) study. J Exp Zool. 1995;273(1):82–6.

    Article  PubMed  CAS  Google Scholar 

  59. Burton DW, et al. Chondrocyte calcium-sensing receptor expression is up-regulated in early guinea pig knee osteoarthritis and modulates PTHrP, MMP-13, and TIMP-3 expression. Osteoarthritis Cartilage. 2005;13(5):395–404.

    Article  PubMed  CAS  Google Scholar 

  60. Huser CA, Davies ME. Calcium signaling leads to mitochondrial depolarization in impact-induced chondrocyte death in equine articular cartilage explants. Arthritis Rheum. 2007;56(7):2322–34.

    Article  PubMed  CAS  Google Scholar 

  61. Zhai Q, et al. Nanorepairers Rescue Inflammation-Induced Mitochondrial Dysfunction in Mesenchymal Stem Cells. Adv Sci (Weinh). 2022;9(4):e2103839.

    Article  PubMed  Google Scholar 

  62. Liu D, et al. Mitochondrial quality control in cartilage damage and osteoarthritis: new insights and potential therapeutic targets. Osteoarthritis Cartilage. 2022;30(3):395–405.

    Article  PubMed  CAS  Google Scholar 

  63. Rodella LF, et al. Aging and vascular dysfunction: beneficial melatonin effects. Age (Dordr). 2013;35(1):103–15.

    Article  PubMed  CAS  Google Scholar 

  64. Qiu L, Luo Y, Chen X. Quercetin attenuates mitochondrial dysfunction and biogenesis via upregulated AMPK/SIRT1 signaling pathway in OA rats. Biomed Pharmacother. 2018;103:1585–91.

    Article  PubMed  CAS  Google Scholar 

  65. Wang L, et al. Puerarin Attenuates Osteoarthritis via Upregulating AMP-Activated Protein Kinase/Proliferator-Activated Receptor-γ Coactivator-1 Signaling Pathway in Osteoarthritis Rats. Pharmacology. 2018;102(3–4):117–25.

    Article  PubMed  CAS  Google Scholar 

  66. Masuda I, et al. Apple procyanidins promote mitochondrial biogenesis and proteoglycan biosynthesis in chondrocytes. Sci Rep. 2018;8(1):7229.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Ansari MY, Novak K, Haqqi TM. ERK1/2-mediated activation of DRP1 regulates mitochondrial dynamics and apoptosis in chondrocytes. Osteoarthritis Cartilage. 2022;30(2):315–28.

    Article  PubMed  CAS  Google Scholar 

  68. Zhuang H et al. β-Hydroxybutyrate enhances chondrocyte mitophagy and reduces cartilage degeneration in osteoarthritis via the HCAR2/AMPK/PINK1/Parkin pathway. Aging Cell, 2024: p. e14294.

  69. Jie L, et al. Protocatechuic aldehyde attenuates chondrocyte senescence via the regulation of PTEN-induced kinase 1/Parkin-mediated mitochondrial autophagy. Int J Immunopathol Pharmacol. 2024;38:3946320241271724.

    Article  PubMed  Google Scholar 

  70. Ikeda G, et al. Nanoparticle-Mediated Targeting of Cyclosporine A Enhances Cardioprotection Against Ischemia-Reperfusion Injury Through Inhibition of Mitochondrial Permeability Transition Pore Opening. Sci Rep. 2016;6:20467.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Sun Y, et al. B-type natriuretic peptide protects cardiomyocytes at reperfusion via mitochondrial calcium uniporter. Biomed Pharmacother. 2010;64(3):170–6.

    Article  PubMed  CAS  Google Scholar 

  72. Shin HJ, et al. 66shc siRNA Nanoparticles Ameliorate Chondrocytic Mitochondrial Dysfunction in Osteoarthritis. Int J Nanomed. 2020;15:2379–90.

    Article  CAS  Google Scholar 

  73. Chen Q, et al. [Research progress on the role of chondrocyte mitochondrial homeostasis imbalance in the pathogenesis of osteoarthritis]. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi. 2023;37(6):748–57.

    PubMed  Google Scholar 

  74. Cao YL, et al. MFN1 structures reveal nucleotide-triggered dimerization critical for mitochondrial fusion. Nature. 2017;542(7641):372–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Shen X et al. Extracellular Calcium Ion Concentration Regulates Chondrocyte Elastic Modulus and Adhesion Behavior. Int J Mol Sci, 2021. 22(18).

  76. Lin H, et al. Edaravone ameliorates compression-induced damage in rat nucleus pulposus cells. Life Sci. 2017;189:76–83.

    Article  PubMed  CAS  Google Scholar 

  77. Tran VA et al. Dual Stimuli-Responsive Multifunctional Silicon Nanocarriers for Specifically Targeting Mitochondria in Human Cancer Cells. Pharmaceutics, 2022. 14(4).

  78. Vallorz EL, et al. Kidney targeting of formoterol containing polymeric nanoparticles improves recovery from ischemia reperfusion-induced acute kidney injury in mice. Kidney Int. 2022;102(5):1073–89.

    Article  PubMed  CAS  Google Scholar 

  79. Tang LX, et al. Intercellular mitochondrial transfer as a means of revitalizing injured glomerular endothelial cells. World J Stem Cells. 2022;14(9):729–43.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Wei B, et al. Mitochondrial transfer from bone mesenchymal stem cells protects against tendinopathy both in vitro and in vivo. Stem Cell Res Ther. 2023;14(1):104.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Li H, et al. Mitochondrial Transfer from Bone Marrow Mesenchymal Stem Cells to Motor Neurons in Spinal Cord Injury Rats via Gap Junction. Theranostics. 2019;9(7):2017–35.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Jin P et al. Platelets Facilitate Wound Healing by Mitochondrial Transfer and Reducing Oxidative Stress in Endothelial Cells. Oxid Med Cell Longev, 2023. 2023: p. 2345279.

  83. Huang X, et al. Mitochondrial transfer between BMSCs and Müller promotes mitochondrial fusion and suppresses gliosis in degenerative retina. iScience. 2024;27(7):110309.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Bourebaba L, et al. Artificial mitochondrial transplantation (AMT) reverses aging of mesenchymal stromal cells and improves their immunomodulatory properties in LPS-induced synoviocytes inflammation. Biochim Biophys Acta Mol Cell Res. 2024;1871(7):119806.

    Article  PubMed  CAS  Google Scholar 

  85. Doyle EC, Wragg NM, Wilson SL. Intraarticular injection of bone marrow-derived mesenchymal stem cells enhances regeneration in knee osteoarthritis. Knee Surg Sports Traumatol Arthrosc. 2020;28(12):3827–42.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Kim KI, et al. Safety and Efficacy of the Intra-articular Injection of Mesenchymal Stem Cells for the Treatment of Osteoarthritic Knee: A 5-Year Follow-up Study. Stem Cells Transl Med. 2022;11(6):586–96.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Lee AR, et al. Mitochondrial Transplantation Ameliorates the Development and Progression of Osteoarthritis. Immune Netw. 2022;22(2):e14.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Zhang S, et al. MSC exosomes alleviate temporomandibular joint osteoarthritis by attenuating inflammation and restoring matrix homeostasis. Biomaterials. 2019;200:35–47.

    Article  PubMed  CAS  Google Scholar 

  89. Hamilton AM, et al. Iron nanoparticle-labeled murine mesenchymal stromal cells in an osteoarthritic model persists and suggests anti-inflammatory mechanism of action. PLoS ONE. 2019;14(12):e0214107.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Toghraie F, et al. Scaffold-free adipose-derived stem cells (ASCs) improve experimentally induced osteoarthritis in rabbits. Arch Iran Med. 2012;15(8):495–9.

    PubMed  Google Scholar 

  91. Murphy JM, et al. Stem cell therapy in a caprine model of osteoarthritis. Arthritis Rheum. 2003;48(12):3464–74.

    Article  PubMed  Google Scholar 

  92. Lee WS, et al. Intra-Articular Injection of Autologous Adipose Tissue-Derived Mesenchymal Stem Cells for the Treatment of Knee Osteoarthritis: A Phase IIb, Randomized, Placebo-Controlled Clinical Trial. Stem Cells Transl Med. 2019;8(6):504–11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Sadri B, et al. Cartilage regeneration and inflammation modulation in knee osteoarthritis following injection of allogeneic adipose-derived mesenchymal stromal cells: a phase II, triple-blinded, placebo controlled, randomized trial. Stem Cell Res Ther. 2023;14(1):162.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Mohammadalipour A, Dumbali SP, Wenzel PL. Mitochondrial Transfer and Regulators of Mesenchymal Stromal Cell Function and Therapeutic Efficacy. Front Cell Dev Biol. 2020;8:603292.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Fahey M, et al. Mesenchymal stromal cells donate mitochondria to articular chondrocytes exposed to mitochondrial, environmental, and mechanical stress. Sci Rep. 2022;12(1):21525.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Mahrouf-Yorgov M, et al. Mesenchymal stem cells sense mitochondria released from damaged cells as danger signals to activate their rescue properties. Cell Death Differ. 2017;24(7):1224–38.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  97. Islam MN, et al. Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat Med. 2012;18(5):759–65.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Desir S, et al. Tunneling nanotube formation is stimulated by hypoxia in ovarian cancer cells. Oncotarget. 2016;7(28):43150–61.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Wang Y, et al. Tunneling-nanotube development in astrocytes depends on p53 activation. Cell Death Differ. 2011;18(4):732–42.

    Article  PubMed  CAS  Google Scholar 

  100. Moschoi R, et al. Protective mitochondrial transfer from bone marrow stromal cells to acute myeloid leukemic cells during chemotherapy. Blood. 2016;128(2):253–64.

    Article  PubMed  CAS  Google Scholar 

  101. Irwin RM et al. Connexin 43 Regulates Intercellular Mitochondrial Transfer from Human Mesenchymal Stromal Cells to Chondrocytes. bioRxiv, 2024.

  102. Spees JL, et al. Mitochondrial transfer between cells can rescue aerobic respiration. Proc Natl Acad Sci U S A. 2006;103(5):1283–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Acquistapace A, et al. Human mesenchymal stem cells reprogram adult cardiomyocytes toward a progenitor-like state through partial cell fusion and mitochondria transfer. Stem Cells. 2011;29(5):812–24.

    Article  PubMed  CAS  Google Scholar 

  104. Han H, et al. Bone marrow-derived mesenchymal stem cells rescue injured H9c2 cells via transferring intact mitochondria through tunneling nanotubes in an in vitro simulated ischemia/reperfusion model. Mol Med Rep. 2016;13(2):1517–24.

    Article  PubMed  CAS  Google Scholar 

  105. Feng Y, et al. Human Bone Marrow Mesenchymal Stem Cells Rescue Endothelial Cells Experiencing Chemotherapy Stress by Mitochondrial Transfer Via Tunneling Nanotubes. Stem Cells Dev. 2019;28(10):674–82.

    Article  PubMed  CAS  Google Scholar 

  106. Liu K, et al. Mesenchymal stem cells transfer mitochondria into cerebral microvasculature and promote recovery from ischemic stroke. Microvasc Res. 2019;123:74–80.

    Article  PubMed  CAS  Google Scholar 

  107. Wang K, et al. Intercellular mitochondrial transfer alleviates pyroptosis in dental pulp damage. Cell Prolif. 2023;56(9):e13442.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  108. Rustom A, et al. Nanotubular highways for intercellular organelle transport. Science. 2004;303(5660):1007–10.

    Article  PubMed  CAS  Google Scholar 

  109. Yang J et al. Extracellular Vesicles and Cx43-Gap Junction Channels Are the Main Routes for Mitochondrial Transfer from Ultra-Purified Mesenchymal Stem Cells, RECs. Int J Mol Sci, 2023. 24(12).

  110. Torralba D, Baixauli F, Sánchez-Madrid F. Mitochondria Know No Boundaries: Mechanisms and Functions of Intercellular Mitochondrial Transfer. Front Cell Dev Biol. 2016;4:107.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Mittal R, et al. Cell communication by tunneling nanotubes: Implications in disease and therapeutic applications. J Cell Physiol. 2019;234(2):1130–46.

    Article  PubMed  CAS  Google Scholar 

  112. Hsu MJ et al. Mitochondrial Transfer by Human Mesenchymal Stromal Cells Ameliorates Hepatocyte Lipid Load in a Mouse Model of NASH. Biomedicines, 2020. 8(9).

  113. Beyer EC, Berthoud VM. Gap junction gene and protein families: Connexins, innexins, and pannexins. Biochim Biophys Acta Biomembr. 2018;1860(1):5–8.

    Article  PubMed  CAS  Google Scholar 

  114. Huang T, et al. Iron oxide nanoparticles augment the intercellular mitochondrial transfer-mediated therapy. Sci Adv. 2021;7(40):eabj0534.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. Yao Y, et al. Connexin 43-Mediated Mitochondrial Transfer of iPSC-MSCs Alleviates Asthma Inflammation. Stem Cell Rep. 2018;11(5):1120–35.

    Article  CAS  Google Scholar 

  116. Liu Z, et al. Mitochondrial transfer/transplantation: an emerging therapeutic approach for multiple diseases. Cell Biosci. 2022;12(1):66.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  117. Paliwal S, et al. Regenerative abilities of mesenchymal stem cells through mitochondrial transfer. J Biomed Sci. 2018;25(1):31.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Zhang Y, et al. iPSC-MSCs with High Intrinsic MIRO1 and Sensitivity to TNF-α Yield Efficacious Mitochondrial Transfer to Rescue Anthracycline-Induced Cardiomyopathy. Stem Cell Rep. 2016;7(4):749–63.

    Article  CAS  Google Scholar 

  119. Li C et al. Mesenchymal stem cells and their mitochondrial transfer: a double-edged sword. Biosci Rep, 2019. 39(5).

  120. Gao L, et al. Mitochondria Are Dynamically Transferring Between Human Neural Cells and Alexander Disease-Associated GFAP Mutations Impair the Astrocytic Transfer. Front Cell Neurosci. 2019;13:316.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  121. Phinney DG, et al. Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs. Nat Commun. 2015;6:8472.

    Article  PubMed  CAS  Google Scholar 

  122. Ma Z, et al. Mesenchymal stem cell-cardiomyocyte interactions under defined contact modes on laser-patterned biochips. PLoS ONE. 2013;8(2):e56554.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  123. Salaud C, et al. Mitochondria transfer from tumor-activated stromal cells (TASC) to primary Glioblastoma cells. Biochem Biophys Res Commun. 2020;533(1):139–47.

    Article  PubMed  CAS  Google Scholar 

  124. Raza T, et al. Efficacy and Safety of Stem Cell Therapy for Orthopedic Conditions, Including Osteoarthritis and Bone Defects. Cureus. 2024;16(7):e63980.

    PubMed  PubMed Central  Google Scholar 

  125. Goto H, et al. Adipose-derived stem cells enhance human breast cancer growth and cancer stem cell-like properties through adipsin. Oncogene. 2019;38(6):767–79.

    Article  PubMed  CAS  Google Scholar 

  126. Stuckey DW, Shah K. Stem cell-based therapies for cancer treatment: separating hope from hype. Nat Rev Cancer. 2014;14(10):683–91.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Tachibana M, et al. Mitochondrial gene replacement in primate offspring and embryonic stem cells. Nature. 2009;461(7262):367–72.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Masuzawa A, et al. Transplantation of autologously derived mitochondria protects the heart from ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 2013;304(7):H966–82.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  129. Kim HR, et al. Fusogenic liposomes encapsulating mitochondria as a promising delivery system for osteoarthritis therapy. Biomaterials. 2023;302:122350.

    Article  PubMed  CAS  Google Scholar 

  130. Kim MJ, et al. Delivery of exogenous mitochondria via centrifugation enhances cellular metabolic function. Sci Rep. 2018;8(1):3330.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Wu S, et al. Polymer Functionalization of Isolated Mitochondria for Cellular Transplantation and Metabolic Phenotype Alteration. Adv Sci (Weinh). 2018;5(3):1700530.

    Article  PubMed  Google Scholar 

  132. Chang JC, et al. Antitumor Actions of Intratumoral Delivery of Membrane-Fused Mitochondria in a Mouse Model of Triple-Negative Breast Cancers. Onco Targets Ther. 2020;13:5241–55.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  133. Chan DC. Mitochondria: dynamic organelles in disease, aging, and development. Cell. 2006;125(7):1241–52.

    Article  PubMed  CAS  Google Scholar 

  134. Caicedo A, et al. MitoCeption as a new tool to assess the effects of mesenchymal stem/stromal cell mitochondria on cancer cell metabolism and function. Sci Rep. 2015;5:p9073.

    Article  Google Scholar 

  135. Macheiner T, et al. Magnetomitotransfer: An efficient way for direct mitochondria transfer into cultured human cells. Sci Rep. 2016;6:35571.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  136. Kitani T, et al. Internalization of isolated functional mitochondria: involvement of macropinocytosis. J Cell Mol Med. 2014;18(8):1694–703.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  137. Thomas MA, et al. Human mesenchymal stromal cells release functional mitochondria in extracellular vesicles. Front Bioeng Biotechnol. 2022;10:870193.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Devhare PB, Ray RB. Extracellular vesicles: Novel mediator for cell to cell communications in liver pathogenesis. Mol Aspects Med. 2018;60:115–22.

    Article  PubMed  CAS  Google Scholar 

  139. Nowak M et al. Extracellular Vesicles as Drug Transporters. Int J Mol Sci, 2023. 24(12).

  140. Atukorala I, Hannan N, Hui L. Immersed in a reservoir of potential: amniotic fluid-derived extracellular vesicles. J Transl Med. 2024;22(1):348.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Zhou H, et al. Extracellular vesicles derived from human umbilical cord mesenchymal stem cells alleviate osteoarthritis of the knee in mice model by interacting with METTL3 to reduce m6A of NLRP3 in macrophage. Stem Cell Res Ther. 2022;13(1):322.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  142. Xu H, Xu B. BMSC-Derived Exosomes Ameliorate Osteoarthritis by Inhibiting Pyroptosis of Cartilage via Delivering miR-326 Targeting HDAC3 and STAT1//NF-κB p65 to Chondrocytes. Mediators Inflamm, 2021. 2021: p. 9972805.

  143. He L, et al. Bone marrow mesenchymal stem cell-derived exosomes protect cartilage damage and relieve knee osteoarthritis pain in a rat model of osteoarthritis. Stem Cell Res Ther. 2020;11(1):276.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  144. Amari L, Germain M. Mitochondrial Extracellular Vesicles - Origins and Roles. Front Mol Neurosci. 2021;14:767219.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  145. D’Souza A, et al. Microvesicles transfer mitochondria and increase mitochondrial function in brain endothelial cells. J Control Release. 2021;338:505–26.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Xia L, et al. AdMSC-derived exosomes alleviate acute lung injury via transferring mitochondrial component to improve homeostasis of alveolar macrophages. Theranostics. 2022;12(6):2928–47.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  147. Zhang Z, et al. Mesenchymal Stem Cell-Conditioned Medium Improves Mitochondrial Dysfunction and Suppresses Apoptosis in Okadaic Acid-Treated SH-SY5Y Cells by Extracellular Vesicle Mitochondrial Transfer. J Alzheimers Dis. 2020;78(3):1161–76.

    Article  PubMed  CAS  Google Scholar 

  148. Contentin R et al. Bone Marrow MSC Secretome Increases Equine Articular Chondrocyte Collagen Accumulation and Their Migratory Capacities. Int J Mol Sci, 2022. 23(10).

  149. Peruzzotti-Jametti L, et al. Neural stem cells traffic functional mitochondria via extracellular vesicles. PLoS Biol. 2021;19(4):e3001166.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  150. Baldari S et al. Challenges and Strategies for Improving the Regenerative Effects of Mesenchymal Stromal Cell-Based Therapies. Int J Mol Sci, 2017. 18(10).

  151. Crewe C, et al. Extracellular vesicle-based interorgan transport of mitochondria from energetically stressed adipocytes. Cell Metab. 2021;33(9):1853–e186811.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  152. D’Acunzo P, et al. Isolation of mitochondria-derived mitovesicles and subpopulations of microvesicles and exosomes from brain tissues. Nat Protoc. 2022;17(11):2517–49.

    Article  PubMed  PubMed Central  Google Scholar 

  153. D’Acunzo P et al. Mitovesicles are a novel population of extracellular vesicles of mitochondrial origin altered in Down syndrome. Sci Adv, 2021. 7(7).

  154. Karise I, et al. Metformin enhances mitochondrial biogenesis and thermogenesis in brown adipocytes of mice. Biomed Pharmacother. 2019;111:1156–65.

    Article  PubMed  CAS  Google Scholar 

  155. Huang T, et al. Efficient intervention for pulmonary fibrosis via mitochondrial transfer promoted by mitochondrial biogenesis. Nat Commun. 2023;14(1):5781.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  156. Wang H, et al. Adiponectin partially rescues high glucose/high fat-induced impairment of mitochondrial biogenesis and function in a PGC-1α dependent manner. Eur Rev Med Pharmacol Sci. 2017;21(3):590–9.

    PubMed  CAS  Google Scholar 

  157. Gurunathan S, et al. Platinum Nanoparticles Enhance Exosome Release in Human Lung Epithelial Adenocarcinoma Cancer Cells (A549): Oxidative Stress and the Ceramide Pathway are Key Players. Int J Nanomed. 2021;16:515–38.

    Article  Google Scholar 

  158. Popara J, et al. Silica nanoparticles actively engage with mesenchymal stem cells in improving acute functional cardiac integration. Nanomed (Lond). 2018;13(10):1121–38.

    Article  CAS  Google Scholar 

  159. Han J, et al. Iron oxide nanoparticle-mediated development of cellular gap junction crosstalk to improve mesenchymal stem cells’ therapeutic efficacy for myocardial infarction. ACS Nano. 2015;9(3):2805–19.

    Article  PubMed  CAS  Google Scholar 

  160. Marzano M, et al. Biogenesis of Extracellular Vesicles Produced from Human-Stem-Cell-Derived Cortical Spheroids Exposed to Iron Oxides. ACS Biomater Sci Eng. 2021;7(3):1111–22.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  161. DiDomenico CD, Lintz M, Bonassar LJ. Molecular transport in articular cartilage - what have we learned from the past 50 years? Nat Rev Rheumatol. 2018;14(7):393–403.

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization and Design, M.H.-Z. and T.C.; Literature Search and Selection, M.H.-Z., J.F-W. and K.H.-C.; Manuscript Writing, M.H.-Z.; Language Editing, Y.L.,B.T.-L., and J.J.-Z.; Review and Revision, C.X.-G., T.C., and J.Z.-X. All authors have read and agreed to the published version of the manuscript.

Corresponding authors

Correspondence to Chenxi Gu or Tao Chen.

Ethics declarations

Competing Interest

The authors declare no conflicts of interest.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, M., Wu, J., Cai, K. et al. From dysfunction to healing: advances in mitochondrial therapy for Osteoarthritis. J Transl Med 22, 1013 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-024-05799-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-024-05799-z

Keywords