Next Article in Journal
Glucose-Lowering Drugs and Primary Prevention of Chronic Kidney Disease in Type 2 Diabetes Patients: A Real-World Primary Care Study
Previous Article in Journal
Drugs Targeting Sirtuin 2 Exhibit Broad-Spectrum Anti-Infective Activity
Previous Article in Special Issue
Analysis of miRNAs in Osteogenesis imperfecta Caused by Mutations in COL1A1 and COL1A2: Insights into Molecular Mechanisms and Potential Therapeutic Targets
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Mitochondrial Homeostasis in Mesenchymal Stem Cell Therapy—Potential Implications in the Treatment of Osteogenesis Imperfecta

1
College of Stomatology, Chongqing Medical University, Chongqing 401147, China
2
Chongqing Key Laboratory of Oral Diseases, Chongqing 401147, China
*
Authors to whom correspondence should be addressed.
Pharmaceuticals 2024, 17(10), 1297; https://doi.org/10.3390/ph17101297 (registering DOI)
Submission received: 20 June 2024 / Revised: 11 September 2024 / Accepted: 18 September 2024 / Published: 29 September 2024
(This article belongs to the Special Issue Osteogenesis Imperfecta—Current and Future Therapies)

Abstract

:
Osteogenesis imperfecta (OI) is a hereditary disorder characterized by bones that are fragile and prone to breaking. The efficacy of existing therapies for OI is limited, and they are associated with potentially harmful side effects. OI is primarily due to a mutation of collagen type I and hence impairs bone regeneration. Mesenchymal stem cell (MSC) therapy is an attractive strategy to take advantage of the potential benefits of these multipotent stem cells to address the underlying molecular defects of OI by differentiating osteoblasts, paracrine effects, or immunomodulation. The maintenance of mitochondrial homeostasis is an essential component for improving the curative efficacy of MSCs in OI by affecting the differentiation, signaling, and immunomodulatory functions of MSCs. In this review, we highlight the MSC-based therapy pathway in OI and introduce the MSC regulation mechanism by mitochondrial homeostasis. Strategies aiming to modulate the metabolism and reduce the oxidative stress, as well as innovative strategies based on the use of compounds (resveratrol, NAD+, α-KG), antioxidants, and nanomaterials, are analyzed. These findings may enable the development of new strategies for the treatment of OI, ultimately resulting in improved patient outcomes.

Graphical Abstract

1. Introduction

OI, also referred to as brittle bone disease, is a collection of hereditary conditions that predominantly impact bone integrity, leading to bones that are susceptible to breaking even from small injuries [1]. OI is caused by mutations in genes responsible for the production of type I collagen, a crucial protein that contributes to the strength and structure of bones as well as skin, ligaments, and tendons [2,3]. Most cases of OI are caused by mutations in the COL1A1 and COL1A2 genes, which are responsible for encoding the pro-α1 and pro-α2 chains of type I collagen, respectively [2,3]. These mutations can either decrease collagen production or result in the creation of aberrant collagen, ultimately leading to the weakening and brittleness of bones [4]. OI can be inherited from one or both parents, or it can arise from a spontaneous mutation in the affected individual [5]. Bisphosphonates are commonly used in the treatment of OI due to their ability to inhibit bone resorption by osteoclasts, thereby increasing bone density and reducing the risk of fractures [6]. However, the long-term use of bisphosphonates may result in the accumulation of microdamage, atypical femoral fracture, and mandibular osteonecrosis [7]. In addition to bisphosphonates, denosumab is an anti-resorptive drug that targets and inhibits RANKL, effectively reducing bone resorption and increasing bone density [8]. Teriparatide is an anabolic bone metabolic agent that stimulates bone formation by promoting osteogenesis, which is a different mechanism of action [9]. Another class of anabolic bone metabolic agents for the treatment of OI includes inhibitors of sclerostin or TGF-β. Sclerostin inhibitors, such as romosozumab, function by blocking the activity of sclerostin, leading to increased bone formation and strength [10]. Similarly, targeting TGF-β, a growth factor involved in bone remodeling, has been demonstrated to ameliorate high-turnover bone disease and enhance bone quality in OI models [11]. However, current treatments like bisphosphonates only partially correct the bone phenotype, and they have some disadvantages such as cytotoxic side effects, a lack of effect in certain patients, or relatively poor efficacy [11,12].
Stem cell therapy has the potential to enhance both the quality and quantity of bone in individuals with OI. The treatment of OI with MSCs offers a multifaceted approach through differentiation into bone cells, paracrine effects on bone tissue, and immunomodulatory actions [13,14,15]. Some studies suggest that extracellular vesicles (EVs) secreted by MSCs may be a crucial pathway for stem cell therapy in treating OI [16]. Although it has been confirmed that the transplantation of stem cells can improve the clinical symptoms of OI patients, the osteoblastic engraftment of donor cells was quite low [17,18]. We hope to find new strategies to enhance the function of mesenchymal stem cells, allowing a small number of implanted stem cells to exert a more significant effect, thereby achieving more notable and sustained symptom improvement in OI patients.
Mitochondria play an indispensable role in the mechanisms by which MSCs mediate the treatment of OI. During osteogenic differentiation, MSCs occur in metabolic reprogramming from glycolysis to oxidative phosphorylation (OXPHOS) [19]. This transition is accompanied by an increase in the production of new mitochondria, the replication of mitochondrial DNA (mtDNA), and the activation of respiratory enzymes. The mitochondria meet the increased energy demands in this process [19,20]. In addition, mitochondria control the generation of reactive oxygen species (ROS), which greatly influences the differentiation process of MSCs. Therefore, the maintenance of the function of MSCs is closely related to mitochondrial homeostasis [19,20].
In addition, the process of treating OI with MSCs through paracrine effects is partially mediated by mitochondrial dynamics. Mitochondria are involved in the secretion of various bioactive factors, such as IGF-1, VEGF, and BMP-2, which can stimulate bone growth and remodeling [21]. OI treatment can benefit from MSC-mediated immune responses that facilitate bone regeneration and repair, and mitochondria can play a role in these processes [15,22]. Mitochondrial transfer from MSCs to CD4+ T cells has been shown to inhibit their proliferation and reduce the secretion of pro-inflammatory cytokines, such as IFN-γ, thereby helping to create an immunosuppressive environment [23]. Based on these promising potential pathways, targeting mitochondrial pathways in MSCs represents a promising strategy for enhancing bone repair in OI [24,25].
We review the relevant research into MSC therapy for OI, focusing on the mechanism and application strategies of improving the function of MSCs, promoting bone regeneration and treating OI through the regulation of mitochondrial homeostasis in MSCs.

2. MSC Therapy Mechanisms in OI

2.1. MSC Differentiation into Osteoblasts

MSCs are multipotent stromal cells capable of differentiating into various cell types, including osteoblasts, which are crucial for bone formation and remodeling. Osteoblasts synthesize and secrete the bone matrix and play a vital role in bone mineralization. This differentiation capacity makes MSCs particularly valuable in treating bone-related disorders like OI [26]. Sinder et al. demonstrated that bone marrow stromal cells (BMSCs) from donors can robustly engraft and differentiate into osteoblasts, generating healthy collagen in individuals with OI [27]. After three months of local transplantation, BMSCs showed a 12% increase in cortical thickness, a 14% rise in the polar moment of inertia, a 30% increase in bone strength, and a 30% increase in stiffness [27]. A recent research article demonstrated that the transplantation of hematopoietic stem cells (HSCs) in mice with OI resulted in the replacement of mutant osteoblasts with healthy osteoblasts that originated from HSCs [28]. This study shows significant improvements in bone morphometrics, mechanics, and turnover parameters [28]. Using a mouse model of OI, the transplantation of human fetal blood MSCs (hfMSCs) into the uterus led to an upregulation of osteogenic genes. These genes include osteocalcin, osteoprotegerin (OPG), Osterix (OSX), and bone morphogenetic protein 2 (BMP-2) [29]. These studies all demonstrate the promising prospects of using MSCs to treat OI, highlighting the importance of promoting MSCs’ osteogenic differentiation. In some clinical studies, the osteoblastic engraftment rate of donor cells after stem cell infusion was only 1–2%, yet patients with OI still experienced improvements in clinical symptoms following stem cell transplantation [17,18]. If we cannot increase the implantation rate of stem cells, can we enhance the potency of the stem cells to enable a small number of implanted MSCs to have a more significant effect?
MSCs achieve osteogenic differentiation through many signaling mechanisms, including bone morphogenetic proteins, as well as the Wnt and Notch pathways, which interact with each other and with transcription factors to regulate MSCs’ differentiation [30]. Transcription factors like Runx2 and OSX occupy a very important position in starting the process of osteogenic differentiation. BMP-2 stimulates the expression of Runx2 and OSX, which are responsible for guiding MSCs to become osteogenic cells and develop into mature osteoblasts [31]. Besides their role in MSCs’ osteogenic differentiation, these transcription factors also have intimate connections with mitochondrial function. Runx2 transcription factor has been shown to negatively regulate SIRT6, a member of the sirtuin family of proteins that regulate gene expression and are known to enhance mitochondrial activity [32]. SIRT6 enhances mitochondrial oxygen consumption, while RUNX2 inhibits SIRT6 expression and reduces oxygen consumption [32]. All of those show a strong correlation in the osteogenic differentiation of MSCs and mitochondrial function, and maintaining mitochondrial homeostasis is a feasible strategy to guide mesenchymal stem cells to achieve our purpose.

2.2. Paracrine Effects in Bone Healing

MSC therapy can improve OI through osteogenic differentiation. However, the efficacy of MSC therapy, regardless of the mechanism and delivery approaches, must consider the homing, adhesion, survival, retention, immunomodulation, angiogenesis, engraftment, and integration of transplanted MSCs at the tissue repair site [15,33]. Otsuru et al. conducted an early study in which they discovered that MSCs release a soluble mediator that indirectly promotes bone formation [34]. In the TERCELOI clinical trial, proteomics and transcriptomics analyses were employed to study two children with severe osteogenesis imperfecta who received multiple injections of MSCs isolated from HLA-matched siblings. They found that the expression of proteins controlled by the TGF-β and BMP signaling pathways increased to varying degrees, and there was also the upregulation of signaling molecules related to osteoprogenitor cells, collagen binding, and the extracellular matrix [26,35]. When MSCs are cultured in an osteogenic medium, they secrete distinct substances at certain times that enhance alkaline phosphatase activity (ALP) in external MSCs and facilitate their movement [36]. Additionally, factors released by bone marrow-derived mesenchymal stem cells (BM-MSCs) through paracrine secretion recruit macrophages and endothelial cells to the injured location [37]. During this process, wounds heal faster, as the cells migrate and multiply, and the presence of progenitor cells and immune cells increases [37]. Several studies have shown that MSCs release various types of EVs, including microvesicles (MVs) and exosomes that can serve as paracrine mediators between MSCs and target cells [38,39,40].
The release of EVs, MVs, and exosomes in MSCs is a complex process that mitochondria play an important role in this process. The release of EVs from MSCs is regulated by mitochondrial stress and dysfunction. Treating MSCs with mitochondrial inhibitors, such as rotenone and antimycin A, which impair mitochondrial function, results in an obvious change in the size distribution of the released EVs [41,42]. This suggests that mitochondrial function influences EV biogenesis and release. Similarly, MSCs cultured under physiological oxygen conditions release fewer EVs compared to cells with mitochondrial dysfunction induced by high oxygen or other stressors, which implies mitochondrial homeostasis regulates EV secretion [43,44]. Therefore, mitochondrial homeostasis is the foundation for the normal function of paracrine secretion in MSCs [45].

2.3. Immunomodulation by MSCs

The occurrence of OI may be related to inflammation. MSCs have immunomodulatory properties, which can regulate immune response, reduce the level of inflammation, and establish a conducive setting for bone healing, thus avoiding the exacerbation of bone fragility due to inflammation and damage to delay the healing process [13,46]. Zhang et al. found that MSCs can alter the cytokine profile in the body, elevating the levels of anti-inflammatory cytokines such as IL-10 and decreasing pro-inflammatory cytokines like IL-6 and TNF-α. Creating a favorable environment for bone regeneration can result from this shift [47]. MSCs inhibit T cell proliferation through direct contact and secrete specific cytokines like TGF-β, which promote the formation of regulatory T cells, aiding in a regulated inflammatory response crucial for bone repair [13]. Moreover, MSCs can influence the immune system by transferring their mitochondria to different immune cells, such as T cells. The transfer of MSC mitochondria to Th1 cells has been shown to reduce the expression of T-bet, which is the key transcription factor for Th1 cell development, leading to the suppression of IFN-γ production [23,48]. The transfer of MSC-derived mitochondria to Th17 cells can impair their IL-17 production and promote the generation of a significant suppressed regulatory T cell (Treg) population [21]. MSCs regulate immune cell function through metabolic reprogramming via mitochondrial transfer. This process enhances mitochondrial respiration in immune cells, leading to immunosuppressive effects, highlighting the importance of mitochondria in MSC-mediated immune regulation [49].
The three mechanisms of differentiation into osteoblasts, paracrine signaling, and immunomodulatory functions by which mesenchymal stem cells treat OI are briefly outlined in Figure 1.

3. Mechanisms of Mitochondrial Homeostasis Regulate the Function of MSCs

Research indicates that introducing human fetal blood MSCs (hfMSCs) into animal models of OI resulted in enhanced bone characteristics and reduced fracture occurrence [29,51]. In previous research, Horwitz et al. documented the clinical outcomes of three children with severe OI after receiving allogeneic mesenchymal bone marrow transplantation. A histological assessment conducted 216 days post-surgery, in addition to a bone densitometry analysis, showed significant improvement in bone structure. All patients had increases in total body bone mineral and growth velocity and reduced frequencies of bone fracture [52]. In the six months after transplantation, three children showed increased growth acceleration and total body bone mineral content in comparison to two non-transplanted OI controls. With extended follow-up periods up to six months after transplantation, growth rates slowed or plateaued, but the amount of bone mineral continued to rise [17]. Horwitz et al. treated six children with severe osteogenesis imperfecta using gene-marked, donor marrow-derived mesenchymal cells to show that mesenchymal cell treatment is feasible. In the first six months following infusion, engraftment was observed in five out of six patients, including the marrow stroma, skin, and bone. Additionally, there was an acceleration of growth velocity [18]. MSC therapy provides a feasible method to reduce the fracture rate in individuals with OI, which can be attributed to the donor cells differentiating into osteoblasts and generating normal bone matrix proteins, such as collagen type I [29,52]. However, Horwitz et al. also pointed out that although the symptoms of OI patients were improved after stem cell transplantation, the implantation rate of stem cells was low [17,18]. The question of how to increase the osteogenic differentiation of successfully implanted stem cells is particularly important, improving the function of MSCs by maintaining mitochondrial homeostasis may achieve this goal.
Mitochondrial homeostasis is implicated in numerous intricate cellular processes, including autophagy, the regulation of MSCs’ differentiation, and immunological responses [53,54]. These processes highlight the pivotal role of mitochondria in physiological regulation, cellular health, and disease. Mitochondrial homeostasis constitutes a prerequisite for the normal functioning of mitochondrial activities [55]. Mitochondrial homeostasis refers to the balance and stability of mitochondrial function, structure, and quantity within cells, ensuring the optimal performance of mitochondria and adaptation to constantly changing cellular demands and environmental conditions [56,57]. Mitochondrial homeostasis includes the complex processes of mitochondrial biogenesis, mitophagy, dynamics, proteostasis, signaling, and metabolic pathways that preserve the integrity and functionality of mitochondria [58]. Mitochondrial dysfunction can lead to MSC function decreases, especially osteogenesis differentiation [19,59]. During the osteogenic differentiation process, mitochondrial biosynthesis, oxygen consumption, and ATP production all increase, indicating the significant role of mitochondria in this process [60]. Mitochondrial dysfunction can result in the overproduction of ROS, leading to oxidative stress and negative impacts on the cellular environment, further affecting the osteogenic differentiation process of MSCs [61]. Maintaining mitochondrial redox balance is crucial for sustaining MSCs’ osteogenic differentiation, which is the basis of MSC therapy of OI [62] (Figure 2).

3.1. Mitochondrial Metabolism in MSCs

Mitochondrial metabolism is indispensable in meeting the diverse metabolic demands of MSCs. During osteoblast formation, there is a great rise in the rate of oxygen consumption and the levels of ATP within the cell, indicating a close association between mitochondrial energy metabolism and MSCs’ differentiation [21]. When stem cells age, damaged mitochondria exhibit abnormal OXPHOS, leading to excessive ROS production. The shift from OXPHOS to glycolysis decreases ATP generation, triggering AMPK activation, which leads to cell cycle arrest and promotes aging [64]. Regulating ROS levels can effectively maintain the functionality of MSCs and decline functional attenuation [65]. Compounds such as 17β-Estradiol have been found to decrease oxidative stress damage, enhance antioxidant enzyme activity, and protect MSCs from the negative effects of ROS induced by mitochondrial production under high-glucose conditions [66]. Furthermore, the PPAR-γ agonist pioglitazone has demonstrated the ability to shield MSCs from damage caused by oxidative stress through regulating ROS levels, indicating its potential in promoting bone regeneration and reducing the risk of osteoporosis [67]. The metabolites produced by the mitochondrial OXPHOS pathway are pivotal in regulating the activity, quiescence, self-renewal, and differentiation of MSCs by influencing ROS levels [21].
MSCs undergo osteogenesis differentiation through a complex interplay between metabolic byproducts and epigenetic regulation, which is driven by mitochondrial metabolism. Metabolites produced by mitochondria serve as essential substrates for various chromatin-modifying enzymes, therefore controlling gene expression in MSCs through enabling specific chromatin alterations [68]. These mitochondrial metabolites are instrumental in regulating gene expression by promoting the acetylation of β-catenin, a process that not only induces its nuclear translocation but also enhances downstream transcriptional activity master regulators such as Runx2 and OSX [69]. This regulatory mechanism plays an important role in directing the differentiation of MSCs towards osteoblasts rather than other cell lineages.
Moreover, mitochondrial metabolites like S-adenosylmethionine (SAM) and α-ketoglutarate (α-KG), which are essential for the OXPHOS process, influence the epigenetic landscape through N6-methyladenine (N6-mA) DNA modification [70]. This modulation of the epigenetic landscape is key to the fate determination of MSCs. The demethylase Alkbh1, regulated by N6-mA modification, has been identified as a significant participant in this regulatory mechanism [71]. The knockout of Alkbh1 has been shown to skew the differentiation potential of BMSCs toward an adipogenic lineage while concurrently inhibiting osteogenic differentiation [72]. This imbalance in differentiation potential underscores the importance of mitochondrial metabolism and epigenetic modifications to the compromised bone quality and increased fracture risk characteristic of OI.
This intricate network of mitochondrial metabolism, epigenetic modifications, and gene expression regulation underscores the significance of mitochondrial pathways in the osteogenic differentiation of MSCs and their potential as therapeutic targets. By influencing key transcriptional regulators and modifying the epigenetic landscape, mitochondrial metabolites directly or indirectly impact the cellular fate decisions of MSCs. Targeting these mitochondrial pathways could offer new strategies for improving bone quality and reducing the fracture risk in OI, emphasizing the necessity for additional investigation into mitochondrial function and its therapeutic potential in OI [68,73,74].

3.2. Mechanism of Mitochondrial Anti-Oxidative Stress

The presence of ROS is pivotal in bone regeneration and reconstruction, but their levels must be finely regulated to ensure optimal therapeutic outcomes. Appropriate levels of ROS are necessary for various signaling pathways involved in bone formation; excessive ROS can impair stem cell function and hinder bone healing processes [75,76]. As MSCs differentiate into osteoblasts, there is a decrease in intracellular ROS levels, accompanied by an upregulation of antioxidant enzymes including superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) [77]. This upregulation of antioxidant enzymes helps counteract the adverse impacts of excessive ROS and promotes osteoblast differentiation and bone reconstruction [78] (Figure 3).
Antioxidants and natural enzymes contribute to the control of oxidative signals and scavenging of excessive ROS in MSCs, thus modulating their behaviors and fate decisions [80,81]. A myriad of papers have indicated that the osteogenic capacity of MSCs could be improved by either providing antioxidant supplementation or overexpressing the antioxidant enzymes [82,83,84]. Nevertheless, it should be pointed out that total ROS depletion is not desirable as regulated ROS levels are needed for cellular signaling and the activation of pathways associated with osteoblast differentiation and bone matrix production [85]. Thus, delicate control over ROS levels would be essential to promote bone regeneration and reconstruction and, on the other hand, to avoid increasing oxidative stress to harmful levels.
These results show that targeting ROS modulation and stimulating antioxidant defense mechanisms in MSCs could be an effective strategy in the MSC treatment of OI [76]. Targeting oxidative stress and antioxidants, therefore, has the potential to fine-tune the redox balance and facilitate osteoblast differentiation to improve bone quality and healing in OI [76].

3.3. Mitochondrial Quality Control in MSCs

Mitochondrial quality control (MQC) systems are essential for maintaining cellular homeostasis and ensuring the normal function of MSCs during bone-forming differentiation. These systems encompass various mechanisms, including protein balance, generation, movement, and mitophagy [86]. The accumulation of mitochondrial defects may impair cellular function and differentiation, and MQC is indispensable for preventing the elimination of damaged or dysfunctional mitochondria [87]. For the MSC treatment of OI, MQC mechanisms are expected to be potential therapeutic targets for enhancing the osteogenic potential of MSCs [45,88].
The NAD+-dependent deacetylase family, sirtuin, which is involved in mitochondrial quality control, has been regarded as a novel therapeutic target for the enhancement of bone quality, highlighting its role in the osteogenesis of MSCs [89]. SIRT1 has been proven to enhance the process of MSCs transforming into bone cells by controlling the creation and operation of mitochondria [88,90]. Furthermore, resveratrol has been found to improve the osteogenic differentiation of aging-affected PO-MSCs (periosteum-derived MSCs) by upregulating the expression of mitochondrial inner membrane proteins (such as Mitofilin) [91]. Research has shown that resveratrol greatly improves extracellular matrix calcification and the expression of osteogenic-related genes, inhibits ROS production in BMSCs, and upregulates AMPK expression, suggesting its potential mechanism in promoting BMSC osteogenic differentiation [92,93].
The mTOR signaling pathway also regulates mitochondrial quality control by affecting mitochondrial biogenesis, dynamics, and autophagy [94]. Rapamycin, an inhibitor of mTOR signaling, upregulates the expression of genes involved in mitochondrial autophagy and mitochondrial fission, while downregulating genes associated with mitochondrial fusion (such as OPA1), thereby maintaining mitochondrial homeostasis [95]. During osteogenic differentiation, an appropriate concentration of rapamycin promotes BMSC osteogenic differentiation through autophagy activation [96].
Collagen membranes crosslinked with 3,4-dihydroxyphenylacetic acid (HPAA-HCM) have been demonstrated to enhance the osteogenic development of MSCs [97]. The self-mineralization ability of these membranes provides a bone-conductive scaffold, supporting the growth and differentiation of MSCs into osteoblasts [98]. HCM accelerates osteogenic differentiation by activating mitochondrial dynamics, which is associated with the expression of key proteins regulating mitochondrial morphology and function, highlighting its role as a core mechanism in osteogenic differentiation [94,97].
In conclusion, MQC mechanisms, such as the regulation of mitochondrial biogenesis, dynamics, and autophagy, are pivotal in the osteogenic differentiation of MSCs and offer potential therapeutic targets for treating OI [86,94]. Sirtuins, resveratrol, mTOR signaling, and HPAA-HCM could become promising approaches to modulate MQC and enhance the osteogenic potential of MSCs.

4. Strategies for Regulating the Function of MSCs through Mitochondrial Homeostasis

4.1. Regulating Mitochondrial Metabolism in MSCs

4.1.1. Resveratrol

Resveratrol, a naturally occurring polyphenolic compound, has been identified as a potent modulator of mitochondrial function and metabolic homeostasis [89,91,94]. It exerts its effects by activating SIRT1 (NAD+-dependent deacetylase), which in turn promotes the deacetylation and activation of PGC-1α. PGC-1α is a key controller of mitochondrial biogenesis [89,92]. This activation cascade enhances mitochondrial function and may counteract the mitochondrial dysfunction associated with OI.
Resveratrol’s therapeutic potential not only promotes mitochondrial biogenesis but also has anti-inflammatory properties that are beneficial in treating inflammatory diseases [93,95]. Studies have demonstrated that resveratrol can promote osteogenic differentiation in various MSC populations, such as gingival MSCs and BMSCs, and can ameliorate age-related osteoporosis by restoring mitochondrial OXPHOS capacity through pathways involving Mitofilin and PGC-1α [96,97]. In osteogenic cultures, resveratrol treatment has been shown to enhance ALP, an early indicator of osteogenic differentiation, and to promote mineralization, a late-stage indicator of bone formation [75,76]. These findings indicate that resveratrol positively influences bone formation and may have therapeutic benefits which enhance bone quality and decrease fracture risk in individuals with OI. The direct supplementation of mitochondrial metabolites, such as resveratrol, offers a novel approach for therapeutic research, potentially leading to innovative treatments for metabolic and degenerative diseases such as OI [95]. By targeting mitochondrial pathways, resveratrol and similar compounds could provide a means to enhance the regenerative capacity of MSCs.

4.1.2. NAD+

Nicotinamide adenine dinucleotide (NAD+), an essential coenzyme in OXPHOS, has been shown to significantly influence MSC differentiation [98]. During the osteogenic differentiation of BMSCs, a marked metabolic shift occurs, characterized by an upregulation of OXPHOS activity, a reduction in glycolysis, and an increase in intracellular NAD+ levels [99,100]. This shift from glycolysis to OXPHOS is necessary for meeting the increased energy demands of differentiation and bone formation. The inhibition of NAD+ synthesis disrupts this metabolic reprogramming, leading to mitochondrial dysfunction and impaired osteogenic differentiation [80]. Conversely, replenishing NAD+ levels can partially restore this impairment, underscoring the crucial importance of NAD+ in supporting mitochondrial function and osteogenesis [101]. Elevated NAD+ levels are crucial for maintaining important metabolic pathways, including glucose glycolysis, the tricarboxylic acid (TCA) cycle, and fatty acid β-oxidation, which are necessary for cellular metabolism and energy generation [102,103]. This highlights the indispensable role of NAD+-mediated mitochondrial OXPHOS in BMSC osteogenic differentiation and provides insights into the mechanisms of bone formation, suggesting potential therapeutic targets for enhancing bone repair and regeneration.
Furthermore, maintaining optimal NAD+ levels has emerged as a novel therapeutic target in regenerative medicine. Studies have demonstrated that the exogenous supplementation of NAD+ can effectively delay senescence in BMSCs induced by external factors, such as D-galactose, and increase intracellular NAD+ levels [81]. The silencing of SIRT1, an NAD+-dependent deacetylase, exacerbates D-galactose-induced senescence, highlighting the defensive impact of exogenous NAD+ on aging BMSCs [84,104]. This suggests that interventions aimed at enhancing NAD+ levels and SIRT1 activity could offer promising strategies for combating an age-related decline in BMSC function and promoting bone regeneration in conditions like OI [105,106].

4.1.3. Alpha-Ketoglutarate (α-KG)

α-KG is a key intermediate in the TCA cycle that plays a crucial role in mitochondrial metabolism. Emerging evidence suggests that α-KG has therapeutic potential for bone disorders [107,108]. α-KG exerts its beneficial effects on MSCs by regulating the epigenetic landscape, specifically through modulating histone methylation patterns. It reduces the enrichment of repressive histone marks, such as H3K9me3 and H3K27me3, on the promoters of key osteogenic genes like BMP2, BMP4, and Nanog [87]. By alleviating this epigenetic repression, α-KG promotes the expression of these genes, thereby stimulating the proliferation, migration, and osteogenic differentiation of MSCs [87].
In vivo research has shown the therapeutic potential of α-KG supplementation in combating osteoporosis and age-related bone loss. The administration of α-KG significantly attenuates bone deterioration in aged mice, preserving bone mass and strength [87,109]. Moreover, α-KG accelerates bone regeneration in aged rodents, highlighting its capacity to improve the regenerative capacity of MSCs and promote bone repair [110]. Beyond its epigenetic effects, α-KG is essential in supporting bone matrix formation and collagen synthesis [111]. As a key intermediate in the TCA cycle, it serves as a significant source of amino acids required for collagen production in cells and organisms [112]. This metabolic function of α-KG helps maintain the integrity of bone tissue and the promote osteogenesis.
Furthermore, α-KG was demonstrated to exert pro-osteogenic effects in osteoblast cell lines through the activation of signaling pathways such as JNK and mTOR/S6K1/S6 [113]. These pathways are involved in regulating cell proliferation, differentiation, and protein synthesis, further supporting the significance of α-KG in promoting bone formation [114]. In conclusion, mitochondrial metabolism, particularly the TCA cycle intermediate α-KG, is crucial in the osteogenic differentiation of MSCs and has substantial therapeutic potential in the treatment of OI and other bone disorders. These relevant studies are summarized in Table 1.

4.2. Mitochondrial Anti-Oxidative Stress Strategy through Antioxidants

4.2.1. N-Acetylcysteine (NAC)

NAC and other antioxidants are essential in preserving cellular homeostasis and supporting the function of MSCs by scavenging ROS. Yamada et al. found that NAC can increase the expression of bone-related genes such as collagen I, osteopontin, osteocalcin, BMP-2, and Runx2/cbfa1, which implies NAC accelerated bone regeneration by activating the differentiation of osteogenic lineages [111]. In a study involving rat femoral defects, the pre-treatment of autologous BMSCs with NAC, followed by implantation using a collagen sponge, yielded markedly higher new bone growth in comparison to the control group [112]. Various studies have shown that NAC has indeed been shown to significantly reduce ROS levels and promote fracture healing [76,113,114]. NAC has been shown to promote bone regeneration through various mechanisms, including its antioxidant activity, its ability to enhance osteogenic differentiation, the activation of pro-osteogenic signaling pathways, improvements in scaffold properties, and the promotion of bone healing in vivo. These properties of NAC suggest its potential therapeutic application in the context of OI.

4.2.2. Vitamin C

Vitamin C, another potent antioxidant, is essential for osteoblast growth and differentiation, making it a key player in musculoskeletal healing. Clinical studies have demonstrated that patients undergoing open reduction and internal fixation surgery who receive vitamin C treatment show increased plasma levels of ALP and osteocalcin, which are markers of increased bone density and faster healing [115]. Roman et al. demonstrated that vitamin C plays a role in orchestrating osteogenic differentiation and function by modulating chromatin accessibility and priming transcriptional activity through epigenetic mechanisms [116]. These findings highlight the advantageous impacts of vitamin C supplementation on bone regeneration and repair.

4.2.3. Alpha-Lipoic Acid (α-LA)

α-LA, a powerful mitochondrial antioxidant, is gaining recognition for its role in maintaining cellular redox balance and promoting bone tissue regeneration. α-LA can protect against bone loss in the rat mandible and promote bone formation by inhibiting oxidative stress [117]. Similarly, another study found that the supplementation of α-LA promotes the healing of femoral fractures in rats [118]. α-LA therapy enhances bone regeneration in bone loss patients by increasing new bone formation, bone volume, and bone mineral density in vivo. α-LA triggers signaling pathways that promote osteogenic differentiation and bone production, such as the PI3K/AKT pathway [119,120]. OI and osteoporosis are both conditions characterized by bone fragility; they have different underlying causes and clinical features, but they have some similarities in terms of treatment, which shows the potential promise of using α-LA to treat OI. The modulation of these signaling pathways could help counteract the impaired osteogenesis observed in OI. These relevant studies have been summarized in Table 2.

4.3. Mitochondrial Anti-Oxidative Stress Strategy through Biomaterials

4.3.1. Graphene Oxide (GO)

GO is a modified form of graphene that has been functionalized with several oxygen-containing groups, showing properties of being hydrophilic and biocompatible. Graphene oxide, when modified with metal nanoparticles, has been demonstrated to improve the anti-inflammatory properties in MSCs for tissue engineering purposes [129]. This characteristic is especially significant in the setting of MSC therapy, where increased oxidative stress and inflammation contribute to the impaired osteogenic differentiation of MSCs and defective bone formation [80].

4.3.2. Fullerene Alcohol/Alginate Hydrogels

Fullerene alcohol/alginate hydrogels are injectable cell delivery carriers with antioxidant properties. These hydrogels can effectively scavenge superoxide anions and hydroxyl radicals, protecting brown adipose-derived stem cells from oxidative stress and enhancing their survival in ROS-rich environments [129]. Similarly, polydopamine (PDA), a material with catechol functional groups, possesses antioxidant properties and has been used to fabricate nanostructures that protect MSCs from oxidative stress [130]. PDA and its derivatives have been utilized to enhance stem cell functionality, remove ROS, and alleviate oxidative stress [131]. By scavenging excessive ROS and mitigating oxidative stress, these nanostructures create a favorable microenvironment for MSC survival and differentiation.

4.3.3. Polydopamine (PDA)

PDA-coated substrates have been shown to significantly reduce oxidative stress and mitochondrial damage in replicative senescent MSCs. This is in part due to the scavenging of extracellular ROS, an aggravating factor of cellular senescence [132]. In addition, the PDA coating may also reduce the expression of senescence-associated genes, including p53 and p21, while enhancing the expression of stemness-associated genes such as OCT4 [133]. PDA nanoparticles, due to their distinct hydroquinone moiety, possess enhanced antioxidant properties, which allows them to scavenge free radicals effectively. This property is crucial for protecting MSCs from oxidative stress-induced damage [134,135].

4.3.4. Cerium Oxide Nanoparticles (CeNPs)

CeNPs demonstrate catalytic behavior by scavenging ROS through redox cycles between Ce3+ and Ce4+ states via electron charge transfer, collecting, storing, or releasing oxygen on their surface [136,137]. This unique feature allows CeNPs to effectively regulate the concentration of ROS in cellular environments, providing a protective effect [138,139]. By utilizing a hydrothermal method to construct a microenvironment-responsive bio-functional metal–organic framework (bio-MOF) coating on titanium surfaces through coordination between pxylylenebisphosphonate (PXBP) and Ce/Sr ions, catalytic properties similar to CAT and SOD are achieved, decomposing ROS in MSCs and restoring their mitochondrial function [136]. This method has demonstrated encouraging outcomes in stimulating the osteogenic differentiation of MSCs and improving bone repair [140].

4.3.5. Manganese Dioxide

The antioxidant mechanism of manganese dioxide (MnO2-x) nanoparticles is attributed to their enzyme-like activity, acting as catalysts to remove superoxide and peroxynitrite, protecting cells from oxidative stress [141]. Manganese-containing bioceramics, such as manganese-doped β-tricalcium phosphate (Mn-TCP), can inhibit osteoclast formation by clearing ROS, promoting bone regeneration in osteoporotic bone defects [142]. These findings suggest that MnO2-x nanoparticles could potentially be employed to modulate the osteogenic differentiation of MSCs and improve bone quality in OI by regulating ROS levels and mitochondrial function [132].

4.3.6. Iron Oxide

Iron oxide (Fe3O4) nanoparticles can be prepared with natural antioxidant activity by incorporating plant or fruit extracts, enhancing their bioactive functions and providing a sustainable method for the environmentally friendly preparation of metal nanoparticles [143,144]. These green-synthesized Fe3O4 nanoparticles have shown promising results in scavenging ROS and promoting the osteogenic differentiation of MSCs [145,146]. By modulating mitochondrial metabolism and reducing oxidative stress, Fe3O4 nanoparticles could potentially be used to treat OI by enhancing the osteogenic potential of MSCs and improving bone formation [84].
Nanomaterials with enzyme-like capabilities offer distinct advantages over natural enzymes, such as enhanced stability, cost-effectiveness, and easy storage, making them well suited for biomedical uses [147,148,149]. These nanomaterials, also known as nanozymes, show promising prospects in the treatment of OI due to their potential in scavenging ROS and regulating the cellular microenvironment [132,150]. These relevant studies have been summarized in Table 3.

5. Conclusions

Exploring MSC therapy for OI by controlling mitochondrial homeostasis is a prospective way of curing this genetic disorder. Current research has demonstrated that MSC therapy can improve clinical symptoms in patients with OI. However, the implantation rate of stem cells remains relatively low. By summarizing previous studies, it has been suggested that maintaining mitochondrial homeostasis to enhance the function of MSCs may allow even a small number of implanted stem cells to have a more significant impact, potentially enhancing the efficacy of MSC therapy for OI patients. Mitochondrial metabolic modulation, anti-oxidative stress approaches, and the application of nanomaterials offer innovative ways of maintaining mitochondrial homeostasis, enhancing bone regeneration, and treating OI. The potential of compounds like resveratrol, nicotinamide adenine dinucleotide (NAD+), and α-KG in regulating mitochondrial function and osteogenic differentiation of MSCs is particularly noteworthy. Furthermore, the role of antioxidants and nanomaterials in protecting MSCs from oxidative stress and improving bone healing outcomes creates new possibilities for OI therapy. Mitochondrial quality control mechanisms, such as protein homeostasis, biogenesis, dynamics, and mitophagy, are indispensable in maintaining cellular homeostasis and ensuring the proper function of MSCs during osteogenic differentiation. Targeting mitochondrial pathways could offer new strategies for improving bone quality and reducing fracture risk in OI patients, emphasizing the need for further research into mitochondrial function and its therapeutic potential in OI. By focusing on the regulation of mitochondrial function, researchers and clinicians can potentially unlock new therapeutic strategies that not only address the symptoms of OI but also target its underlying molecular defects. As we advance our understanding of mitochondrial biology in the context of stem cell therapy, the prospects for developing effective treatments for OI and other bone-related disorders become increasingly promising.

Author Contributions

Q.G. wrote the manuscript and participated in the literature search and related data sorting. Q.Z. and P.J. conceived and revised the manuscript. All authors contributed to the article and approved the submitted version. All authors have read and agreed to the published version of the manuscript.

Funding

This study was sponsored by the National Natural Science Foundation of China (82201059, 82071115, 82220108019); the Chongqing Postdoctoral Science Special Foundation (2021XM1031); the Natural Science Foundation of Chongqing, China (CSTB2022NSCQ-BHX0003); and the China Postdoctoral Science Foundation (2022M720599).

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as potential conflicts of interest.

References

  1. Etich, J.; Lessmeier, L.; Rehberg, M.; Sill, H.; Zaucke, F.; Netzer, C.; Semler, O. Osteogenesis imperfecta-pathophysiology and therapeutic options. Mol. Cell. Pediatr. 2020, 7, 9. [Google Scholar] [CrossRef] [PubMed]
  2. Zhytnik, L.; Maasalu, K.; Reimann, E.; Prans, E.; Koks, S.; Martson, A. Mutational analysis of COL1A1 and COL1A2 genes among Estonian osteogenesis imperfecta patients. Hum. Genom. 2017, 11, 19. [Google Scholar] [CrossRef] [PubMed]
  3. Ho Duy, B.; Zhytnik, L.; Maasalu, K.; Kandla, I.; Prans, E.; Reimann, E.; Martson, A.; Koks, S. Mutation analysis of the COL1A1 and COL1A2 genes in Vietnamese patients with osteogenesis imperfecta. Hum. Genom. 2016, 10, 27. [Google Scholar] [CrossRef]
  4. Lu, Y.; Zhang, S.; Wang, Y.; Ren, X.; Han, J. Molecular mechanisms and clinical manifestations of rare genetic disorders associated with type I collagen. Intractable Rare Dis. Res. 2019, 8, 98–107. [Google Scholar] [CrossRef] [PubMed]
  5. Basel, D.; Steiner, R.D. Osteogenesis imperfecta: Recent findings shed new light on this once well-understood condition. Genet. Med. 2009, 11, 375–385. [Google Scholar] [CrossRef] [PubMed]
  6. Ott, S.M. Long-term safety of bisphosphonates. J. Clin. Endocrinol. Metab. 2005, 90, 1897–1899. [Google Scholar] [CrossRef]
  7. Drake, M.T.; Clarke, B.L.; Khosla, S. Bisphosphonates: Mechanism of action and role in clinical practice. Mayo Clin. Proc. 2008, 83, 1032–1045. [Google Scholar] [CrossRef]
  8. Majdoub, F.; Ferjani, H.L.; Nessib, D.B.; Kaffel, D.; Maatallah, K.; Hamdi, W. Denosumab use in osteogenesis imperfecta: An update on therapeutic approaches. Ann. Pediatr. Endocrinol. Metab. 2023, 28, 98–106. [Google Scholar] [CrossRef]
  9. Orwoll, E.S.; Shapiro, J.; Veith, S.; Wang, Y.; Lapidus, J.; Vanek, C.; Reeder, J.L.; Keaveny, T.M.; Lee, D.C.; Mullins, M.A.; et al. Evaluation of teriparatide treatment in adults with osteogenesis imperfecta. J. Clin. Investig. 2014, 124, 491–498. [Google Scholar] [CrossRef]
  10. Rauner, M.; Taipaleenmaki, H.; Tsourdi, E.; Winter, E.M. Osteoporosis Treatment with Anti-Sclerostin Antibodies-Mechanisms of Action and Clinical Application. J. Clin. Med. 2021, 10, 787. [Google Scholar] [CrossRef]
  11. Botor, M.; Fus-Kujawa, A.; Uroczynska, M.; Stepien, K.L.; Galicka, A.; Gawron, K.; Sieron, A.L. Osteogenesis Imperfecta: Current and Prospective Therapies. Biomolecules 2021, 11, 1493. [Google Scholar] [CrossRef] [PubMed]
  12. Battle, L.; Yakar, S.; Carriero, A. A systematic review and meta-analysis on the efficacy of stem cell therapy on bone brittleness in mouse models of osteogenesis imperfecta. Bone Rep. 2021, 15, 101108. [Google Scholar] [CrossRef] [PubMed]
  13. Medhat, D.; Rodriguez, C.I.; Infante, A. Immunomodulatory Effects of MSCs in Bone Healing. Int. J. Mol. Sci. 2019, 20, 5467. [Google Scholar] [CrossRef] [PubMed]
  14. Linero, I.; Chaparro, O. Paracrine effect of mesenchymal stem cells derived from human adipose tissue in bone regeneration. PLoS ONE 2014, 9, e107001. [Google Scholar] [CrossRef] [PubMed]
  15. Gotherstrom, C.; Walther-Jallow, L. Stem Cell Therapy as a Treatment for Osteogenesis Imperfecta. Curr. Osteoporos. Rep. 2020, 18, 337–343. [Google Scholar] [CrossRef]
  16. Otsuru, S.; Desbourdes, L.; Guess, A.J.; Hofmann, T.J.; Relation, T.; Kaito, T.; Dominici, M.; Iwamoto, M.; Horwitz, E.M. Extracellular vesicles released from mesenchymal stromal cells stimulate bone growth in osteogenesis imperfecta. Cytotherapy 2018, 20, 62–73. [Google Scholar] [CrossRef]
  17. Horwitz, E.M.; Prockop, D.J.; Gordon, P.L.; Koo, W.W.; Fitzpatrick, L.A.; Neel, M.D.; McCarville, M.E.; Orchard, P.J.; Pyeritz, R.E.; Brenner, M.K. Clinical responses to bone marrow transplantation in children with severe osteogenesis imperfecta. Blood 2001, 97, 1227–1231. [Google Scholar] [CrossRef]
  18. Horwitz, E.M.; Gordon, P.L.; Koo, W.K.; Marx, J.C.; Neel, M.D.; McNall, R.Y.; Muul, L.; Hofmann, T. Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: Implications for cell therapy of bone. Proc. Natl. Acad. Sci. USA 2002, 99, 8932–8937. [Google Scholar] [CrossRef]
  19. Li, Q.; Gao, Z.; Chen, Y.; Guan, M.X. The role of mitochondria in osteogenic, adipogenic and chondrogenic differentiation of mesenchymal stem cells. Protein Cell 2017, 8, 439–445. [Google Scholar] [CrossRef]
  20. Jorgensen, C.; Khoury, M. Musculoskeletal Progenitor/Stromal Cell-Derived Mitochondria Modulate Cell Differentiation and Therapeutical Function. Front. Immunol. 2021, 12, 606781. [Google Scholar] [CrossRef]
  21. Yan, W.; Diao, S.; Fan, Z. The role and mechanism of mitochondrial functions and energy metabolism in the function regulation of the mesenchymal stem cells. Stem Cell Res. Ther. 2021, 12, 140. [Google Scholar] [CrossRef] [PubMed]
  22. Fan, X.L.; Zhang, Y.; Li, X.; Fu, Q.L. Mechanisms underlying the protective effects of mesenchymal stem cell-based therapy. Cell. Mol. Life Sci. 2020, 77, 2771–2794. [Google Scholar] [CrossRef] [PubMed]
  23. Akhter, W.; Nakhle, J.; Vaillant, L.; Garcin, G.; Le Saout, C.; Simon, M.; Crozet, C.; Djouad, F.; Jorgensen, C.; Vignais, M.L.; et al. Transfer of mesenchymal stem cell mitochondria to CD4+ T cells contributes to repress Th1 differentiation by downregulating T-bet expression. Stem Cell Res. Ther. 2023, 14, 12. [Google Scholar] [CrossRef] [PubMed]
  24. Zhang, L.; Liu, Q.; Hu, H.; Zhao, L.; Zhu, K. Progress in mesenchymal stem cell mitochondria transfer for the repair of tissue injury and treatment of disease. Biomed. Pharmacother. 2022, 153, 113482. [Google Scholar] [CrossRef] [PubMed]
  25. Oryan, A.; Kamali, A.; Moshiri, A.; Baghaban Eslaminejad, M. Role of Mesenchymal Stem Cells in Bone Regenerative Medicine: What Is the Evidence? Cells Tissues Organs 2017, 204, 59–83. [Google Scholar] [CrossRef] [PubMed]
  26. Infante, A.; Gener, B.; Vazquez, M.; Olivares, N.; Arrieta, A.; Grau, G.; Llano, I.; Madero, L.; Bueno, A.M.; Sagastizabal, B.; et al. Reiterative infusions of MSCs improve pediatric osteogenesis imperfecta eliciting a pro-osteogenic paracrine response: TERCELOI clinical trial. Clin. Transl. Med. 2021, 11, e265. [Google Scholar] [CrossRef]
  27. Sinder, B.P.; Novak, S.; Wee, N.K.Y.; Basile, M.; Maye, P.; Matthews, B.G.; Kalajzic, I. Engraftment of skeletal progenitor cells by bone-directed transplantation improves osteogenesis imperfecta murine bone phenotype. Stem Cells 2020, 38, 530–541. [Google Scholar] [CrossRef]
  28. Kang, I.H.; Baliga, U.K.; Wu, Y.; Mehrotra, S.; Yao, H.; LaRue, A.C.; Mehrotra, M. Hematopoietic stem cell-derived functional osteoblasts exhibit therapeutic efficacy in a murine model of osteogenesis imperfecta. Stem Cells 2021, 39, 1457–1477. [Google Scholar] [CrossRef]
  29. Guillot, P.V.; Abass, O.; Bassett, J.H.; Shefelbine, S.J.; Bou-Gharios, G.; Chan, J.; Kurata, H.; Williams, G.R.; Polak, J.; Fisk, N.M. Intrauterine transplantation of human fetal mesenchymal stem cells from first-trimester blood repairs bone and reduces fractures in osteogenesis imperfecta mice. Blood 2008, 111, 1717–1725. [Google Scholar] [CrossRef]
  30. Thomas, S.; Jaganathan, B.G. Signaling network regulating osteogenesis in mesenchymal stem cells. J. Cell Commun. Signal. 2022, 16, 47–61. [Google Scholar] [CrossRef]
  31. Grotheer, V.; Skrynecki, N.; Oezel, L.; Windolf, J.; Grassmann, J. Osteogenic differentiation of human mesenchymal stromal cells and fibroblasts differs depending on tissue origin and replicative senescence. Sci. Rep. 2021, 11, 11968. [Google Scholar] [CrossRef] [PubMed]
  32. Choe, M.; Brusgard, J.L.; Chumsri, S.; Bhandary, L.; Zhao, X.F.; Lu, S.; Goloubeva, O.G.; Polster, B.M.; Fiskum, G.M.; Girnun, G.D.; et al. The RUNX2 Transcription Factor Negatively Regulates SIRT6 Expression to Alter Glucose Metabolism in Breast Cancer Cells. J. Cell. Biochem. 2015, 116, 2210–2226. [Google Scholar] [CrossRef] [PubMed]
  33. Chang, C.; Yan, J.; Yao, Z.; Zhang, C.; Li, X.; Mao, H.Q. Effects of Mesenchymal Stem Cell-Derived Paracrine Signals and Their Delivery Strategies. Adv. Healthc. Mater. 2021, 10, e2001689. [Google Scholar] [CrossRef] [PubMed]
  34. Otsuru, S.; Gordon, P.L.; Shimono, K.; Jethva, R.; Marino, R.; Phillips, C.L.; Hofmann, T.J.; Veronesi, E.; Dominici, M.; Iwamoto, M.; et al. Transplanted bone marrow mononuclear cells and MSCs impart clinical benefit to children with osteogenesis imperfecta through different mechanisms. Blood 2012, 120, 1933–1941. [Google Scholar] [CrossRef] [PubMed]
  35. Ramesh, S.; Daniel, D.; Gotherstrom, C.; Madhuri, V. Trophic effects of multiple administration of mesenchymal stem cells in children with osteogenesis imperfecta. Clin. Transl. Med. 2021, 11, e385. [Google Scholar] [CrossRef] [PubMed]
  36. Li, F.; Whyte, N.; Niyibizi, C. Differentiating multipotent mesenchymal stromal cells generate factors that exert paracrine activities on exogenous MSCs: Implications for paracrine activities in bone regeneration. Biochem. Biophys. Res. Commun. 2012, 426, 475–479. [Google Scholar] [CrossRef]
  37. Chen, L.; Tredget, E.E.; Wu, P.Y.; Wu, Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS ONE 2008, 3, e1886. [Google Scholar] [CrossRef]
  38. Heldring, N.; Mager, I.; Wood, M.J.; Le Blanc, K.; Andaloussi, S.E. Therapeutic Potential of Multipotent Mesenchymal Stromal Cells and Their Extracellular Vesicles. Hum. Gene Ther. 2015, 26, 506–517. [Google Scholar] [CrossRef]
  39. Mendt, M.; Kamerkar, S.; Sugimoto, H.; McAndrews, K.M.; Wu, C.C.; Gagea, M.; Yang, S.; Blanko, E.V.R.; Peng, Q.; Ma, X.; et al. Generation and testing of clinical-grade exosomes for pancreatic cancer. JCI Insight 2018, 3, e99263. [Google Scholar] [CrossRef]
  40. Mendt, M.; Rezvani, K.; Shpall, E. Mesenchymal stem cell-derived exosomes for clinical use. Bone Marrow Transplant. 2019, 54, 789–792. [Google Scholar] [CrossRef]
  41. Zhao, M.; Liu, S.; Wang, C.; Wang, Y.; Wan, M.; Liu, F.; Gong, M.; Yuan, Y.; Chen, Y.; Cheng, J.; et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles Attenuate Mitochondrial Damage and Inflammation by Stabilizing Mitochondrial DNA. ACS Nano 2021, 15, 1519–1538. [Google Scholar] [CrossRef] [PubMed]
  42. She, Z.; Xie, M.; Hun, M.; Abdirahman, A.S.; Li, C.; Wu, F.; Luo, S.; Wan, W.; Wen, C.; Tian, J. Immunoregulatory Effects of Mitochondria Transferred by Extracellular Vesicles. Front. Immunol. 2020, 11, 628576. [Google Scholar] [CrossRef] [PubMed]
  43. Crewe, C.; Funcke, J.B.; Li, S.; Joffin, N.; Gliniak, C.M.; Ghaben, A.L.; An, Y.A.; Sadek, H.A.; Gordillo, R.; Akgul, Y.; et al. Extracellular vesicle-based interorgan transport of mitochondria from energetically stressed adipocytes. Cell Metab. 2021, 33, 1853–1868.e11. [Google Scholar] [CrossRef] [PubMed]
  44. Liang, W.; Sagar, S.; Ravindran, R.; Najor, R.H.; Quiles, J.M.; Chi, L.; Diao, R.Y.; Woodall, B.P.; Leon, L.J.; Zumaya, E.; et al. Mitochondria are secreted in extracellular vesicles when lysosomal function is impaired. Nat. Commun. 2023, 14, 5031. [Google Scholar] [CrossRef]
  45. Mukkala, A.N.; Jerkic, M.; Khan, Z.; Szaszi, K.; Kapus, A.; Rotstein, O. Therapeutic Effects of Mesenchymal Stromal Cells Require Mitochondrial Transfer and Quality Control. Int. J. Mol. Sci. 2023, 24, 15788. [Google Scholar] [CrossRef]
  46. Cao, Y.J.; Wei, Z.; Zhang, H.; Zhang, Z.L. Expanding the Clinical Spectrum of Osteogenesis Imperfecta Type V: 13 Additional Patients and Review. Front. Endocrinol. 2019, 10, 375. [Google Scholar] [CrossRef]
  47. Zhang, R.; Liu, Y.; Yan, K.; Chen, L.; Chen, X.R.; Li, P.; Chen, F.F.; Jiang, X.D. Anti-inflammatory and immunomodulatory mechanisms of mesenchymal stem cell transplantation in experimental traumatic brain injury. J. Neuroinflamm. 2013, 10, 106. [Google Scholar] [CrossRef]
  48. Piekarska, K.; Urban-Wojciuk, Z.; Kurkowiak, M.; Pelikant-Malecka, I.; Schumacher, A.; Sakowska, J.; Spodnik, J.H.; Arcimowicz, L.; Zielinska, H.; Tymoniuk, B.; et al. Mesenchymal stem cells transfer mitochondria to allogeneic Tregs in an HLA-dependent manner improving their immunosuppressive activity. Nat. Commun. 2022, 13, 856. [Google Scholar] [CrossRef]
  49. Li, H.; Dai, H.; Li, J. Immunomodulatory properties of mesenchymal stromal/stem cells: The link with metabolism. J. Adv. Res. 2023, 45, 15–29. [Google Scholar] [CrossRef]
  50. Amarasekara, D.S.; Kim, S.; Rho, J. Regulation of Osteoblast Differentiation by Cytokine Networks. Int. J. Mol. Sci. 2021, 22, 2851. [Google Scholar] [CrossRef]
  51. Vanleene, M.; Saldanha, Z.; Cloyd, K.L.; Jell, G.; Bou-Gharios, G.; Bassett, J.H.; Williams, G.R.; Fisk, N.M.; Oyen, M.L.; Stevens, M.M.; et al. Transplantation of human fetal blood stem cells in the osteogenesis imperfecta mouse leads to improvement in multiscale tissue properties. Blood 2011, 117, 1053–1060. [Google Scholar] [CrossRef] [PubMed]
  52. Horwitz, E.M.; Prockop, D.J.; Fitzpatrick, L.A.; Koo, W.W.; Gordon, P.L.; Neel, M.; Sussman, M.; Orchard, P.; Marx, J.C.; Pyeritz, R.E.; et al. Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta. Nat. Med. 1999, 5, 309–313. [Google Scholar] [CrossRef] [PubMed]
  53. Li, A.; Gao, M.; Liu, B.; Qin, Y.; Chen, L.; Liu, H.; Wu, H.; Gong, G. Mitochondrial autophagy: Molecular mechanisms and implications for cardiovascular disease. Cell Death Dis. 2022, 13, 444. [Google Scholar] [CrossRef] [PubMed]
  54. Chen, S.; Liao, Z.; Xu, P. Mitochondrial control of innate immune responses. Front. Immunol. 2023, 14, 1166214. [Google Scholar] [CrossRef]
  55. Mohammadalipour, A.; Dumbali, S.P.; Wenzel, P.L. Mitochondrial Transfer and Regulators of Mesenchymal Stromal Cell Function and Therapeutic Efficacy. Front. Cell Dev. Biol. 2020, 8, 603292. [Google Scholar] [CrossRef]
  56. Collier, J.J.; Olahova, M.; McWilliams, T.G.; Taylor, R.W. Mitochondrial signalling and homeostasis: From cell biology to neurological disease. Trends Neurosci. 2023, 46, 137–152. [Google Scholar] [CrossRef]
  57. Liu, A.R.; Lv, Z.; Yan, Z.W.; Wu, X.Y.; Yan, L.R.; Sun, L.P.; Yuan, Y.; Xu, Q. Association of mitochondrial homeostasis and dynamic balance with malignant biological behaviors of gastrointestinal cancer. J. Transl. Med. 2023, 21, 27. [Google Scholar] [CrossRef]
  58. Liu, L.; Li, Y.; Chen, G.; Chen, Q. Crosstalk between mitochondrial biogenesis and mitophagy to maintain mitochondrial homeostasis. J. Biomed. Sci. 2023, 30, 86. [Google Scholar] [CrossRef]
  59. Wang, Y.; Liu, Y.; Chen, E.; Pan, Z. The role of mitochondrial dysfunction in mesenchymal stem cell senescence. Cell Tissue Res. 2020, 382, 457–462. [Google Scholar] [CrossRef]
  60. Gao, J.; Feng, Z.; Wang, X.; Zeng, M.; Liu, J.; Han, S.; Xu, J.; Chen, L.; Cao, K.; Long, J.; et al. SIRT3/SOD2 maintains osteoblast differentiation and bone formation by regulating mitochondrial stress. Cell Death Differ. 2018, 25, 229–240. [Google Scholar] [CrossRef]
  61. Wang, F.S.; Wu, R.W.; Chen, Y.S.; Ko, J.Y.; Jahr, H.; Lian, W.S. Biophysical Modulation of the Mitochondrial Metabolism and Redox in Bone Homeostasis and Osteoporosis: How Biophysics Converts into Bioenergetics. Antioxidants 2021, 10, 1394. [Google Scholar] [CrossRef] [PubMed]
  62. Lee, S.Y.; An, H.J.; Kim, J.M.; Sung, M.J.; Kim, D.K.; Kim, H.K.; Oh, J.; Jeong, H.Y.; Lee, Y.H.; Yang, T.; et al. PINK1 deficiency impairs osteoblast differentiation through aberrant mitochondrial homeostasis. Stem Cell Res. Ther. 2021, 12, 589. [Google Scholar] [CrossRef] [PubMed]
  63. Loussouarn, C.; Pers, Y.M.; Bony, C.; Jorgensen, C.; Noel, D. Mesenchymal Stromal Cell-Derived Extracellular Vesicles Regulate the Mitochondrial Metabolism via Transfer of miRNAs. Front. Immunol. 2021, 12, 623973. [Google Scholar] [CrossRef] [PubMed]
  64. Mas-Bargues, C. Mitochondria pleiotropism in stem cell senescence: Mechanisms and therapeutic approaches. Free Radic. Biol. Med. 2023, 208, 657–671. [Google Scholar] [CrossRef] [PubMed]
  65. Ahlqvist, K.J.; Suomalainen, A.; Hamalainen, R.H. Stem cells, mitochondria and aging. Biochim. Biophys. Acta 2015, 1847, 1380–1386. [Google Scholar] [CrossRef]
  66. Saeed, K.; Jo, M.H.; Park, J.S.; Alam, S.I.; Khan, I.; Ahmad, R.; Khan, A.; Ullah, R.; Kim, M.O. 17β-Estradiol Abrogates Oxidative Stress and Neuroinflammation after Cortical Stab Wound Injury. Antioxidants 2021, 10, 1682. [Google Scholar] [CrossRef]
  67. Hu, Y.; Huang, L.; Shen, M.; Liu, Y.; Liu, G.; Wu, Y.; Ding, F.; Ma, K.; Wang, W.; Zhang, Y.; et al. Pioglitazone Protects Compression-Mediated Apoptosis in Nucleus Pulposus Mesenchymal Stem Cells by Suppressing Oxidative Stress. Oxid. Med. Cell. Longev. 2019, 2019, 4764071. [Google Scholar] [CrossRef]
  68. Zhang, C.; Meng, Y.; Han, J. Emerging roles of mitochondrial functions and epigenetic changes in the modulation of stem cell fate. Cell. Mol. Life Sci. 2024, 81, 26. [Google Scholar] [CrossRef]
  69. Morganti, C.; Bonora, M.; Marchi, S.; Ferroni, L.; Gardin, C.; Wieckowski, M.R.; Giorgi, C.; Pinton, P.; Zavan, B. Citrate Mediates Crosstalk between Mitochondria and the Nucleus to Promote Human Mesenchymal Stem Cell In Vitro Osteogenesis. Cells 2020, 9, 1034. [Google Scholar] [CrossRef]
  70. Li, H.; Zhang, N.; Wang, Y.; Xia, S.; Zhu, Y.; Xing, C.; Tian, X.; Du, Y. DNA N6-Methyladenine Modification in Eukaryotic Genome. Front. Genet. 2022, 13, 914404. [Google Scholar] [CrossRef]
  71. Zhou, C.; Liu, Y.; Li, X.; Zou, J.; Zou, S. DNA N6-methyladenine demethylase ALKBH1 enhances osteogenic differentiation of human MSCs. Bone Res. 2016, 4, 16033. [Google Scholar] [CrossRef] [PubMed]
  72. Cai, G.P.; Liu, Y.L.; Luo, L.P.; Xiao, Y.; Jiang, T.J.; Yuan, J.; Wang, M. Alkbh1-mediated DNA N6-methyladenine modification regulates bone marrow mesenchymal stem cell fate during skeletal aging. Cell Prolif. 2022, 55, e13178. [Google Scholar] [CrossRef] [PubMed]
  73. Gremminger, V.L.; Harrelson, E.N.; Crawford, T.K.; Ohler, A.; Schulz, L.C.; Rector, R.S.; Phillips, C.L. Skeletal muscle specific mitochondrial dysfunction and altered energy metabolism in a murine model (oim/oim) of severe osteogenesis imperfecta. Mol. Genet. Metab. 2021, 132, 244–253. [Google Scholar] [CrossRef] [PubMed]
  74. Xin, L.; Wen, Y.; Song, J.; Chen, T.; Zhai, Q. Bone regeneration strategies based on organelle homeostasis of mesenchymal stem cells. Front. Endocrinol. 2023, 14, 1151691. [Google Scholar] [CrossRef] [PubMed]
  75. Biswas, L.; Niveria, K.; Verma, A.K. Paradoxical role of reactive oxygen species in bone remodelling: Implications in osteoporosis and possible nanotherapeutic interventions. Explor. Med. 2022, 3, 393–413. [Google Scholar] [CrossRef]
  76. Sheppard, A.J.; Barfield, A.M.; Barton, S.; Dong, Y. Understanding Reactive Oxygen Species in Bone Regeneration: A Glance at Potential Therapeutics and Bioengineering Applications. Front. Bioeng. Biotechnol. 2022, 10, 836764. [Google Scholar] [CrossRef]
  77. Hu, C.; Zhao, L.; Peng, C.; Li, L. Regulation of the mitochondrial reactive oxygen species: Strategies to control mesenchymal stem cell fates ex vivo and in vivo. J. Cell. Mol. Med. 2018, 22, 5196–5207. [Google Scholar] [CrossRef]
  78. Nugud, A.; Sandeep, D.; El-Serafi, A.T. Two faces of the coin: Minireview for dissecting the role of reactive oxygen species in stem cell potency and lineage commitment. J. Adv. Res. 2018, 14, 73–79. [Google Scholar] [CrossRef]
  79. Li, X.; Li, B.; Shi, Y.; Wang, C.; Ye, L. Targeting reactive oxygen species in stem cells for bone therapy. Drug Discov. Today 2021, 26, 1226–1244. [Google Scholar] [CrossRef]
  80. Lee, J.H.; Jung, H.K.; Han, Y.S.; Yoon, Y.M.; Yun, C.W.; Sun, H.Y.; Cho, H.W.; Lee, S.H. Antioxidant effects of Cirsium setidens extract on oxidative stress in human mesenchymal stem cells. Mol. Med. Rep. 2016, 14, 3777–3784. [Google Scholar] [CrossRef]
  81. Varesi, A.; Chirumbolo, S.; Campagnoli, L.I.M.; Pierella, E.; Piccini, G.B.; Carrara, A.; Ricevuti, G.; Scassellati, C.; Bonvicini, C.; Pascale, A. The Role of Antioxidants in the Interplay between Oxidative Stress and Senescence. Antioxidants 2022, 11, 1224. [Google Scholar] [CrossRef] [PubMed]
  82. Fus-Kujawa, A.; Mendrek, B.; Bajdak-Rusinek, K.; Diak, N.; Strzelec, K.; Gutmajster, E.; Janelt, K.; Kowalczuk, A.; Trybus, A.; Rozwadowska, P.; et al. Gene-repaired iPS cells as novel approach for patient with osteogenesis imperfecta. Front. Bioeng. Biotechnol. 2023, 11, 1205122. [Google Scholar] [CrossRef] [PubMed]
  83. Valenti, M.T.; Dalle Carbonare, L.; Mottes, M. Osteogenic Differentiation in Healthy and Pathological Conditions. Int. J. Mol. Sci. 2016, 18, 41. [Google Scholar] [CrossRef] [PubMed]
  84. Dinulescu, A.; Pasarica, A.S.; Carp, M.; Dusca, A.; Dijmarescu, I.; Pavelescu, M.L.; Pacurar, D.; Ulici, A. New Perspectives of Therapies in Osteogenesis Imperfecta-A Literature Review. J. Clin. Med. 2024, 13, 1065. [Google Scholar] [CrossRef]
  85. Ren, X.; Liu, H.; Wu, X.; Weng, W.; Wang, X.; Su, J. Reactive Oxygen Species (ROS)-Responsive Biomaterials for the Treatment of Bone-Related Diseases. Front. Bioeng. Biotechnol. 2021, 9, 820468. [Google Scholar] [CrossRef]
  86. Yan, C.; Shi, Y.; Yuan, L.; Lv, D.; Sun, B.; Wang, J.; Liu, X.; An, F. Mitochondrial quality control and its role in osteoporosis. Front. Endocrinol. 2023, 14, 1077058. [Google Scholar] [CrossRef]
  87. Roca-Portoles, A.; Tait, S.W.G. Mitochondrial quality control: From molecule to organelle. Cell. Mol. Life Sci. 2021, 78, 3853–3866. [Google Scholar] [CrossRef]
  88. Li, Q.; Cheng, J.C.; Jiang, Q.; Lee, W.Y. Role of sirtuins in bone biology: Potential implications for novel therapeutic strategies for osteoporosis. Aging Cell 2021, 20, e13301. [Google Scholar] [CrossRef]
  89. Zhang, T.; Wang, L.; Duan, X.; Niu, Y.; Li, M.; Yun, L.; Sun, H.; Ma, Y.; Guo, Y. Sirtuins mediate mitochondrial quality control mechanisms: A novel therapeutic target for osteoporosis. Front. Endocrinol. 2023, 14, 1281213. [Google Scholar] [CrossRef]
  90. Simic, P.; Zainabadi, K.; Bell, E.; Sykes, D.B.; Saez, B.; Lotinun, S.; Baron, R.; Scadden, D.; Schipani, E.; Guarente, L. SIRT1 regulates differentiation of mesenchymal stem cells by deacetylating β-catenin. EMBO Mol. Med. 2013, 5, 430–440. [Google Scholar] [CrossRef]
  91. Moon, D.K.; Kim, B.G.; Lee, A.R.; In Choe, Y.; Khan, I.; Moon, K.M.; Jeon, R.H.; Byun, J.H.; Hwang, S.C.; Woo, D.K. Resveratrol can enhance osteogenic differentiation and mitochondrial biogenesis from human periosteum-derived mesenchymal stem cells. J. Orthop. Surg. Res. 2020, 15, 203. [Google Scholar] [CrossRef] [PubMed]
  92. Zhou, T.; Yan, Y.; Zhao, C.; Xu, Y.; Wang, Q.; Xu, N. Resveratrol improves osteogenic differentiation of senescent bone mesenchymal stem cells through inhibiting endogenous reactive oxygen species production via AMPK activation. Redox Rep. 2019, 24, 62–69. [Google Scholar] [CrossRef] [PubMed]
  93. Li, J.; Xin, Z.; Cai, M. The role of resveratrol in bone marrow-derived mesenchymal stem cells from patients with osteoporosis. J. Cell. Biochem. 2019, 120, 16634–16642. [Google Scholar] [CrossRef] [PubMed]
  94. Gorrell, L.; Makareeva, E.; Omari, S.; Otsuru, S.; Leikin, S. ER, Mitochondria, and ISR Regulation by mt-HSP70 and ATF5 upon Procollagen Misfolding in Osteoblasts. Adv. Sci. 2022, 9, e2201273. [Google Scholar] [CrossRef]
  95. Ruolan, W.; Liangjiao, C.; Longquan, S. The mTOR/ULK1 signaling pathway mediates the autophagy-promoting and osteogenic effects of dicalcium silicate nanoparticles. J. Nanobiotechnol. 2020, 18, 119. [Google Scholar] [CrossRef]
  96. Chen, J.; Long, F. mTORC1 Signaling Promotes Osteoblast Differentiation from Preosteoblasts. PLoS ONE 2015, 10, e0130627. [Google Scholar] [CrossRef]
  97. Wan, M.C.; Tang, X.Y.; Li, J.; Gao, P.; Wang, F.; Shen, M.J.; Gu, J.T.; Tay, F.; Chen, J.H.; Niu, L.N.; et al. Upregulation of mitochondrial dynamics is responsible for osteogenic differentiation of mesenchymal stem cells cultured on self-mineralized collagen membranes. Acta Biomater. 2021, 136, 137–146. [Google Scholar] [CrossRef]
  98. de Melo Pereira, D.; Eischen-Loges, M.; Birgani, Z.T.; Habibovic, P. Proliferation and Osteogenic Differentiation of hMSCs on Biomineralized Collagen. Front. Bioeng. Biotechnol. 2020, 8, 554565. [Google Scholar] [CrossRef]
  99. Choi, Y.; Yoon, D.S.; Lee, K.M.; Choi, S.M.; Lee, M.H.; Park, K.H.; Han, S.H.; Lee, J.W. Enhancement of Mesenchymal Stem Cell-Driven Bone Regeneration by Resveratrol-Mediated SOX2 Regulation. Aging Dis. 2019, 10, 818–833. [Google Scholar] [CrossRef]
  100. Shakibaei, M.; Shayan, P.; Busch, F.; Aldinger, C.; Buhrmann, C.; Lueders, C.; Mobasheri, A. Resveratrol mediated modulation of Sirt-1/Runx2 promotes osteogenic differentiation of mesenchymal stem cells: Potential role of Runx2 deacetylation. PLoS ONE 2012, 7, e35712. [Google Scholar] [CrossRef]
  101. Tseng, P.C.; Hou, S.M.; Chen, R.J.; Peng, H.W.; Hsieh, C.F.; Kuo, M.L.; Yen, M.L. Resveratrol promotes osteogenesis of human mesenchymal stem cells by upregulating RUNX2 gene expression via the SIRT1/FOXO3A axis. J. Bone Miner. Res. 2011, 26, 2552–2563. [Google Scholar] [CrossRef] [PubMed]
  102. Dai, Z.; Li, Y.; Quarles, L.D.; Song, T.; Pan, W.; Zhou, H.; Xiao, Z. Resveratrol enhances proliferation and osteoblastic differentiation in human mesenchymal stem cells via ER-dependent ERK1/2 activation. Phytomedicine 2007, 14, 806–814. [Google Scholar] [CrossRef] [PubMed]
  103. Han, X.; Jia, G.-f.; Zhu, F. Resveratrol Alleviates Osteoporosis by Promoting Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells via SITR1/PI3K/AKT Pathway. Int. J. Morphol. 2024, 42, 216–224. [Google Scholar] [CrossRef]
  104. Kim, H.N.; Ponte, F.; Warren, A.; Ring, R.; Iyer, S.; Han, L.; Almeida, M. A decrease in NAD+ contributes to the loss of osteoprogenitors and bone mass with aging. NPJ Aging Mech. Dis. 2021, 7, 8. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, J.; Liu, L.; Ding, Z.; Luo, Q.; Ju, Y.; Song, G. Exogenous NAD+ Postpones the D-Gal-Induced Senescence of Bone Marrow-Derived Mesenchymal Stem Cells via Sirt1 Signaling. Antioxidants 2021, 10, 254. [Google Scholar] [CrossRef]
  106. Li, B.; Shi, Y.; Liu, M.; Wu, F.; Hu, X.; Yu, F.; Wang, C.; Ye, L. Attenuates of NAD+ impair BMSC osteogenesis and fracture repair through OXPHOS. Stem Cell Res. Ther. 2022, 13, 77. [Google Scholar] [CrossRef]
  107. Liu, L.; Zhang, W.; Liu, T.; Tan, Y.; Chen, C.; Zhao, J.; Geng, H.; Ma, C. The physiological metabolite α-ketoglutarate ameliorates osteoarthritis by regulating mitophagy and oxidative stress. Redox Biol. 2023, 62, 102663. [Google Scholar] [CrossRef]
  108. Zurek, A.; Mizerska-Kowalska, M.; Slawinska-Brych, A.; Kalawaj, K.; Bojarska-Junak, A.; Kandefer-Szerszen, M.; Zdzisinska, B. Alpha ketoglutarate exerts a pro-osteogenic effect in osteoblast cell lines through activation of JNK and mTOR/S6K1/S6 signaling pathways. Toxicol. Appl. Pharmacol. 2019, 374, 53–64. [Google Scholar] [CrossRef]
  109. Li, Y.; Liu, L.; Li, Y.; Song, W.; Shao, B.; Li, H.; Lin, W.; Li, Q.; Shuai, X.; Bai, M.; et al. Alpha-ketoglutarate promotes alveolar bone regeneration by modulating M2 macrophage polarization. Bone Rep. 2023, 18, 101671. [Google Scholar] [CrossRef]
  110. Wang, Y.; Deng, P.; Liu, Y.; Wu, Y.; Chen, Y.; Guo, Y.; Zhang, S.; Zheng, X.; Zhou, L.; Liu, W.; et al. Alpha-ketoglutarate ameliorates age-related osteoporosis via regulating histone methylations. Nat. Commun. 2020, 11, 5596. [Google Scholar] [CrossRef]
  111. Yamada, M.; Tsukimura, N.; Ikeda, T.; Sugita, Y.; Att, W.; Kojima, N.; Kubo, K.; Ueno, T.; Sakurai, K.; Ogawa, T. N-acetyl cysteine as an osteogenesis-enhancing molecule for bone regeneration. Biomaterials 2013, 34, 6147–6156. [Google Scholar] [CrossRef] [PubMed]
  112. Watanabe, J.; Yamada, M.; Niibe, K.; Zhang, M.; Kondo, T.; Ishibashi, M.; Egusa, H. Preconditioning of bone marrow-derived mesenchymal stem cells with N-acetyl-L-cysteine enhances bone regeneration via reinforced resistance to oxidative stress. Biomaterials 2018, 185, 25–38. [Google Scholar] [CrossRef] [PubMed]
  113. Duryee, M.J.; Dusad, A.; Hunter, C.D.; Kharbanda, K.K.; Bruenjes, J.D.; Easterling, K.C.; Siebler, J.C.; Thiele, G.M.; Chakkalakal, D.A. N-Acetyl Cysteine Treatment Restores Early Phase Fracture Healing in Ethanol-Fed Rats. Alcohol Clin. Exp. Res. 2018, 42, 1206–1216. [Google Scholar] [CrossRef] [PubMed]
  114. Meng, Z.; Liu, J.; Feng, Z.; Guo, S.; Wang, M.; Wang, Z.; Li, Z.; Li, H.; Sui, L. N-acetylcysteine regulates dental follicle stem cell osteogenesis and alveolar bone repair via ROS scavenging. Stem Cell Res. Ther. 2022, 13, 466. [Google Scholar] [CrossRef] [PubMed]
  115. Sandukji, A.; Al-Sawaf, H.; Mohamadin, A.; Alrashidi, Y.; Sheweita, S.A. Oxidative stress and bone markers in plasma of patients with long-bone fixative surgery: Role of antioxidants. Hum. Exp. Toxicol. 2011, 30, 435–442. [Google Scholar] [CrossRef] [PubMed]
  116. Thaler, R.; Khani, F.; Sturmlechner, I.; Dehghani, S.S.; Denbeigh, J.M.; Zhou, X.; Pichurin, O.; Dudakovic, A.; Jerez, S.S.; Zhong, J.; et al. Vitamin C epigenetically controls osteogenesis and bone mineralization. Nat. Commun. 2022, 13, 5883. [Google Scholar] [CrossRef] [PubMed]
  117. Radzki, R.P.; Bienko, M.; Wolski, D.; Oniszczuk, T.; Radzka-Pogoda, A.; Polak, P.; Borzecki, A.; Stasiak, M. Lipoic acid (LA) dose-dependently protects bone losses in the mandible of rats during the development of osteopenia by inhibiting oxidative stress and promoting bone formation. Biomed. Pharmacother. 2022, 146, 112467. [Google Scholar] [CrossRef]
  118. Aydin, A.; Halici, Z.; Akoz, A.; Karaman, A.; Ferah, I.; Bayir, Y.; Aksakal, A.M.; Akpinar, E.; Selli, J.; Kovaci, H. Treatment with α-lipoic acid enhances the bone healing after femoral fracture model of rats. Naunyn Schmiedebergs Arch. Pharmacol. 2014, 387, 1025–1036. [Google Scholar] [CrossRef]
  119. Roberts, J.L.; Moreau, R. Emerging role of α-lipoic acid in the prevention and treatment of bone loss. Nutr. Rev. 2015, 73, 116–125. [Google Scholar] [CrossRef]
  120. Lu, S.Y.; Wang, C.Y.; Jin, Y.; Meng, Q.; Liu, Q.; Liu, Z.H.; Liu, K.X.; Sun, H.J.; Liu, M.Z. The osteogenesis-promoting effects of α-lipoic acid against glucocorticoid-induced osteoporosis through the NOX4, NF-kappaB, JNK and PI3K/AKT pathways. Sci. Rep. 2017, 7, 3331. [Google Scholar] [CrossRef]
  121. Li, C.J.; Sun, L.Y.; Pang, C.Y. Synergistic protection of N-acetylcysteine and ascorbic acid 2-phosphate on human mesenchymal stem cells against mitoptosis, necroptosis and apoptosis. Sci. Rep. 2015, 5, 9819. [Google Scholar] [CrossRef] [PubMed]
  122. Ye, G.; Xie, Z.; Zeng, H.; Wang, P.; Li, J.; Zheng, G.; Wang, S.; Cao, Q.; Li, M.; Liu, W.; et al. Oxidative stress-mediated mitochondrial dysfunction facilitates mesenchymal stem cell senescence in ankylosing spondylitis. Cell Death Dis. 2020, 11, 775. [Google Scholar] [CrossRef] [PubMed]
  123. Shaban, S.; El-Husseny, M.W.A.; Abushouk, A.I.; Salem, A.M.A.; Mamdouh, M.; Abdel-Daim, M.M. Effects of Antioxidant Supplements on the Survival and Differentiation of Stem Cells. Oxid. Med. Cell. Longev. 2017, 2017, 5032102. [Google Scholar] [CrossRef] [PubMed]
  124. Tangtrongsup, S.; Kisiday, J.D. Differential Effects of the Antioxidants N-Acetylcysteine and Pyrrolidine Dithiocarbamate on Mesenchymal Stem Cell Chondrogenesis. Cell. Mol. Bioeng. 2019, 12, 153–163. [Google Scholar] [CrossRef]
  125. Packer, L.; Witt, E.H.; Tritschler, H.J. α-Lipoic acid as a biological antioxidant. Free Radic. Biol. Med. 1995, 19, 227–250. [Google Scholar] [CrossRef]
  126. Voloboueva, L.A.; Liu, J.; Suh, J.H.; Ames, B.N.; Miller, S.S. (R)-α-lipoic acid protects retinal pigment epithelial cells from oxidative damage. Investig. Ophthalmol. Vis. Sci. 2005, 46, 4302–4310. [Google Scholar] [CrossRef]
  127. Perez-Araluce, M.; Jungst, T.; Sanmartin, C.; Prosper, F.; Plano, D.; Mazo, M.M. Biomaterials-Based Antioxidant Strategies for the Treatment of Oxidative Stress Diseases. Biomimetics 2024, 9, 23. [Google Scholar] [CrossRef]
  128. Maldonado, E.; Morales-Pison, S.; Urbina, F.; Solari, A. Aging Hallmarks and the Role of Oxidative Stress. Antioxidants 2023, 12, 651. [Google Scholar] [CrossRef]
  129. Hao, T.; Li, J.; Yao, F.; Dong, D.; Wang, Y.; Yang, B.; Wang, C. Injectable Fullerenol/Alginate Hydrogel for Suppression of Oxidative Stress Damage in Brown Adipose-Derived Stem Cells and Cardiac Repair. ACS Nano 2017, 11, 5474–5488. [Google Scholar] [CrossRef]
  130. Battaglini, M.; Emanet, M.; Carmignani, A.; Ciofani, G. Polydopamine-based nanostructures: A new generation of versatile, multi-tasking, and smart theranostic tools. Nano Today 2024, 55, 102151. [Google Scholar] [CrossRef]
  131. Shafiq, M.; Chen, Y.; Hashim, R.; He, C.; Mo, X.; Zhou, X. Reactive Oxygen Species-Based Biomaterials for Regenerative Medicine and Tissue Engineering Applications. Front. Bioeng. Biotechnol. 2021, 9, 821288. [Google Scholar] [CrossRef] [PubMed]
  132. Liu, J.; Han, X.; Zhang, T.; Tian, K.; Li, Z.; Luo, F. Reactive oxygen species (ROS) scavenging biomaterials for anti-inflammatory diseases: From mechanism to therapy. J. Hematol. Oncol. 2023, 16, 116. [Google Scholar] [CrossRef] [PubMed]
  133. Deng, Z.; Wang, W.; Xu, X.; Nie, Y.; Liu, Y.; Gould, O.E.C.; Ma, N.; Lendlein, A. Biofunction of Polydopamine Coating in Stem Cell Culture. ACS Appl. Mater. Interfaces 2021, 13, 10748–10759. [Google Scholar] [CrossRef] [PubMed]
  134. Han, J.; Wang, J.; Shi, H.; Li, Q.; Zhang, S.; Wu, H.; Li, W.; Gan, L.; Brown-Borg, H.M.; Feng, W.; et al. Ultra-small polydopamine nanomedicine-enabled antioxidation against senescence. Mater. Today Bio 2023, 19, 100544. [Google Scholar] [CrossRef] [PubMed]
  135. Singh, I.; Dhawan, G.; Gupta, S.; Kumar, P. Recent Advances in a Polydopamine-Mediated Antimicrobial Adhesion System. Front. Microbiol. 2020, 11, 607099. [Google Scholar] [CrossRef] [PubMed]
  136. Abdullah, J.A.A.; Jimenez-Rosado, M.; Perez-Puyana, V.; Guerrero, A.; Romero, A. Green Synthesis of Fe(x)O(y) Nanoparticles with Potential Antioxidant Properties. Nanomaterials 2022, 12, 2449. [Google Scholar] [CrossRef]
  137. Yadav, N.; Patel, V.; McCourt, L.; Ruppert, M.; Miller, M.; Inerbaev, T.; Mahasivam, S.; Bansal, V.; Vinu, A.; Singh, S.; et al. Tuning the enzyme-like activities of cerium oxide nanoparticles using a triethyl phosphite ligand. Biomater. Sci. 2022, 10, 3245–3258. [Google Scholar] [CrossRef]
  138. Estevez, A.Y.; Ganesana, M.; Trentini, J.F.; Olson, J.E.; Li, G.; Boateng, Y.O.; Lipps, J.M.; Yablonski, S.E.R.; Donnelly, W.T.; Leiter, J.C.; et al. Antioxidant Enzyme-Mimetic Activity and Neuroprotective Effects of Cerium Oxide Nanoparticles Stabilized with Various Ratios of Citric Acid and EDTA. Biomolecules 2019, 9, 562. [Google Scholar] [CrossRef]
  139. Ju, X.; Fucikova, A.; Smid, B.; Novakova, J.; Matolinova, I.; Matolin, V.; Janata, M.; Belinova, T.; Hubalek Kalbacova, M. Colloidal stability and catalytic activity of cerium oxide nanoparticles in cell culture media. RSC Adv. 2020, 10, 39373–39384. [Google Scholar] [CrossRef]
  140. Chen, M.; Wang, D.; Li, M.; He, Y.; He, T.; Chen, M.; Hu, Y.; Luo, Z.; Cai, K. Nanocatalytic Biofunctional MOF Coating on Titanium Implants Promotes Osteoporotic Bone Regeneration through Cooperative Pro-osteoblastogenesis MSC Reprogramming. ACS Nano 2022, 16, 15397–15412. [Google Scholar] [CrossRef]
  141. Filippova, A.D.; Sozarukova, M.M.; Baranchikov, A.E.; Kottsov, S.Y.; Cherednichenko, K.A.; Ivanov, V.K. Peroxidase-like Activity of CeO2 Nanozymes: Particle Size and Chemical Environment Matter. Molecules 2023, 28, 3811. [Google Scholar] [CrossRef] [PubMed]
  142. Li, J.; Deng, C.; Liang, W.; Kang, F.; Bai, Y.; Ma, B.; Wu, C.; Dong, S. Mn-containing bioceramics inhibit osteoclastogenesis and promote osteoporotic bone regeneration via scavenging ROS. Bioact. Mater. 2021, 6, 3839–3850. [Google Scholar] [CrossRef] [PubMed]
  143. Kang, E.S.; Kim, D.S.; Suhito, I.R.; Choo, S.S.; Kim, S.J.; Song, I.; Kim, T.H. Guiding osteogenesis of mesenchymal stem cells using carbon-based nanomaterials. Nano Converg. 2017, 4, 2. [Google Scholar] [CrossRef] [PubMed]
  144. Thao, N.T.M.; Do, H.D.K.; Nam, N.N.; Tran, N.K.S.; Dan, T.T.; Trinh, K.T.L. Antioxidant Nanozymes: Mechanisms, Activity Manipulation, and Applications. Micromachines 2023, 14, 1017. [Google Scholar] [CrossRef]
  145. Ge, X.; Cao, Z.; Chu, L. The Antioxidant Effect of the Metal and Metal-Oxide Nanoparticles. Antioxidants 2022, 11, 791. [Google Scholar] [CrossRef] [PubMed]
  146. Ullah, M.; Kim, D.-S.; Hun Park, K. Evaluating antioxidant activity of phenolic mediated Fe3O4 nanoparticles using Usnea Longissimma methanol extract. Results Chem. 2022, 4, 100661. [Google Scholar] [CrossRef]
  147. Jiang, D.; Ni, D.; Rosenkrans, Z.T.; Huang, P.; Yan, X.; Cai, W. Nanozyme: New horizons for responsive biomedical applications. Chem. Soc. Rev. 2019, 48, 3683–3704. [Google Scholar] [CrossRef]
  148. Cormode, D.P.; Gao, L.; Koo, H. Emerging Biomedical Applications of Enzyme-Like Catalytic Nanomaterials. Trends Biotechnol. 2018, 36, 15–29. [Google Scholar] [CrossRef]
  149. Shang, Y.; Liu, F.; Wang, Y.; Li, N.; Ding, B. Enzyme Mimic Nanomaterials and Their Biomedical Applications. Chembiochem 2020, 21, 2408–2418. [Google Scholar] [CrossRef]
  150. Ren, X.; Chen, D.; Wang, Y.; Li, H.; Zhang, Y.; Chen, H.; Li, X.; Huo, M. Nanozymes-recent development and biomedical applications. J. Nanobiotechnol. 2022, 20, 92. [Google Scholar] [CrossRef]
  151. Ikram, R.; Shamsuddin, S.A.A.; Mohamed Jan, B.; Abdul Qadir, M.; Kenanakis, G.; Stylianakis, M.M.; Anastasiadis, S.H. Impact of Graphene Derivatives as Artificial Extracellular Matrices on Mesenchymal Stem Cells. Molecules 2022, 27, 379. [Google Scholar] [CrossRef] [PubMed]
  152. Gurunathan, S.; Arsalan Iqbal, M.; Qasim, M.; Park, C.H.; Yoo, H.; Hwang, J.H.; Uhm, S.J.; Song, H.; Park, C.; Do, J.T.; et al. Evaluation of Graphene Oxide Induced Cellular Toxicity and Transcriptome Analysis in Human Embryonic Kidney Cells. Nanomaterials 2019, 9, 969. [Google Scholar] [CrossRef]
  153. Carmignani, A.; Battaglini, M.; Sinibaldi, E.; Marino, A.; Vighetto, V.; Cauda, V.; Ciofani, G. In Vitro and Ex Vivo Investigation of the Effects of Polydopamine Nanoparticle Size on Their Antioxidant and Photothermal Properties: Implications for Biomedical Applications. ACS Appl. Nano Mater. 2022, 5, 1702–1713. [Google Scholar] [CrossRef]
  154. Gaharwar, U.S.; Paulraj, R. Iron Oxide Nanoparticles Induced Oxidative Damage in Peripheral Blood Cells of Rat. J. Biomed. Sci. Eng. 2015, 08, 274–286. [Google Scholar] [CrossRef]
  155. Mehta, K.J. Iron Oxide Nanoparticles in Mesenchymal Stem Cell Detection and Therapy. Stem Cell Rev. Rep. 2022, 18, 2234–2261. [Google Scholar] [CrossRef] [PubMed]
  156. Huang, T.; Zhang, T.; Jiang, X.; Li, A.; Su, Y.; Bian, Q.; Wu, H.; Lin, R.; Li, N.; Cao, H.; et al. Iron oxide nanoparticles augment the intercellular mitochondrial transfer-mediated therapy. Sci. Adv. 2021, 7, eabj0534. [Google Scholar] [CrossRef]
  157. Li, X.; Wei, Z.; Lv, H.; Wu, L.; Cui, Y.; Yao, H.; Li, J.; Zhang, H.; Yang, B.; Jiang, J. Iron oxide nanoparticles promote the migration of mesenchymal stem cells to injury sites. Int. J. Nanomed. 2019, 14, 573–589. [Google Scholar] [CrossRef]
Figure 1. MSCs’ treatment of OI is multifaceted and involves several aspects of MSC biology, including their ability to differentiate into osteoblasts, their immunomodulatory effects, and their capacity to secrete bioactive molecules that can influence the bone healing process. (A) Differentiation into osteoblasts: BMP-2 stimulates the expression of Runx2 and OSX, and mitochondria provide energy for this process, promoting the differentiation of mesenchymal stem cells into bone [50]. (B) Paracrine signaling: MSCs release soluble mediators (cytokines, enzymes, and growth factors) that promote cell migration and proliferation to enhance bone healing and quality [14,33]. (C) Immunomodulatory effects: MSCs alter the cytokine profile in the body, elevating anti-inflammatory cytokines and decreasing pro-inflammatory cytokines [13].
Figure 1. MSCs’ treatment of OI is multifaceted and involves several aspects of MSC biology, including their ability to differentiate into osteoblasts, their immunomodulatory effects, and their capacity to secrete bioactive molecules that can influence the bone healing process. (A) Differentiation into osteoblasts: BMP-2 stimulates the expression of Runx2 and OSX, and mitochondria provide energy for this process, promoting the differentiation of mesenchymal stem cells into bone [50]. (B) Paracrine signaling: MSCs release soluble mediators (cytokines, enzymes, and growth factors) that promote cell migration and proliferation to enhance bone healing and quality [14,33]. (C) Immunomodulatory effects: MSCs alter the cytokine profile in the body, elevating anti-inflammatory cytokines and decreasing pro-inflammatory cytokines [13].
Pharmaceuticals 17 01297 g001
Figure 2. Mitochondrial metabolism is indispensable in meeting the diverse metabolic demands of MSCs. An increase in reactive oxygen species (ROS) generated by OXPHOS leads to the activation of AMP-activated protein kinase (AMPK). A decrease in ROS levels is associated with the maintenance of MSC function [63,64].
Figure 2. Mitochondrial metabolism is indispensable in meeting the diverse metabolic demands of MSCs. An increase in reactive oxygen species (ROS) generated by OXPHOS leads to the activation of AMP-activated protein kinase (AMPK). A decrease in ROS levels is associated with the maintenance of MSC function [63,64].
Pharmaceuticals 17 01297 g002
Figure 3. ROS are crucial for bone regeneration and reconstruction, but their levels must be regulated for optimal therapeutic outcomes. Excessive ROS can impair stem cell function and hinder healing. As MSCs differentiate into osteoblasts, ROS levels decrease and antioxidant enzymes upregulate, promoting osteoblast differentiation and reconstruction [75,76,79].
Figure 3. ROS are crucial for bone regeneration and reconstruction, but their levels must be regulated for optimal therapeutic outcomes. Excessive ROS can impair stem cell function and hinder healing. As MSCs differentiate into osteoblasts, ROS levels decrease and antioxidant enzymes upregulate, promoting osteoblast differentiation and reconstruction [75,76,79].
Pharmaceuticals 17 01297 g003
Table 1. Strategies for regulating mitochondrial metabolism.
Table 1. Strategies for regulating mitochondrial metabolism.
Metabolic ModulationSummaryKey MechanismBiological EffectRef.
ResveratrolResveratrol can maintain the therapeutic potential of MSCs during long-term culture by acting through the SIRT1-SOX2 axisSIRT1-SOX2 AxisImproved bone regeneration[74]
Resveratrol promotes osteogenic differentiation and mitochondrial biogenesis in periosteum-derived MSCsMitochondrial BiogenesisEnhanced osteogenesis[75]
Runx2 acetylation/deacetylation is a main mechanism during osteogenic differentiation in MSCs in vitroSirt-1/Runx2 PathwayPromoted osteogenic differentiation[76]
Resveratrol enhances osteogenesis in human MSCs by upregulating the expression of the RUNX2 gene through the SIRT1/FOXO3A pathwaySIRT1/FOXO3A AxisEnhanced osteogenesis[77]
Resveratrol enhances the proliferation and osteoblastic differentiation of human MSCs through ER-dependent ERK1/2 activationERK1/2 ActivationIncreased proliferation and differentiation[78]
Resveratrol can attenuate osteoporosis by promoting the osteogenic differentiation of bone marrow MSCs through the SIRT1/PI3K/AKT pathwaySIRT1/PI3K/AKT PathwayAttenuation of osteoporosis[80]
NAD+NAD+ levels affect osteoblastogenesis in cells from old mice, showing that reduced NAD+ impairs mineralization under osteogenic conditionsNAD+ Level ImpactImpaired mineralization[81]
NAD+ levels impair mitochondrial fusion, leading to mitochondrial dysfunction and reduced activity of OXPHOS, which subsequently blocks osteogenesis and diminishes bone fracture healingMitochondrial DysfunctionBlocked osteogenesis and fracture repair[82]
Exogenous NAD+ can delay senescence in bone marrow-derived MSCs through the activation of the Sirt1 signaling pathwaySirt1 Signaling ActivationDelayed senescence in MSCs[83]
miR-34a uses the NAD+-Sirt1 pathway to further mediate its role in MSC replicative senescence and natural senescence by targeting NamptNAD+-Sirt1 PathwayAmeliorated MSC senescence[84]
α-KGα-KG promotes alveolar bone regeneration following jawbone injury by modulating macrophage polarization towards an M2 phenotype, which is conducive to healing and tissue repairModulation of Macrophage PolarizationEnhanced bone regeneration[85]
α-KG supplementation increases bone mass in aged mice and accelerates bone regeneration by decreasing histone methylations and upregulating BMP signaling and Nanog expressionRegulation of Histone ModificationsAccelerated bone regeneration[86]
α-KG influences stem cell fate and promotes osteogenic differentiation through mitochondrial nuclear signalingMitochondrial SignalingPromoted osteogenic differentiation[68]
Table 2. Summary of antioxidant strategies of different antioxidants.
Table 2. Summary of antioxidant strategies of different antioxidants.
AntioxidantsSummaryMain MechanismBiological EffectRef.
NACNAC and ascorbic acid protect MSCs from oxidative stress-induced mitochondrial dysfunction by enhancing mitochondrial fusion and reducing fragmentationMitochondrial ProtectionEnhanced mitochondrial function[121]
NAC inhibit ROS production and rescue MSCs from senescence by improving mitochondrial function and reducing oxidative stressMitochondrial ProtectionRescued MSCs from senescence[122]
NAC and mitochondria-targeted ubiquinone can reduce oxidative damage and improve the survival and differentiation of MSCsReduction in ROSImproved survival and differentiation[123]
NAC and pyrrolidine dithiocarbamate reduce intracellular ROS and their effects on MSC chondrogenesisReduction in ROSReduced oxidative stress in chondrogenesis[124]
α-LAα-LA has potential effects on MSCs by protecting them from oxidative stressEnhancement of Antioxidant MechanismsProtected MSCs from oxidative stress[125]
α-LA can protect mitochondria from oxidative stress by enhancing cellular antioxidant mechanismsEnhancement of Antioxidant MechanismsProtected mitochondria from oxidative stress[126]
Vitamin CVitamin C hydrogel scaffolds enhance cell survival and minimize ROS levels under H2O2-induced oxidative stress conditionsFree Radical ScavengingImproved cell survival under oxidative stress[127]
Vitamin C can protect MSCs from oxidative stress-induced mitochondrial dysfunctionMitochondrial ProtectionProtected MSCs from oxidative stress-induced mitochondrial dysfunction[128]
Table 3. Strategy of scavenging ROS by new biomaterials.
Table 3. Strategy of scavenging ROS by new biomaterials.
Biological MaterialSummaryMain MechanismBiological EffectRef.
GOGO has potential to mitigate cadmium-induced cytotoxicity and oxidative stressMitigation of cadmium-induced cytotoxicityReduced oxidative stress[151]
GO exposure leads to significant decreases in mitochondrial membrane potential and ATP productionMitochondrial dysfunction and ATP reductionDecreased mitochondrial membrane potential and ATP generation[152]
Fullerenol/Alginate HydrogelFullerenol/alginate hydrogel can effectively scavenge superoxide anion and hydroxyl radicals, improving the survival of stem cells under oxidative stress Antioxidant activity and cell deliverySuppression of oxidative stress damage in MSCs[129]
Fullerene/alginate hydrogels in bone regeneration strategies bond to modulation of mitochondrial function and redox homeostasisOrganelle homeostasis and bone regenerationImproved bone regeneration through organelle homeostasis of MSCs[74]
PDAPDA-coated substrate can reduce oxidative stress and mitochondrial damage in mesenchymal stem cells, enhancing their expansion and reducing senescenceAntioxidant properties and cellular protectionReduction in oxidative stress and mitochondrial damage in MSCs[133]
PAD nanoparticles have enhanced antioxidant properties and cellular uptake, which could be beneficial for protecting MSCs from oxidative stressAntioxidant effects and mitochondrial healthEnhancement of antioxidant properties and cellular uptake[153]
CeNPsCeNPs support the mitochondrial health of MSCs in regenerative contextsAntioxidant and anti-inflammatory effectsPotential applications in wound healing and tissue regeneration[154]
CeNPs has potential in mitigating oxidative stress and protecting mitochondrial function in various cell typesBiocompatibility and cytotoxicityReduction in ROS levels and protection against oxidative stress[155]
Fe3O4 nanoparticlesIron oxide nanoparticles could augment intercellular mitochondrial transfer from MSCs Oxidative stress and cytotoxicityEnhanced intercellular mitochondrial transfer from MSCs to diseased cells[156]
Fe3O4 nanoparticles can be used for magnetic targeting and delivery of mesenchymal stem cells, improving their retention and therapeutic effectsMagnetic targeting and cell deliveryImproved cell retention and therapeutic effects in various disease models[157]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Guo, Q.; Zhai, Q.; Ji, P. The Role of Mitochondrial Homeostasis in Mesenchymal Stem Cell Therapy—Potential Implications in the Treatment of Osteogenesis Imperfecta. Pharmaceuticals 2024, 17, 1297. https://doi.org/10.3390/ph17101297

AMA Style

Guo Q, Zhai Q, Ji P. The Role of Mitochondrial Homeostasis in Mesenchymal Stem Cell Therapy—Potential Implications in the Treatment of Osteogenesis Imperfecta. Pharmaceuticals. 2024; 17(10):1297. https://doi.org/10.3390/ph17101297

Chicago/Turabian Style

Guo, Qingling, Qiming Zhai, and Ping Ji. 2024. "The Role of Mitochondrial Homeostasis in Mesenchymal Stem Cell Therapy—Potential Implications in the Treatment of Osteogenesis Imperfecta" Pharmaceuticals 17, no. 10: 1297. https://doi.org/10.3390/ph17101297

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop