Abstract
-
The pathogenesis of steroid-induced osteonecrosis of the femoral head (SONFH) remains unclear; however, emerging evidence suggests that mitochondrial injury plays a significant role.
-
This review aims to elucidate the involvement of mitochondrial dysfunction in SONFH and explore potential therapeutic targets.
-
A comprehensive literature search was conducted in PubMed, Web of Science, and Elsevier ScienceDirect, focusing on mitochondrial homeostasis, including mitophagy, mitochondrial biogenesis, mitochondrial dynamics, and oxidative stress in SONFH. Ultimately, we included and analyzed a total of 16 studies.
-
Glucocorticoids initially promote but later inhibit mitochondrial biogenesis in osteoblasts, leading to excessive ROS production and mitochondrial dysfunction. This dysfunction impairs osteoblast survival and bone formation, contributing to SONFH progression.
-
Key proteins such as mitochondrial transcription factor A (TFAM) and peroxisome proliferator-activated receptor γ coactivator 1-α (PGC1α) are potential therapeutic targets for promoting mitochondrial biogenesis and reducing ROS-induced damage.
-
Enhancing mitochondrial function and reducing oxidative stress in osteoblasts may prevent or slow the progression of SONFH. Future research should focus on developing these strategies.
Introduction
Steroids, widely used to treat various inflammatory and autoimmune diseases (1), can increase the risk of femoral head necrosis with long-term or high-dose use (2), which in severe cases can lead to disability (3). Steroid-induced osteonecrosis of the femoral head (SONFH) may account for 26.35% of all cases of femoral head osteonecrosis among men and 55.75% of cases among women (4). However, due to a lack of effective early intervention in clinical settings, over 80% of SONFH patients require total hip arthroplasty (5). The imaging results for patients with SONFH are presented in Fig. 1.
While the pathogenesis of SONFH is still entirely unclear, steroids are thought to injure vascular endothelial cells, perturb osteogenic differentiation, and induce hyperactivity of osteoclasts as well as apoptosis of osteoblasts, osteocytes, and bone marrow mesenchymal stromal cells (BMSCs) (6, 7). These alterations interfere with bone remodeling, leading to the local loss of bone mass and potentially causing femoral head collapse (7, 8). At present, drugs such as bisphosphonates and calcitonin are used to treat early stages of SONFH. Although these drugs can inhibit osteoclast activity and show good clinical effects in the early stages of SONFH, they cannot sustain effective bone turnover (9).
Glucocorticoids (GCs) significantly impact bone cell proliferation, differentiation, function, and apoptosis. They inhibit osteoblast differentiation by promoting adipogenic factors and suppressing the Wnt signaling pathway, reduce osteoblast proliferation, increase oxidative stress, and elevate reactive oxygen species (ROS) production. Prolonged exposure leads to mitochondrial dysfunction and apoptosis of osteoblasts and osteocytes by disrupting mitochondrial membrane potential and releasing pro-apoptotic factors like cytochrome C. These effects contribute to bone loss and osteoporosis (10).
Emerging evidence suggests that SONFH involves mitochondrial injury (11), making it a potential novel therapeutic target. Mitochondria are essential for cellular energy production, regulating oxidative stress, and apoptosis. Steroids can disrupt mitochondrial homeostasis, leading to excessive ROS production and apoptosis. Impaired mitochondrial function due to steroid exposure can exacerbate tissue damage and necrosis. Thus, understanding and targeting mitochondrial dysfunction could provide new avenues for treating SONFH. Disruption of mitochondrial homeostasis has already been implicated in other diseases (12, 13), and it is known to impair osteogenic differentiation (14, 15). The steroid dexamethasone appears to induce apoptosis in osteoblastic cells through a pathway mediated by mitochondria (16). Bone cell apoptosis in SONFH appears to involve the impairment of mitochondrial function and the release of cytochrome C (17).
Based on relevant research published in recent years, we carried out this review to expound on the relationship between mitochondrial homeostasis and the pathogenesis of SONFH, including the potential mechanisms of mitophagy, mitochondrial biogenesis, mitochondrial dynamics, and ROS generation, with the aim of assisting orthopedists in gaining a comprehensive understanding of these mechanisms and providing a novel target for SONFH treatment.
Searching strategies
This narrative review is conducted and reported according to the Scale for the Assessment of Narrative Review Articles (SANRA) guidelines to ensure a structured and comprehensive review process (18).
We searched the literature in PubMed, Web of Science, and Elsevier ScienceDirect. The search strategy was as follows: (osteonecrosis of the femoral head or aseptic necrosis of the femoral head or steroid-induced osteonecrosis of the femoral head or glucocorticoid-induced osteonecrosis of the femoral head or ONFH or ANFH or SONFH or GONFH) and (mitochondrion or mitochondrial dysfunction or mitochondrial homeostasis or mitophagy or mitochondrial biogenesis or mitochondrial dynamics or oxidative stress).
The research explored the role of mitochondrial homeostasis (mitophagy, mitochondrial biogenesis, mitochondrial dynamics, and mitochondrial ROS metabolism) in SONFH. The exclusion criteria are as follows: (i) Studies that did not specifically explore mitochondrial homeostasis, including mitophagy, mitochondrial biogenesis, mitochondrial dynamics, or mitochondrial ROS metabolism in the context of SONFH, were excluded; (ii) Research that did not involve subjects relevant to SONFH, such as studies on other types of osteonecrosis or unrelated diseases, was excluded; (iii) Review articles, case reports, and studies without experimental or clinical data related to mitochondrial functions in SONFH were excluded; (iv) Studies that lacked sufficient data or had inconclusive results regarding the role of mitochondria in SONFH were excluded.
Literature selection followed these steps: first, all retrieved studies were imported into Endnote X7 and duplicates were excluded. Then, irrelevant literature was excluded by two independent researchers based on titles and abstracts. Finally, the same two researchers included the studies that met the selection criteria after scrutinizing full texts. Any disagreement was resolved by discussion with a third researcher.
Following the aforementioned retrieval strategy, a total of 262 studies were identified through a search. After removing duplicates, 108 studies were left, and then 75 studies were excluded based on the irrelevance of their titles and abstracts. Additionally, 17 studies were removed because they did not meet the selection criteria. Finally, 16 studies were included in this review.
Mitochondrial homeostasis
The mitochondrion acts as a hub of metabolic and signaling processes. It comprises four functional regions: outer membrane, intermembrane space, inner membrane, and matrix. These four regions participate in oxidative phosphorylation to produce ATP, fatty acid oxidation, calcium buffering, phospholipid synthesis, generation of ROS, maintenance, synthesis of iron-sulfur clusters, and innate immune signaling (19). Mitochondria are crucial for energy production through oxidative phosphorylation, which occurs in the inner mitochondrial membrane. The electron transport chain (ETC) drives the synthesis of ATP by creating a proton gradient used by ATP synthase. During this process, ROS are produced as by-products. While ROS are vital for cell signaling, excessive ROS can cause oxidative stress, leading to cellular damage. Maintaining mitochondrial homeostasis, therefore, involves a balance between ATP production and the management of ROS levels (Fig. 2).
The cell’s population and morphology of mitochondria are highly dynamic (20). The lifespan of mitochondria is short, and the function of new mitochondria noticeably declines after only a few days. This means that mitochondria are essentially in an environment of continuous exposure to high levels of ROS (21, 22), which damage mitochondrial proteins and induce mutations in mitochondrial DNA (23). Older mitochondria continuously undergo fission and fusion to produce new mitochondria, or they undergo mitophagy to be removed from the cell as waste (24). During their function, mitochondria must maintain a robust antioxidant system that can keep levels of potentially toxic ROS in check. The balance among mitochondrial biogenesis, mitophagy, and the dynamics of mitochondrial fusion/fission – collectively referred to as ‘mitochondrial homeostasis’ – is important for maintaining a stable pool of functional mitochondria in the cell (25).
When damaged mitochondria cannot be degraded by mitophagy, the normal ROS metabolism becomes disordered, and abundant Ca2+ and cytochrome C are released into the cytosol, triggering cell apoptosis. In this way, mitochondrial damage has already been implicated in diabetes, neurodegenerative diseases, and the effects of aging (12, 13).
Mitochondrial homeostasis in SONFH
While the pathogenesis of SONFH remains unclear, the most widely accepted mechanism involves apoptosis of osteogenic cells, abnormal osteogenic differentiation, and impaired microcirculation (6). Recently, a study demonstrated that glucocorticoids may inhibit the transcriptional activity of glucose transporter member 1 (GLUT1) in SONFH, which is responsible for transporting glucose from the extracellular space into the cell. This inhibition reduces the amount and activity of GLUT1 in cells, leading to decreased mitochondrial activity, reduced ATP production, the promotion of apoptosis, and inhibition of osteoblast ossification via the GC/GR/GLUT1 axis (26). In addition, another study found that BID and FTH1, two genes involved in the mitochondrial apoptotic pathway, were highly expressed in SONFH and promoted apoptosis of bone cells (27).
A key driver of the disease appears to be oxidative stress: long-term and/or high doses of corticosteroids within the hypoxic environment of the femoral head produce more ROS than osteogenic cells can remove (17, 28, 29). These steroids can increase mitochondrial activity, leading to higher levels of oxidative phosphorylation, but they cause a decrease in the activity of antioxidant enzymes such as SOD1, HO-1, and catalase, which in turn generates more ROS (30). The abundant ROS disrupts the outer and inner mitochondrial membranes, disturbing oxidative phosphorylation, resulting in a decrease in energy production. Furthermore, the damaged membranes allow the leakage of Ca2+ and cytochrome C into the cytosol, the fluid within the cell. Mitochondrial dysfunction occurs when cytochrome C, crucial for ATP production, is released into the cytoplasm, reducing ATP and affecting energy supply; this release also triggers apoptosis by forming a complex with apoptotic protease activating factor-1 and caspase-9, activating the caspase cascade. Excess Ca2+ can lower mitochondrial membrane potential, decrease ATP synthesis, increase membrane permeability, and release pro-apoptotic factors, while both cytochrome C release and Ca2+ influx elevate oxidative stress, leading to cell damage and apoptosis (29, 31, 32). This inhibits osteogenic differentiation and bone mineralization function (33, 34, 35).
Mitochondria injury also induces the apoptosis of bone microvascular endothelial cells (36), reducing blood flow to the femoral head and creating a hypercoagulable state conducive to ischemia and thrombus formation (37). Ischemia and hypoxia of the femoral head impair the function of the oxygenated mitochondrial respiratory chain to produce excessive ROS, exacerbating the development of SONFH. Oxygen is crucial for the operation of the mitochondrial ETC. Under ischemic and hypoxic conditions, mitochondria are unable to carry out normal electron transfer, leading to the accumulation of electrons between various complexes of the ETC. Electrons are prone to leak from ETC complexes I and III into the mitochondrial matrix, reacting with oxygen molecules to generate superoxide (O2 −), a type of ROS, which leads to the overexpression of SOD2. SOD2 is an upregulated gene in SONFH, and this may lead to the accumulation of hydrogen peroxide (27). An excessive increase in oxides leads to mitochondrial dysfunction and ultimately cell apoptosis.
P53, a tumor suppressor protein, contributes to the apoptosis of osteogenic cells in the femoral head. After being activated by the glucocorticoid receptor, p53 translocates to the mitochondria, where it forms a complex with cyclophilin-D (38), disrupting the normal mitochondrial membrane potential. Additionally, oxidative stress can upregulate p53 in BMSCs, further exacerbating mitochondrial dysfunction (33).
Ferroptosis, a distinct form of programmed cell death characterized by mitochondrial damage, can lead to cell death via iron-dependent mechanisms. It has been shown to play a role in the pathogenesis of femoral head necrosis. A recent study has shown that increased p53 expression inhibits the expression of SLC7A1 and GPX4 in BMSCs, MC3T3-E1, and MLOY4 cells in SONFH, leading to a reduction in intracellular GSH levels. This reduction results in elevated levels of MDA, ROS, and lipid ROS within the cells, causing mitochondrial shrinkage, a decrease in mitochondrial cristae, and impaired mitochondrial function, ultimately leading to ferroptosis and abnormal differentiation of osteoblasts and apoptosis of bone cells (39, 40). FTH1 gene associated with ferroptosis is highly expressed in SONFH (27). Antioxidants such as melatonin (MT) have been shown to attenuate GCs-induced ROS generation and promote osteogenic differentiation by inhibiting ferroptosis mediated through MT2, which can relieve the SONFH process (40).
Furthermore, mitochondrial dysfunction may affect the differentiation of adipocytes, promote apoptosis of bone cells, and accelerate the process of SONFH. The Wnt/β-catenin pathway is crucial for regulating the differentiation of BMSCs, with increased expression promoting osteoblast development (41, 42). In SONFH rats, glucocorticoid exposure has been shown to disrupt the osteogenic and adipogenic differentiation of BMSCs by inhibiting β-catenin signaling (43). In another study, it was also proven that adipocyte formation was promoted and bone cell apoptosis was accelerated when the Wnt/β-catenin pathway was inhibited in SONFH (42). Additionally, mitochondrial damage produces excessive ROS, which promote the binding of FoxO to β-catenin, ultimately leading to reduced osteoblast formation (41).
These considerations suggest that mitochondrial dysfunction is associated with the development of SONFH, and restoring the function of mitochondria may be an effective treatment against SONFH. The following sections elaborate on the pathophysiological mechanisms and the potential treatment of molecular targets involved in the four important components of mitochondrial homeostasis: mitophagy, mitochondrial biogenesis, mitochondrial dynamics, and mitochondrial ROS metabolism.
Mitophagy
Mitophagy, in which damaged or ineffective mitochondria are delivered to autophagosomes for degradation (44), is one of the major mitochondrial quality control processes (45). An appropriate level of mitophagy is required to prevent oxidative stress and apoptosis by reducing ROS and proapoptotic factors released by the damaged mitochondria (46). Oxidative stress initially promotes mitophagy in BMSCs via a pathway involving c-Jun N-terminal kinases, but later it inhibits mitophagy and promotes apoptosis (47). When oxidative stress damages mitochondria, leading to depolarization of their inner membranes, the Pink1/Parkin system acts as a sensor for mitochondrial quality and is activated (48). PINK1 and Parkin function as the first steps of a signaling pathway that activates mitochondrial quality control pathways in response to mitochondrial damage. PINK1 accumulates on the outer mitochondrial membrane, where it phosphorylates ubiquitin and recruits the E3 ubiquitin ligase Parkin from the cytosol to the outer membrane. Parkin ubiquitinates several proteins in the outer membrane, and the adaptor proteins p62/SQSTM1 bind to these ubiquitin moieties and autophagic protein LC3-II on autophagosomes to mediate sequestration of the organelle in an autophagosomal membrane, allowing mitophagy to proceed (48, 49, 50) (Fig. 3).
Deficiency of PINK1 inhibits osteoblast differentiation and increases mitochondrial production of ROS (15). Upregulating Parkin exerts analogous effects, but only if P53 is simultaneously downregulated (33), probably because P53 can inhibit the activity of Parkin by binding to its RING0 region (46, 51), and downregulation of P53 can effectively reduce the decline of mitochondrial membrane potential (38). Dexamethasone appears to inhibit hypoxia-induced mitophagy by downregulating BNIP3, NIX, and LC3-II, which are critical for the mitophagy process. BNIP3 and NIX are pro-apoptotic proteins that promote the removal of damaged mitochondria by mediating their recognition and engulfment by autophagosomes, while LC3-II is essential for the formation and elongation of autophagosomes. This inhibition, therefore, promotes apoptosis of osteocytes. These effects of dexamethasone can be reversed by overexpression of Hypoxia-inducible factor-1α (52). A study has shown that high-dose hormones can effectively inhibit the expression of PINK1, Parkin, and LC3-II, upregulate p62, significantly weaken the autophagy function of osteoblast mitochondria, and affect the survival rate of osteoblasts (53). However, Vitamin K2 administration can significantly attenuate the dexamethasone-induced downregulation of LC3-II, PINK1, and Parkin in osteoblasts, thereby restoring mitophagic processes and normal osteoblastic activity (53).
Mitochondrial biogenesis
While undifferentiated BMSCs and osteoblasts show relatively weak mitochondrial activity, osteogenic induction induces a large increase in oxygen consumption. To provide this additional metabolic power, mitochondrial biogenesis is strongly upregulated (35, 54). Glucocorticoid initially promotes mitochondrial biogenesis in osteoblasts but later inhibits it (55). The resulting excess levels of ROS may further inhibit biogenesis by interfering with the cytosolic synthesis of mitochondrial proteins and their targeting to the appropriate submitochondrial compartments (25, 56).
Several proteins promote osteogenesis by promoting mitochondrial biogenesis. For example, adding mitochondrial transcription factor A (TFAM) to osteocytes under oxidative stress protects them from apoptosis (57). Taurine can reverse the downregulation of TFAM in osteocytes under glucocorticoid and hypoxia stimulation and prevent SONFH in rabbits (58). The protein ‘peroxisome proliferator-activated receptor γ coactivator 1-α’ (PGC-1α) interacts with the transcription factors NRF1 and NRF2, which in turn stimulate the expression of TFAM to promote mitochondrial biogenesis (59), and the polypeptide hormone liraglutide, which is a glucagon-like peptide-1 (GLP-1) receptor agonist, stimulates signaling involving the GLP-1 receptor, cyclic AMP, phosphorylated AMP kinase, and the Adipo1 receptor to promote mitochondrial biogenesis and osteogenesis (60). Resveratrol, a polyphenolic compound found in various plants, can activate sirtuin 1 and rescue mitochondrial biogenesis in osteoblasts that have been exposed to dexamethasone (61). A neuroprotective bovine colostrum has been shown to attenuate mitochondria-induced apoptosis in osteoblasts treated with dexamethasone, potentially by activating the Hsp70 system to correct and stabilize the structure of proteins necessary for mitochondrial biogenesis (62, 63). These examples build a strong case that these proteins could be promising targets. Regulating their activity may enhance mitochondrial biosynthesis in osteoblasts, thereby promoting osteogenesis and aiding in the repair of femoral head necrosis.
Mitochondria dynamics
New mitochondria can be generated from old ones through fusion mediated by mitochondrial fusion protein and optic atrophy protein 1 (64), and by fission mediated by dynamic-associated protein 1 (DRP1), mitochondrial fission protein 1 (FIS1), and mitochondrial fission factor (MFF) (23). These two processes must be kept within appropriate limits as part of mitochondrial homeostasis. Excessive fission, for example, can lead to abnormal morphology, permeabilization of the outer mitochondrial membrane, release of ROS, and cell death (65). As glucocorticoids increase mitochondrial fusion, it initially enhances mitochondrial function. However, when followed by excessive fission, it leads to mitochondrial dysfunction by promoting the release of pro-apoptotic factors like cytochrome C into the cytosol, ultimately resulting in the apoptosis of osteoblasts (55).
Treating endothelial progenitor cells with glucocorticoid methylprednisolone increases the number of granulated mitochondria, raises the level of ROS, upregulates MFF and FIS1, and eliminates normal cristae structure (66). On the other hand, treating BMSCs with dexamethasone downregulates DRP1, MFF, and FIS1 (34). Thus, further research is needed to clarify how GCs alter mitochondrial dynamics. Nevertheless, studies have identified several molecules that alter the dynamics in ways that may alleviate SONFH. Blockade of DRP1 helps protect osteoblasts from oxidative stress by inhibiting excessive mitochondrial fission (67). Amyloid precursor protein, which is expressed in bone cells and many other cell types (68), stimulates mitochondrial fusion to promote osteoblast survival and bone formation in the presence of oxidative stress. At the same time, the protein may promote antioxidant responses to ROS and prevent cytochrome C release, helping prevent osteoblast apoptosis (69).
Mitochondrial ROS metabolism
Mitochondria are at once the centers of ROS production in cells and the organelles most sensitive to ROS (21). There are two antioxidant systems according to the distribution of antioxidant enzymes in the cell, and the enzymes in mitochondria and cytoplasm can both significantly clear the excessive ROS in the cell. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a vital transcription factor that is dysregulated in various oxidative stress-related pathologies (70). It combats intracellular oxidative stress by activating multiple downstream genes, such as Heme Oxygenase-1 (HO-1), superoxide dismutase (SOD), glutathione (GSH), and catalase (CAT) (71, 72), by binding to the upstream antioxidant response element (ARE) sequence (73). The phosphatase and tensin homolog inhibitor VO-OHpic attenuates GC-associated endothelial progenitor cell dysfunction and osteonecrosis of the femoral head by activating Nrf2 signaling and inhibiting the mitochondrial apoptosis pathway (66). PARK7 promotes repair in early SONFH by enhancing resistance to stress-induced apoptosis in BMSCs via regulation of the Nrf2 signaling pathway (74). In addition, mitochondria contain a strong antioxidant system involving SOD2, Gpx1/4, Prx3, Trx2, and TrxR2 (75). SOD2, for example, can eliminate excess superoxide and protein oxidation in mitochondria during osteoblast differentiation. To promote this activity, the enzyme is deacetylated at lysine 68 by SIRT3 (35). Melatonin can target mitochondria to promote the activity of SIRT3 and SOD2 to reduce oxidative stress, which has been shown to promote osteoblast survival and increase bone mass (76, 77). These considerations illustrate the possibility of treating SONFH by increasing the antioxidant capacity of mitochondria or their host cells.
Figure 4 shows and depicts the relevant regulatory mechanisms of mitochondrial homeostasis in SONFH.
Conclusions
Accumulating evidence suggests that the dysfunction of mitochondrial homeostasis helps drive SONFH. The suppression of mitophagy and mitochondrial biogenesis, excessive fission, and disorder of mitochondrial ROS metabolism lead to the dysfunction of mitochondrial homeostasis.
Efforts to target the major components of that homeostasis-mitochondrial biogenesis, mitophagy, mitochondrial dynamics, and mitochondrial antioxidant capacity – may be effective against this debilitating condition. Future research should focus on regulating these processes to restore mitochondrial homeostasis. Potential therapeutic targets include enhancing mitophagy via PINK1/Parkin, promoting mitochondrial biogenesis through TFAM and PGC-1α, balancing mitochondrial dynamics by inhibiting DRP1, and reducing oxidative stress with antioxidants like melatonin and SIRT3 activators. Exploring natural compounds, such as those in traditional Chinese medicine, may also offer novel treatments. However, it should be noted that mitochondrial activity is ubiquitous across all tissues and involved in numerous physiological processes. Therefore, we expect new drug interventions targeting mitochondria to be able to selectively affect the femoral head to minimize systemic side effects. Continued investigation into these pathways and their clinical applications is essential for developing effective therapies for SONFH.
ICMJE Conflict of Interest Statement
The authors declare that the study was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Funding Statement
This work is supported in China by the National Natural Science Foundation (82074472) and Zhejiang Provincial Natural Science Foundation (LQ22H060003).
Acknowledgement
We would like to thank A Chapin Rodríguez PhD for English language editing.
References
- 1↑
Peters MJ, Symmons DP, McCarey D, Dijkmans BA, Nicola P, Kvien TK, McInnes IB, Haentzschel H, Gonzalez-Gay MA, Provan S, et al.EULAR evidence-based recommendations for cardiovascular risk management in patients with rheumatoid arthritis and other forms of inflammatory arthritis. Annals of the Rheumatic Diseases 2010 69 325–331. (https://doi.org/10.1136/ard.2009.113696)
- 2↑
Caplan A, Fett N, Rosenbach M, Werth VP, & Micheletti RG. Prevention and management of glucocorticoid-induced side effects: a comprehensive review: a review of glucocorticoid pharmacology and bone health. Journal of the American Academy of Dermatology 2017 76 1–9. (https://doi.org/10.1016/j.jaad.2016.01.062)
- 3↑
Powell C, Chang C, Naguwa SM, Cheema G, & Gershwin ME. Steroid induced osteonecrosis: an analysis of steroid dosing risk. Autoimmunity Reviews 2010 9 721–743. (https://doi.org/10.1016/j.autrev.2010.06.007)
- 4↑
Zhao DW, Yu M, Hu K, Wang W, Yang L, Wang BJ, Gao XH, Guo YM, Xu YQ, Wei YS, et al.Prevalence of nontraumatic osteonecrosis of the femoral head and its associated risk factors in the Chinese population: results from a nationally representative survey. Chinese Medical Journal 2015 128 2843–2850. (https://doi.org/10.4103/0366-6999.168017)
- 5↑
Johnson AJ, Mont MA, Tsao AK, & Jones LC. Treatment of femoral head osteonecrosis in the United States: 16-year analysis of the nationwide inpatient sample. Clinical Orthopaedics and Related Research 2014 472 617–623. (https://doi.org/10.1007/s11999-013-3220-3)
- 6↑
Chang C, Greenspan A, & Gershwin ME. The pathogenesis, diagnosis and clinical manifestations of steroid-induced osteonecrosis. Journal of Autoimmunity 2020 110 102460. (https://doi.org/10.1016/j.jaut.2020.102460)
- 7↑
Weinstein RS, Hogan EA, Borrelli MJ, Liachenko S, O’Brien CA, & Manolagas SC. The pathophysiological sequence of glucocorticoid-induced osteonecrosis of the femoral head in male mice. Endocrinology 2017 158 3817–3831. (https://doi.org/10.1210/en.2017-00662)
- 8↑
Wang C, Meng H, Wang Y, Zhao B, Zhao C, Sun W, Zhu Y, Han B, Yuan X, Liu R, et al.Analysis of early stage osteonecrosis of the human femoral head and the mechanism of femoral head collapse. International Journal of Biological Sciences 2018 14 156–164. (https://doi.org/10.7150/ijbs.18334)
- 9↑
Zhao D, Zhang F, Wang B, Liu B, Li L, Kim SY, Goodman SB, Hernigou P, Cui Q, Lineaweaver WC, et al.Guidelines for clinical diagnosis and treatment of osteonecrosis of the femoral head in adults (2019 version). Journal of Orthopaedic Translation 2020 21 100–110. (https://doi.org/10.1016/j.jot.2019.12.004)
- 10↑
Cheng CH, Chen LR, & Chen KH. Osteoporosis due to hormone imbalance: an overview of the effects of estrogen deficiency and glucocorticoid overuse on bone turnover. International Journal of Molecular Sciences 2022 23 1376. (https://doi.org/10.3390/ijms23031376)
- 11↑
Tsuchiya M, Ichiseki T, Ueda S, Ueda Y, Shimazaki M, Kaneuji A, & Kawahara N. Mitochondrial stress and redox failure in steroid-associated osteonecrosis. International Journal of Medical Sciences 2018 15 205–209. (https://doi.org/10.7150/ijms.22525)
- 12↑
Huang S, Wang Y, Gan X, Fang D, Zhong C, Wu L, Hu G, Sosunov AA, McKhann GM, Yu H, et al.Drp1-mediated mitochondrial abnormalities link to synaptic injury in diabetes model. Diabetes 2015 64 1728–1742. (https://doi.org/10.2337/db14-0758)
- 13↑
Chan DC. Mitochondrial dynamics and its involvement in disease. Annual Review of Pathology 2020 15 235–259. (https://doi.org/10.1146/annurev-pathmechdis-012419-032711)
- 14↑
Shen Y, Wu L, Qin D, Xia Y, Zhou Z, Zhang X, & Wu X. Carbon black suppresses the osteogenesis of mesenchymal stem cells: the role of mitochondria. Particle and Fibre Toxicology 2018 15 16. (https://doi.org/10.1186/s12989-018-0253-5)
- 15↑
Lee SY, An HJ, Kim JM, Sung MJ, Kim DK, Kim HK, Oh J, Jeong HY, Lee YH, Yang T, et al.PINK1 deficiency impairs osteoblast differentiation through aberrant mitochondrial homeostasis. Stem Cell Research and Therapy 2021 12 589. (https://doi.org/10.1186/s13287-021-02656-4)
- 16↑
Lin H, Wei B, Li G, Zheng J, Sun J, Chu J, Zeng R, & Niu Y. Sulforaphane reverses glucocorticoid-induced apoptosis in osteoblastic cells through regulation of the Nrf2 pathway. Drug Design, Development and Therapy 2014 8 973–982. (https://doi.org/10.2147/DDDT.S65410)
- 17↑
Zhan J, Yan Z, Zhao M, Qi W, Lin J, Lin Z, Huang Y, Pan X, & Xue X. Allicin inhibits osteoblast apoptosis and steroid-induced necrosis of femoral head progression by activating the PI3K/AKT pathway. Food and Function 2020 11 7830–7841. (https://doi.org/10.1039/d0fo00837k)
- 18↑
Baethge C, Goldbeck-Wood S, & Mertens S. SANRA-a scale for the quality assessment of narrative review articles. Research Integrity and Peer Review 2019 4 5. (https://doi.org/10.1186/s41073-019-0064-8)
- 19↑
Spinelli JB, & Haigis MC. The multifaceted contributions of mitochondria to cellular metabolism. Nature Cell Biology 2018 20 745–754. (https://doi.org/10.1038/s41556-018-0124-1)
- 20↑
Westermann B. Mitochondrial fusion and fission in cell life and death. Nature Reviews. Molecular Cell Biology 2010 11 872–884. (https://doi.org/10.1038/nrm3013)
- 21↑
Alam TI, Kanki T, Muta T, Ukaji K, Abe Y, Nakayama H, Takio K, Hamasaki N, & Kang D. Human mitochondrial DNA is packaged with TFAM. Nucleic Acids Research 2003 31 1640–1645. (https://doi.org/10.1093/nar/gkg251)
- 22↑
Rossmann MP, Dubois SM, Agarwal S, & Zon LI. Mitochondrial function in development and disease. Disease Models and Mechanisms 2021 14 dmm048912. (https://doi.org/10.1242/dmm.048912)
- 23↑
Willems PHM, Rossignol R, Dieteren CEJ, Murphy MP, & Koopman WJH. Redox homeostasis and mitochondrial dynamics. Cell Metabolism 2015 22 207–218. (https://doi.org/10.1016/j.cmet.2015.06.006)
- 24↑
Ma K, Chen G, Li W, Kepp O, Zhu Y, & Chen Q. Mitophagy, mitochondrial homeostasis, and cell fate. Frontiers in Cell and Developmental Biology 2020 8 467. (https://doi.org/10.3389/fcell.2020.00467)
- 25↑
Ng MYW, Wai T, & Simonsen A. Quality control of the mitochondrion. Developmental Cell 2021 56 881–905. (https://doi.org/10.1016/j.devcel.2021.02.009)
- 26↑
Luo H, Wei J, Wu S, Zheng Q, Lin X, & Chen P. Elucidating the role of the GC/GR/GLUT1 axis in steroid-induced osteonecrosis of the femoral head: a proteomic approach. Bone 2024 183 117074. (https://doi.org/10.1016/j.bone.2024.117074)
- 27↑
Ma Z, Sun J, Jiang Q, Zhao Y, Jiang H, Sun P, & Feng W. Identification and analysis of mitochondria-related central genes in steroid-induced osteonecrosis of the femoral head, along with drug prediction. Frontiers in Endocrinology 2024 15 1341366. (https://doi.org/10.3389/fendo.2024.1341366)
- 28↑
Xue XH, Feng ZH, Li ZX, & Pan XY. Salidroside inhibits steroid-induced avascular necrosis of the femoral head via the PI3K/Akt signaling pathway: in vitro and in vivo studies. Molecular Medicine Reports 2018 17 3751–3757. (https://doi.org/10.3892/mmr.2017.8349)
- 29↑
Yan Z, Zhan J, Qi W, Lin J, Huang Y, Xue X, & Pan X. The protective effect of luteolin in glucocorticoid-induced osteonecrosis of the femoral head. Frontiers in Pharmacology 2020 11 1195. (https://doi.org/10.3389/fphar.2020.01195)
- 30↑
Chen K, Liu Y, He J, Pavlos N, Wang C, Kenny J, Yuan J, Zhang Q, Xu J, & He W. Steroid-induced osteonecrosis of the femoral head reveals enhanced reactive oxygen species and hyperactive osteoclasts. International Journal of Biological Sciences 2020 16 1888–1900. (https://doi.org/10.7150/ijbs.40917)
- 31↑
Shimasaki M, Ueda S, Ichiseki T, Hirata H, Kawahara N, & Ueda Y. Resistance of bone marrow mesenchymal stem cells in a stressed environment - comparison with osteocyte cells. International Journal of Medical Sciences 2021 18 1375–1381. (https://doi.org/10.7150/ijms.52104)
- 32↑
Xu J, Gong H, Lu S, Deasey MJ, & Cui Q. Animal models of steroid-induced osteonecrosis of the femoral head-a comprehensive research review up to 2018. International Orthopaedics 2018 42 1729–1737. (https://doi.org/10.1007/s00264-018-3956-1)
- 33↑
Zhang F, Peng W, Zhang J, Dong W, Wu J, Wang T, & Xie Z. P53 and Parkin co-regulate mitophagy in bone marrow mesenchymal stem cells to promote the repair of early steroid-induced osteonecrosis of the femoral head. Cell Death and Disease 2020 11 42. (https://doi.org/10.1038/s41419-020-2238-1)
- 34↑
Jing X, Du T, Yang X, Zhang W, Wang G, Liu X, Li T, & Jiang Z. Desferoxamine protects against glucocorticoid-induced osteonecrosis of the femoral head via activating HIF-1alpha expression. Journal of Cellular Physiology 2020 235 9864–9875. (https://doi.org/10.1002/jcp.29799)
- 35↑
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 and Differentiation 2018 25 229–240. (https://doi.org/10.1038/cdd.2017.144)
- 36↑
Yu H, Liu P, Zuo W, Sun X, Liu H, Lu F, Guo W, & Zhang Q. Decreased angiogenic and increased apoptotic activities of bone microvascular endothelial cells in patients with glucocorticoid-induced osteonecrosis of the femoral head. BMC Musculoskeletal Disorders 2020 21 277. (https://doi.org/10.1186/s12891-020-03225-1)
- 37↑
Boss JH, & Misselevich I. Osteonecrosis of the femoral head of laboratory animals: the lessons learned from a comparative study of osteonecrosis in man and experimental animals. Veterinary Pathology 2003 40 345–354. (https://doi.org/10.1354/vp.40-4-345)
- 38↑
Zhen YF, Wang GD, Zhu LQ, Tan SP, Zhang FY, Zhou XZ, & Wang XD. P53 dependent mitochondrial permeability transition pore opening is required for dexamethasone-induced death of osteoblasts. Journal of Cellular Physiology 2014 229 1475–1483. (https://doi.org/10.1002/jcp.24589)
- 39↑
Sun F, Zhou JL, Liu ZL, Jiang ZW, & Peng H. Dexamethasone induces ferroptosis via P53/SLC7A11/GPX4 pathway in glucocorticoid-induced osteonecrosis of the femoral head. Biochemical and Biophysical Research Communications 2022 602 149–155. (https://doi.org/10.1016/j.bbrc.2022.02.112)
- 40↑
Li W, Li W, Zhang W, Wang H, Yu L, Yang P, Qin Y, Gan M, Yang X, Huang L, et al.Exogenous melatonin ameliorates steroid-induced osteonecrosis of the femoral head by modulating ferroptosis through GDF15-mediated signaling. Stem Cell Research and Therapy 2023 14 171. (https://doi.org/10.1186/s13287-023-03371-y)
- 41↑
Almeida M, Han L, Ambrogini E, Weinstein RS, & Manolagas SC. Glucocorticoids and tumor necrosis factor α increase oxidative stress and suppress Wnt protein signaling in osteoblasts. Journal of Biological Chemistry 2011 286 44326–44335. (https://doi.org/10.1074/jbc.M111.283481)
- 42↑
Tan G, Kang PD, & Pei FX. Glucocorticoids affect the metabolism of bone marrow stromal cells and lead to osteonecrosis of the femoral head: a review. Chinese Medical Journal 2012 125 134–139.
- 43↑
Xia C, Xu H, Fang L, Chen J, Yuan W, Fu D, Wang X, He B, Xiao L, Wu C, et al.β-catenin inhibition disrupts the homeostasis of osteogenic/adipogenic differentiation leading to the development of glucocorticoid-induced osteonecrosis of the femoral head. eLife 2024 12 RP92469. (https://doi.org/10.7554/eLife.92469)
- 44↑
Palikaras K, Lionaki E, & Tavernarakis N. Mechanisms of mitophagy in cellular homeostasis, physiology and pathology. Nature Cell Biology 2018 20 1013–1022. (https://doi.org/10.1038/s41556-018-0176-2)
- 45↑
Pickles S, Vigie P, & Youle RJ. Mitophagy and quality control mechanisms in mitochondrial maintenance. Current Biology 2018 28 R170–R185. (https://doi.org/10.1016/j.cub.2018.01.004)
- 46↑
Wang DB, Kinoshita C, Kinoshita Y, & Morrison RS. p53 and mitochondrial function in neurons. Biochimica et Biophysica Acta 2014 1842 1186–1197. (https://doi.org/10.1016/j.bbadis.2013.12.015)
- 47↑
Fan P, Yu XY, Xie XH, Chen CH, Zhang P, Yang C, Peng X, & Wang YT. Mitophagy is a protective response against oxidative damage in bone marrow mesenchymal stem cells. Life Sciences 2019 229 36–45. (https://doi.org/10.1016/j.lfs.2019.05.027)
- 48↑
Rüb C, Wilkening A, & Voos W. Mitochondrial quality control by the Pink1/Parkin system. Cell and Tissue Research 2017 367 111–123. (https://doi.org/10.1007/s00441-016-2485-8)
- 49↑
Vincow ES, Merrihew G, Thomas RE, Shulman NJ, Beyer RP, MacCoss MJ, & Pallanck LJ. The PINK1-Parkin pathway promotes both mitophagy and selective respiratory chain turnover in vivo. PNAS 2013 110 6400–6405. (https://doi.org/10.1073/pnas.1221132110)
- 50↑
Ge P, Dawson VL, & Dawson TM. PINK1 and Parkin mitochondrial quality control: a source of regional vulnerability in Parkinson’s disease. Molecular Neurodegeneration 2020 15 20. (https://doi.org/10.1186/s13024-020-00367-7)
- 51↑
Jung YY, Son DJ, Lee HL, Kim DH, Song MJ, Ham YW, Kim Y, Han SB, Park MH, & Hong JT. Loss of Parkin reduces inflammatory arthritis by inhibiting p53 degradation. Redox Biology 2017 12 666–673. (https://doi.org/10.1016/j.redox.2017.04.007)
- 52↑
Xu K, Lu C, Ren X, Wang J, Xu P, & Zhang Y. Overexpression of HIF-1alpha enhances the protective effect of mitophagy on steroid-induced osteocytes apoptosis. Environmental Toxicology 2021 36 2123–2137. (https://doi.org/10.1002/tox.23327)
- 53↑
Chen L, Shi X, Weng SJ, Xie J, Tang JH, Yan DY, Wang BZ, Xie ZJ, Wu ZY, & Yang L. Vitamin K2 can rescue the dexamethasone-induced downregulation of osteoblast autophagy and mitophagy thereby restoring osteoblast function in vitro and in vivo. Frontiers in Pharmacology 2020 11 1209. (https://doi.org/10.3389/fphar.2020.01209)
- 54↑
Li Q, Gao Z, Chen Y, & Guan MX. The role of mitochondria in osteogenic, adipogenic and chondrogenic differentiation of mesenchymal stem cells. Protein and Cell 2017 8 439–445. (https://doi.org/10.1007/s13238-017-0385-7)
- 55↑
Hsu CN, Jen CY, Chen YH, Peng SY, Wu SC, & Yao CL. Glucocorticoid transiently upregulates mitochondrial biogenesis in the osteoblast. Chinese Journal of Physiology 2020 63 286–293. (https://doi.org/10.4103/CJP.CJP_51_20)
- 56↑
Wiedemann N, & Pfanner N. Mitochondrial machineries for protein import and assembly. Annual Review of Biochemistry 2017 86 685–714. (https://doi.org/10.1146/annurev-biochem-060815-014352)
- 57↑
Ueda S, Shimasaki M, Ichiseki T, Hirata H, Kawahara N, & Ueda Y. Mitochondrial transcription factor A added to osteocytes in a stressed environment has a cytoprotective Effect. International Journal of Medical Sciences 2020 17 1293–1299. (https://doi.org/10.7150/ijms.45335)
- 58↑
Hirata H, Ueda S, Ichiseki T, Shimasaki M, Ueda Y, Kaneuji A, & Kawahara N. Taurine inhibits glucocorticoid-induced bone mitochondrial injury, preventing osteonecrosis in rabbits and cultured osteocytes. International Journal of Molecular Sciences 2020 21 6892. (https://doi.org/10.3390/ijms21186892)
- 59↑
Popov LD. Mitochondrial biogenesis: an update. Journal of Cellular and Molecular Medicine 2020 24 4892–4899. (https://doi.org/10.1111/jcmm.15194)
- 60↑
Pal S, Maurya SK, Chattopadhyay S, Pal China S, Porwal K, Kulkarni C, Sanyal S, Sinha RA, & Chattopadhyay N. The osteogenic effect of liraglutide involves enhanced mitochondrial biogenesis in osteoblasts. Biochemical Pharmacology 2019 164 34–44. (https://doi.org/10.1016/j.bcp.2019.03.024)
- 61↑
Wang L, Li Q, Yan H, Jiao G, Wang H, Chi H, Zhou H, Chen L, Shan Y, & Chen Y. Resveratrol protects osteoblasts against dexamethasone-induced cytotoxicity through activation of AMP-activated protein kinase. Drug Design, Development and Therapy 2020 14 4451–4463. (https://doi.org/10.2147/DDDT.S266502)
- 62↑
Martin-Aragon S, Bermejo-Bescos P, Benedi J, Raposo C, Marques F, Kydonaki EK, Gkiata P, Koutedakis Y, Ntina G, Carrillo AE, et al.A neuroprotective bovine colostrum attenuates apoptosis in dexamethasone-treated MC3T3-E1 osteoblastic cells. International Journal of Molecular Sciences 2021 22 10195. (https://doi.org/10.3390/ijms221910195)
- 63↑
Kim S, & Sieburth D. Sphingosine kinase activates the mitochondrial unfolded protein response and is targeted to mitochondria by stress. Cell Reports 2018 24 2932–2945.e4. (https://doi.org/10.1016/j.celrep.2018.08.037)
- 64↑
Chen H, McCaffery JM, & Chan DC. Mitochondrial fusion protects against neurodegeneration in the cerebellum. Cell 2007 130 548–562. (https://doi.org/10.1016/j.cell.2007.06.026)
- 65↑
Archer SL. Mitochondrial dynamics--mitochondrial fission and fusion in human diseases. New England Journal of Medicine 2013 369 2236–2251. (https://doi.org/10.1056/NEJMra1215233)
- 66↑
Yao X, Yu S, Jing X, Guo J, Sun K, Guo F, & Ye Y. PTEN inhibitor VO-OHpic attenuates GC-associated endothelial progenitor cell dysfunction and osteonecrosis of the femoral head via activating Nrf2 signaling and inhibiting mitochondrial apoptosis pathway. Stem Cell Research and Therapy 2020 11 140. (https://doi.org/10.1186/s13287-020-01658-y)
- 67↑
Gan X, Huang S, Yu Q, Yu H, & Yan SS. Blockade of Drp1 rescues oxidative stress-induced osteoblast dysfunction. Biochemical and Biophysical Research Communications 2015 468 719–725. (https://doi.org/10.1016/j.bbrc.2015.11.022)
- 68↑
Xia WF, Jung JU, Shun C, Xiong S, Xiong L, Shi XM, Mei L, & Xiong WC. Swedish mutant APP suppresses osteoblast differentiation and causes osteoporotic deficit, which are ameliorated by N-acetyl-L-cysteine. Journal of Bone and Mineral Research 2013 28 2122–2135. (https://doi.org/10.1002/jbmr.1954)
- 69↑
Pan JX, Tang F, Xiong F, Xiong L, Zeng P, Wang B, Zhao K, Guo H, Shun C, Xia WF, et al.APP promotes osteoblast survival and bone formation by regulating mitochondrial function and preventing oxidative stress. Cell Death and Disease 2018 9 1077. (https://doi.org/10.1038/s41419-018-1123-7)
- 70↑
Kumar H, Kim IS, More SV, Kim BW, & Choi DK. Natural product-derived pharmacological modulators of Nrf2/ARE pathway for chronic diseases. Natural Product Reports 2014 31 109–139. (https://doi.org/10.1039/c3np70065h)
- 71↑
Kensler TW, Wakabayashi N, & Biswal S. Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annual Review of Pharmacology and Toxicology 2007 47 89–116. (https://doi.org/10.1146/annurev.pharmtox.46.120604.141046)
- 72↑
Malhotra D, Portales-Casamar E, Singh A, Srivastava S, Arenillas D, Happel C, Shyr C, Wakabayashi N, Kensler TW, Wasserman WW, et al.Global mapping of binding sites for Nrf2 identifies novel targets in cell survival response through ChIP-Seq profiling and network analysis. Nucleic Acids Research 2010 38 5718–5734. (https://doi.org/10.1093/nar/gkq212)
- 73↑
Shi X, & Zhou B. The role of Nrf2 and MAPK pathways in PFOS-induced oxidative stress in zebrafish embryos. Toxicological Sciences 2010 115 391–400. (https://doi.org/10.1093/toxsci/kfq066)
- 74↑
Zhang F, Yan Y, Peng W, Wang L, Wang T, Xie Z, Luo H, Zhang J, & Dong W. PARK7 promotes repair in early steroid-induced osteonecrosis of the femoral head by enhancing resistance to stress-induced apoptosis in bone marrow mesenchymal stem cells via regulation of the Nrf2 signaling pathway. Cell Death and Disease 2021 12 940. (https://doi.org/10.1038/s41419-021-04226-1)
- 75↑
Apostolova N, & Victor VM. Molecular strategies for targeting antioxidants to mitochondria: therapeutic implications. Antioxidants and Redox Signaling 2015 22 686–729. (https://doi.org/10.1089/ars.2014.5952)
- 76↑
Reiter RJ, Mayo JC, Tan DX, Sainz RM, Alatorre-Jimenez M, & Qin L. Melatonin as an antioxidant: under promises but over delivers. Journal of Pineal Research 2016 61 253–278. (https://doi.org/10.1111/jpi.12360)
- 77↑
Zhou W, Liu Y, Shen J, Yu B, Bai J, Lin J, Guo X, Sun H, Chen Z, Yang H, et al.Melatonin increases bone mass around the prostheses of OVX rats by ameliorating mitochondrial oxidative stress via the SIRT3/SOD2 signaling pathway. Oxidative Medicine and Cellular Longevity 2019 2019 4019619. (https://doi.org/10.1155/2019/4019619)