Abstract
Introduction
Bone-degenerative conditions, including osteoporosis, rheumatoid arthritis, and osteoarthritis, are major public health concerns worldwide, associated with oxidative stress and iron overload that disrupts bone homeostasis. Ferroptosis, an iron-mediated form of cell death, has emerged as a critical factor in bone degeneration, necessitating a comprehensive review of its role in these conditions.
Content
This review comprehensively examined the latest research on oxidative stress, iron metabolism, and ferroptosis related to bone biology and degeneration, focusing on their interconnections and potential therapeutic implications. The review revealed that oxidative stress affects various bone cell types, including osteoclasts, osteoblasts, and chondrocytes, contributing to bone loss and cartilage degradation. Iron homeostasis was found to be crucial for bone cell function, with both iron overload and deficiency potentially leading to pathological conditions. Ferroptosis regulation involves a complex interplay between iron metabolism, lipid peroxidation, and antioxidant systems, including the SLC7A11-GSH-GPX4 network and the FSP1-CoQ10H2 pathway. Different bone cell lineages, including mesenchymal stem cells, osteoblasts, osteoclasts, and chondrocytes, exhibit varied responses to ferroptosis induction and regulation.
Summary
Understanding the molecular mechanisms underlying ferroptosis regulation in bone cells offers promising avenues for developing targeted therapies for bone-degenerative conditions.
Outlook
Future research should focus on elucidating the specific roles of ferroptosis in different bone disorders and exploring potential therapeutic interventions targeting oxidative stress, iron overload, and ferroptosis pathways to improve the management of these debilitating conditions.
Introduction
Bone-related degenerative conditions, encompassing diseases such as osteoporosis, rheumatoid arthritis, osteoarthritis, diabetic bone deterioration, and lumbar disc herniation, have emerged as significant public health concerns worldwide [1], [2], [3]. Osteoporosis, featured by reduced bone mass and bone tissue degeneration, affects over 200 million people globally and accounts for substantial healthcare expenditures [4]. Rheumatoid arthritis, an autoimmune disorder that primarily impairs synovial joints, affects approximately 0.5–1% of the population worldwide and can cause joint tissue disruption and disability [5]. The incidence of these age-related conditions is expected to rise due to an aging population [6]. Although pharmacological interventions and surgical procedures are available, managing these debilitating conditions remains challenging, necessitating the formulation of more effective therapeutic approaches.
Oxidative stress and iron overload are emerging driving factors in the progression of bone-related disorders. Cellular damage and tissue degeneration can result from oxidative stress, which arises when there is an imbalance between the generation of reactive oxygen species (ROS) and the body’s antioxidant defense systems [7]. In osteoporosis, increased oxidative levels have been associated with the dysregulation of osteoblast and osteoclast activities, leading to bone loss [8]. Similarly, in rheumatoid arthritis, heightened ROS generation can activate inflammatory pathways, exacerbating joint destruction [9]. Furthermore, iron accumulation has been observed in various bone disorders, and excess iron can catalyze the production of aggressively active hydroxyl radicals, potentiating ROS stress [10]. For instance, in osteoarthritis, iron deposition in articular cartilage has been associated with chondrocyte death and cartilage degradation [11]. This knowledge regarding the roles of oxidative burden and iron dysregulation in bone-related diseases offer promising avenues for developing targeted therapeutic strategies.
Ferroptosis, a novel modality of iron-mediated cell death, has been identified as a critical player in bone degeneration [12]. This unique mode of cell demise is instigated by an imbalance in the production and neutralization of lipid peroxides, which eventually causes membrane rupture [13]. Ferroptosis has been linked to the maintenance of bone cell balance. For instance, iron overload can promote osteoclast differentiation and suppress osteoblast function, thereby triggering excessive bone resorption [14], 15]. The induction of ferroptosis in osteoblasts has been well-documented in bone-degenerative disorders, such as osteoporosis [16]. Elevated levels of lipid peroxidation and iron accumulation were found in osteoporotic bone, indicating a potential role for ferroptosis in driving pathological bone loss. Given the increasing importance of ferroptosis in bone health and illness, targeting ferroptotic processes has garnered intense interest in the research field, holding promise for alleviating the progression of bone-degenerative conditions [17].
In the current review, we discuss recent understanding of the interconnections between redox imbalance, iron homeostasis, and ferroptosis in terms of bone biology and degeneration. We highlight the critical roles of these processes in regulating bone cell homeostasis and function, and their dysregulation in various bone-degenerative conditions. By elucidating the molecular mechanisms underlying ferroptosis regulation in different bone cell lineages, we aim to shed light on the pathogenesis of debilitating disorders like osteoporosis, rheumatoid arthritis, and osteoarthritis. Ultimately, this insight could eventually lead to the creation of new therapeutic approaches that target ferroptosis pathways, offering promising prospects for preventing and managing bone-degenerative conditions.
Oxidative stress and iron metabolism in bone homeostasis
Oxidative stress and bone homeostasis
Oxidative stress has become an important modulator of bone homeostasis, exerting profound effects on various bone cell types and their functions [18]. In osteoclasts, the bone-resorbing cells, elevated ROS levels were found to promote their differentiation and functional activity, contributing to accelerated bone loss [19], 20]. Conversely, in osteoblasts, the bone-forming cells, oxidative stress can impair their differentiation, mineralization capacity, and survival, thereby compromising bone formation [21]. Additionally, chondrocytes, the cellular components of cartilage, are vulnerable to oxidative injury, which can lead to cartilage degradation in osteoarthritis [22]. Notably, oxidative stress also impinges on cell fate determination of mesenchymal stem cells, hindering their osteogenic differentiation potential while favoring adipogenic lineage commitment, ultimately disrupting bone homeostasis [23]. These findings highlight the intricate interaction between oxidative burden and various bone lineages, underscoring the potential benefits of mitigating oxidative stress in the management of bone-degenerative conditions.
Iron homeostasis in bone and cartilage lineages
Iron metabolism is meticulously regulated in different cell types within the bone and cartilage to maintain proper cell differentiation and functional homeostasis. In osteoclasts, which are responsible for bone resorption, iron uptake is critically important for their differentiation and function [24], 25]. During osteoclastogenesis, the expression of key genes facilitating iron import, such as transferrin receptor 1 (TFR1) and the non-transferrin-bound iron (NTBI) transporter solute carrier family 39 member 14 (SLC39A14), is significantly upregulated [26]. This increased iron acquisition is essential for supporting the high energy demands of osteoclasts, as iron is a crucial cofactor for mitochondrial respiration and biogenesis [27]. Notably, TFR1 depletion in osteoclasts leads to a substantial decrease in labile iron levels and attenuated bone resorption activity [28]. Conversely, iron chelation by agents like deferoxamine (DFO) dose-dependently inhibits osteoclast development and activity [29], 30]. Osteoblasts, on the other hand, exhibit a more complex relationship with iron. While moderate levels of iron are necessary for osteoblast function, excessive iron can impair their differentiation, mineralization, and survival [31]. Iron overload conditions lead to the upregulation of iron-importing factors like divalent metal transporter 1 (DMT1) and TFR1 in osteoblasts, accompanied by increased oxidative stress and potential ferroptosis induction [32], 33]. Furthermore, iron overload suppresses osteogenic master regulators such as runt-related transcription factor 2 (Runx2) and its target genes, including osteocalcin and alkaline phosphatase (ALP), hampering osteoblast differentiation and matrix mineralization [34].
In mesenchymal stem cells (MSCs), the precursors of osteoblasts and chondrocytes, iron dysregulation can break the balanced differentiation between osteogenesis and adipogenesis. Iron overload impairs the osteogenic commitment of MSCs [35], while iron chelation can rescue this anti-osteogenic effect [36]. Chondrocytes, the key cellular component of cartilage, are also susceptible to iron-induced damage. Excessive iron induces catabolic markers like matrix metalloproteinases (MMPs) in chondrocytes, leading to extracellular matrix degradation and cartilage degeneration [37]. Iron chelators and antioxidants can alleviate these detrimental effects, underscoring the involvement of iron-dependent oxidative burden in cartilage deterioration [38], 39]. In summary, iron metabolism can functionally impinge on different cell types within the bone and cartilage microenvironment. Maintaining an optimal balance of iron levels is critical for preserving bone homeostasis, as both iron overload and deficiency can contribute to pathological conditions like osteoporosis and cartilage degeneration.
Regulation of ferroptosis: intricate interplay of diverse metabolic and signaling pathways
Iron regulation and ferroptosis induction
Iron is a mediator of oxidative stress and indispensable for ferroptosis initiation. Iron ions within cells mainly originate from heme group or non-heme iron found in dietary food. Non-heme iron can be present as Fe2+ or Fe3+. The enzymatic action of duodenal cytochrome B (DCYTB) in the intestine converts Fe3+ to Fe2+, facilitating its uptake through epithelial cells via DMT1 [40]. Ferritin iron and heme iron can also be internalized by intestinal epithelial cells via ferritin receptors and the cluster of differentiation 91 (CD91), respectively [41]. Excess Fe2+ can bind to ferritin within intestinal epithelial cells for storage [42]. In times of need, Fe2+ can be extruded from intestinal epithelial cells through ferroportin 1 (FPN1) and undergo oxidation to Fe3+ by hephaestin (HEPH) for onward transportation [43], 44]. Hepatic transferrin (TF) is loaded with Fe3+, with each TF molecule capable of binding two Fe3+ ions [45]. Fe3+-TF undergoes translocation to target cells through the interaction with transferrin receptor 1 (TFR1), ushering Fe3+ to endosomal compartments [46]. Within the acidic milieu of endosomes, Fe3+ is converted to Fe2+ by metal-containing reductases like six-transmembrane epithelial antigen of prostate 3 (STEAP3) before being delivered to the cellular cytoplasm by DMT1 [47].
Intracellular regulation of Fe2+ is tightly controlled to maintain homeostasis. Cellular insufficiency of Fe2+ can hinder various biological processes, while an abundance of Fe2+ triggers the Fenton reaction. This reaction involves the excess Fe2+ reacting with hydrogen peroxide [48], resulting in ROS production. The accumulation of iron and ROS can trigger ferroptosis [49] (Figure 1) Furthermore, an excess of Fe2+ can stimulate lipid peroxidation through participation in the enzymatic subunit of lipoxygenase (LOX) [50], 51]. Additionally, surplus intracellular Fe2+ can be converted to Fe3+ by ferritin and stored in the structure of ferritin heavy chain 1 (FTH1) or ferritin light chain (FTL) [52]. Ferritin protein may undergo degradation by nuclear receptor coactivator 4 (NCOA4), releasing labile iron [53]. Cells can also eliminate excessive Fe2+ through the action of FPN1. FPN1, along with its governing factor hepcidin, has a pivotal role in iron metabolism. FPN1 has currently been recognized as the sole iron export avenue [54]. Irregular expression or malfunction of these proteins can result in heightened levels of intracellular labile iron.

Current understanding of ferroptosis induction and inhibition. Ferroptosis induction mechanisms (left panel) and the inhibition mechanisms (right panel) are summarized. Iron metabolism: Extracellular Fe3+ binds to TF, forming the Fe3+-TF1 complex, which interacts with TFR1 for Fe3+ entry into the cytosol. Fe3+ is converted to Fe2+, catalyzing hydroxyl radicals (·OH) production through the Fenton reaction. Both ·OH and Fe2+-containing LOX enzyme induce lipid peroxidation. Mitochondria: the central organelle in iron homeostasis, the mitochondrion, requires iron for heme groups and sulfur-iron clusters essential for mitochondrial ETC components. In cancer cells, increased iron levels enhance electron transport and ROS production in mitochondria. Lipid metabolism: PUFAs are converted to PUFA-CoA by ACSL4 and then to PUFA-PL by LPCAT. LOXs or ·OH oxidize PUFA-PL to produce PLOOH. GPX4: a selenoprotein, GPX4, utilizes GSH to reduce PLOOH and prevent ferroptosis. GSH metabolism: the antiporter system Xc− exchanges cystine for glutamate, converting cystine to cysteine for GSH biosynthesis. Blocking system Xc− leads to GSH depletion, promoting ferroptosis. Mevalonate pathway: IPP, synthesized from acetyl-CoA, activates GPX4 with a selenocysteine residue. FSP1-dependent pathway: FSP1 converts CoQ10 to ubiquinol to counteract PLOOH. DHODH-dependent pathway: DHODH reduces CoQ10 to ubiquinol-10, suppressing mitochondrial lipid peroxidation and ferroptosis. GCH1-dependent pathway: GCH-1 generates BH4 from BH2 to counteract PLOOH and inhibit ferroptosis. ACSL4, acyl-CoA synthetase long-chain family member 4; BH2, dihydrobiopterin; BH4, tetrahydrobiopterin; CoQ10, coenzyme Q10 (also known as ubiquinone); DHODH, dihydroorotate dehydrogenase; ETC, electron transport chain; FSP1, ferroptosis suppression protein 1; GCH1, GTP cyclohydrolase 1; GPX4, glutathione peroxidase 4; IPP, isopentenyl pyrophosphate; LPCAT3, lysophosphatidylcholine acyltransferase 3; LOX, lipoxygenase; NOX, NADPH oxidase; PLOOH, phospholipid hydroperoxides; PUFA, polyunsaturated fatty acid; PUFA-PL, polyunsaturated phospholipid; RTA, radical-trapping antioxidant; TF, transferrin; TFRC, transferrin receptor.
Another important process in ferroptosis induction is iron-dependent lipid peroxidation. Phospholipids (PLs) are essential constituents of plasma membranes, with the ability to form diverse fatty acyl chains [55]. Polyunsaturated fatty acyl (PUFA) can form PUFA-PLs by attaching to the sn2 position of PL with the assistance of acyl-CoA synthetase long-chain family member 4 (ACSL4) and Lys phosphatidylcholine acyltransferase 3 (LPCAT3) [56]. Deletion of these enzymes could suppress the generation of PUFA-PLs [57], 58]. PUFA-PLs are significant for maintaining cell membrane fluidity and functional flexibility [59]. However, PUFAs with bis-allylic hydrogen atoms are highly susceptible to oxidation by potent oxidants. This reaction produces phospholipid radicals (PL·) which are further transformed into phospholipid peroxyl radicals (PLOO·) [60]. It is noteworthy that PLOO· could remove hydrogen atoms from the bis-allylic positions of a different PUFA-PL, leading to the production of PUFA-PL-OOH [61]. The continual synthesis and buildup of PUFA-PL-OOH, influenced by intracellular iron and ROS levels, undermines the plasma membrane integrity to induce cell death [62]. Additionally, lipoxygenases (LOXs) can promote the oxidation of PUFA-PLs and support ferroptosis [63].
Negative regulators of ferroptosis
Cells, under normal conditions, can counteract lipid peroxidation efficiently to avoid excessive accumulation. One crucial enzyme responsible for detoxifying PL-OOH is Glutathione peroxidase 4 (GPX4) from the GPXs family (Figure 1). GPX4 can transform PL-OOH into harmless phospholipid alcohol (PL-OH) through the reducing power of glutathione (GSH) [64]. GSH acts as a vital reducing cofactor to facilitate lipid peroxidation detoxification mediated by GPX4 [65]. The activity of GPX4 is impaired when there is a decrease in GSH synthesis. GSH is composed of glutamate, glycine, and cysteine, with cysteine being a key intermediate amino acid that must be obtained extracellularly for GSH synthesis [66]. Cellular import of cysteine relies on the cystine/glutamate transporter (system Xc−), consisting of the light chain subunit solute carrier family 7 member 11 (SLC7A11) and the heavy chain subunit solute carrier family 3 member 2 (SLC3A2). The exchange of intracellular glutamate with extracellular cystine is facilitated by SLC7A11 [67]. Intracellular cystine is further converted to cysteine to contribute to GSH production [68]. Thus, the primary defense mechanism against ferroptosis involves GSH biosynthesis and the GPX4-mediated detoxification of GSH-dependent PL-OOH, forming the SLC7A11-GSH-GPX4 network. Disruption of this network, such as through inhibition of SLC7A11-GSH-GPX4, can trigger ferroptosis. Notably, Dixon et al. demonstrated that inhibitors like Ras-selective lethal small molecule 3 (RSL3) and Erastin induce ferroptosis by blocking GPX4 and System Xc-, resulting in cellular PL-OOH overload [69].
Additional pathways have been identified to combat ferroptosis. One such mechanism involves ubiquinone (UQ, also known as coenzyme Q10, or CoQ10), a lipophilic molecule that is prevalent across various cell types. Coenzyme Q10 (CoQ10), primarily produced in mitochondria, plays a crucial role in the electron transport chain due to its redox properties [70]. CoQ10 exists in various oxidation states, with its fully reduced form ubiquinol (CoQ10H2) acting as a potent antioxidant that combats lipophilic free radicals and suppresses ferroptosis. The enzyme ferroptosis suppressor protein 1 (FSP1) catalyzes the reduction of CoQ10 to CoQ10H2, utilizing NAD(P)H in the process [71]. FSP1’s N-terminal myristoylation motif is crucial for its plasma membrane localization and function, with modifications to this region potentially impairing its activity [72]. The FSP1-CoQ10H2 system functions as an alternative ferroptosis defense mechanism, complementing the SLC7A11-GSH-GPX4 pathway by continuously generating CoQ10H2 and eliminating lipid free radicals. Studies by Bersuker et al. demonstrated FSP1’s synergistic action with GPX4 in ferroptosis prevention. While FSP1 knockout did not affect GSH levels, it led to increased lipid peroxidation. Moreover, GPX4-deficient H460 cells remained viable with functional FSP1, but simultaneous knockout of both GPX4 and FSP1 resulted in pronounced ferroptosis [73]. Further research by Doll et al. showed that FSP1 overexpression mitigated lipid peroxidation in GPX4-deficient cells [74]. Another protective pathway against ferroptosis involves GTP cyclohydrolase-1 (GCH1) and tetrahydrobiopterin (BH4). BH4 serves as a crucial cofactor, possessing significant antioxidant qualities that aid in reducing lipid peroxidation directly and enhancing CoQ10 synthesis [75], 76]. GCH1 functions as a key factor catalyzing the reduction of guanosine triphosphate (GTP) to BH4 [76]. Research has demonstrated that the GCH1-BH4 pathway can inhibit ferroptosis by facilitating lipid remodeling [77], 78].
Ferroptosis regulation in different bone cell lineages
The process of bone remodeling relies on the coordinated actions of several key cell types. These include bone marrow mesenchymal stem cells (MSCs), which give rise to osteoblasts and orchestrate various aspects of remodeling; osteoblasts, which are located on the bone surface and produce the bone matrix; osteocytes, which develop from matrix-embedded osteoblasts and form a mechanosensitive network within the bone while mediating paracrine signaling; and osteoclasts, which continuously degrade and resorb bone matrix [79]. This complex interplay maintains a delicate balance between bone formation and breakdown. As ferroptosis has emerged as a significant mode of regulated cell death, its potential involvement in the function of bone-related cells such as MSCs, osteoblasts, osteoclasts, and chondrocytes has become a focal point of research in recent years [80].
MSCs have been shown to have the ability to inhibit the oxidation of phospholipids and mitigate ferroptotic cell death in both cell and animal models. The involvement of MSC-derived exosomes in these processes has been documented, as they were able to suppress prostaglandin-endoperoxide synthase 2 (PTGS2) expression and upregulate SLC7A11 [81]. The suppressive capacity of MSCs on ferroptosis has also been observed in nerve cells within a mouse model suffering from an acute spinal cord injury, wherein MSCs and their exosomes improved the injury condition by enhancing the levels of FSP1 [82]. MSC-mediated ferroptosis regulation is also achieved by NOP2/Sun RNA methyltransferase 5 (NSUN5)-dependent posttranscriptional methylation of ferritin heavy chain/light-chain (FTH1/FTL) mRNA. When NSUN5 is deficient, there is an increase in intracellular iron level along with a diminished GPX4 level, indicating a critical role of the NSUN5-FTH1/FTL axis in orchestrating ferroptosis in MSCs [83].
Osteoblasts play a crucial role in the formation and mineralization of bone, as well as in the reconstruction of osteocytes. Advanced glycation end products (AGEs) induce osteoblastic ferroptosis and promote osteoporosis [84], while melatonin ameliorates osteoblastic ferroptosis and enhances osteogenic capacity through activating nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling [85]. Mitochondrial ferritin (FtMt) regulates osteoblastic ferroptosis by sequestering iron ions and preventing the accumulation of toxic ferrous ions in the mitochondria. Activation of FtMt ameliorates osteoblastic ferroptosis, while inhibition induces mitophagy through ROS/PTEN-induced kinase 1 (PINK1)/Parkin signaling [86]. Exosomes from vascular endothelial cells and endothelial progenitor cells can prevent osteoblastic ferroptosis by suppressing ferritinophagy and the ferroptotic pathway [87], 88]. Osteoclasts are induced to differentiate and resorb bone by iron ions through ROS production [89], 90]. However, iron overload could induce cell death in osteoclasts through ferroptosis. Zoledronic acid represses osteoclast growth through ferroptosis induction, suppressing cell viability and decreasing GPX4 and GSH levels [91]. Ferroptosis is also involved in osteoclast function during receptor activator of nuclear factor kappa-B ligand (RANKL)-induced differentiation, and hypoxic condition inhibits ferroptosis in osteoclasts by inhibiting RANKL-induced ferritinophagy [92]. Chondrocytes undergo ferroptosis in the pathogenesis of osteoarthritis (OA). Interleukin-1 beta (IL-1β) can trigger ROS generation and alter the expression of ferroptosis-associated genes within chondrocytes, which leads to an increase in MMP13 expression and a reduction in collagen II [93]. Deferoxamine (DFO) and d-mannose ameliorate OA by suppressing chondrocyte ferroptosis, with d-mannose attenuating chondrocyte sensitivity to ferroptosis via hypoxia-inducible factor-2α (HIF-2α) [94], 95]. The insights into ferroptosis regulation in different bone cell lineages suggest promising therapeutic targets for various bone-related diseases. Targeted interventions that enhance the protective mechanisms in MSCs, osteoblasts, and chondrocytes, or inhibit detrimental processes in osteoclasts, hold promise in enhancing bone health and prevent the progression of bone degeneration.
Emerging evidence of ferroptosis in bone-degenerative diseases
Ferroptosis, which involves an intricate interplay of iron metabolism, redox regulation and lipid ROS balance, is garnering significant attention for its implications in bone-degenerative diseases. Characterizing molecular players in the pathogenic ferroptosis of various bone disorders, including osteoporosis, rheumatoid arthritis, osteoarthritis, diabetic bone loss, and lumbar disc herniation, has offered novel perspectives on the mechanisms underlying bone homeostasis and degeneration (Table 1).
Emerging role of ferroptosis in bone-degenerative diseases.
Disease | Cell type | Molecular players | Effect of ferroptosis | Ref. |
---|---|---|---|---|
Diabetic bone loss | Osteoblasts, osteoclasts | GPX4, FtMt, FPN, system Xc-, Nrf2, HO-1, Tfr2 | Contributes to bone loss | [86], [96], [97], [98], [99] |
Osteoarthritis (OA) | Chondrocytes | GPX4, SLC3A2, Nrf2, HO-1, Piezo1, SLC7A11, FSP1, CoQ10 | Promotes chondrocyte death and cartilage damage | [100], [101], [102], [103], [104], [105], [106], [107] |
Rheumatoid arthritis (RA) | Fibroblast-like synoviocytes (FLSs), chondrocytes | SLC7A11, GPX4, FTH1, ACSL4, SEMA5A, SCD1, miR-19b-3p | FLSs resist ferroptosis, promoting RA progression; chondrocyte ferroptosis contributes to cartilage damage | [108], [109], [110], [111], [112], [113], [114], [115], [116], [117], [118], [119] |
Osteoporosis | Bone mesenchymal stem cells (BMSCs), osteoblasts, osteoclasts, osteocytes | FANCD2, FtMt, Nrf2, HO-1, GPX4, system Xc-, HIF-1α, ferritin, hepcidin, sclerostin, RANKL | Contributes to bone loss by affecting differentiation and survival of bone cells | [85], [86], [87], [88, 92], [120], [121], [122], [123], [124] |
Lumbar disc degeneration | Nucleus pulposus cells (NPCs), annulus fibrosus cells, chondrocytes | GPX4, FTH, PTGS2, ACSL4, HO-1, FPN, MTF1, miR-10a-5p, IL-6R | Promotes degeneration of intervertebral disc, nucleus pulposus, and cartilage | [125], [126], [127], [128], [129], [130], [131], [132] |
Diabetic bone loss
Iron dysregulation and ferroptosis have been documented in the development of diabetic bone loss. Under high glucose conditions, iron metabolism is disrupted, culminating in iron over-accumulation and excessive ROS production in bone cells. This iron-dependent oxidative stress induces cell death featured by lipid ROS generation and mitochondrial dysfunction [96]. Several ferroptosis-related proteins have also been implicated in this process. For example, GPX4, a key antioxidant enzyme that protects against lipid peroxidation, is downregulated in the bone tissue of diabetic osteoporosis models. The FtMt protein, which sequesters iron in mitochondria, can suppress high glucose induced ferroptosis in osteoblasts, and FtMt knockdown triggers mitophagy and ferroptosis [86]. The iron exporter ferroportin (FPN) is also involved, as its inhibition by hepcidin causes an increase in intracellular iron and increased ROS production in osteoclasts, promoting osteoporosis [97]. Additionally, the cystine/glutamate antiporter system Xc-, which supplies cysteine in GSH production pathway, was found to be impaired in diabetic bone loss, further compromising antioxidant defenses and facilitating ferroptosis [98]. Contrarily, Nrf2/HO-1 axis has been shown to protect against osteoblast ferroptosis in diabetic osteoporosis by enhancing antioxidant capacity [98]. Lastly, transferrin receptor 2 (Tfr2), which regulates iron uptake, has been found to control bone mass by modulating the BMP-p38 MAPK-Wnt signaling axis in osteoblasts [99]. Together, these studies delineated the complex interplay between iron metabolism, oxidative stress, and ferroptosis in the pathogenesis of diabetic bone loss, with various ferroptosis-related proteins serving as potential therapeutic targets.
Osteoarthritis (OA)
Ferroptosis in chondrocytes has been recognized as a detrimental event in osteoarthritis development. Several lines of evidence suggest the dysregulation of key ferroptosis regulators in OA cartilage and chondrocytes. Transcriptomics profiling along with various other studies have shown a significant decrease in GPX4 expression within OA cartilage [100], [101], [102]. Additionally, it has been documented that the levels of solute carrier family 3 member 2 (SLC3A2) are decreased in OA cartilage [103]. On the other hand, inhibitors of ferroptosis, such as ferrostatin-1 (Fer-1) and desferrioxamine (DFO), have demonstrated the ability to suppress the ferroptosis of chondrocytes and mitigate OA [104]. Yan et al. identified that the ferroptosis activator Erastin instigated chondrocyte ferroptosis while suppressing type II collagen expression; conversely, Fer-1 was able to counteract this process and activate the Nrf2/GPX4 and Nrf2/HO-1 signaling [105]. Excessive mechanical stress also accelerates the progression of OA. Piezo1, a key mechanosensing ion channel, has been implicated in regulating iron homeostasis [106]. Mechanical disruptions activate Piezo1 protein, resulting in an increased inflow of calcium ions. This activation results in a reduction of GSH and inhibition of GPX4, which promotes chondrocyte ferroptosis and subsequently OA [107]. The above evidence highlights chondrocyte ferroptosis as a key player in OA. Nonetheless, the precise underlying mechanisms remain to be elucidated. One potential therapeutic approach for OA is the targeted suppression of chondrocyte ferroptosis, particularly focusing on the GSH-GPX4 pathway.
Rheumatoid arthritis (RA)
Rheumatoid arthritis (RA) is a prevalent autoimmune condition marked by immune cell infiltration and the proliferation of synovial fibroblasts, culminating in cartilage and bone destruction and resulting in aggressive arthritis [108]. The role of ferroptosis is significantly associated with the development of RA. When compared to healthy individuals, patients with RA and osteoarthritis (OA), particularly those with RA, exhibit elevated synovial iron levels [109]. Fibroblast-like synoviocytes (FLSs) are a crucial cell type involved in RA, which promote RA progression by secreting cytokines, supporting neovascularization, and degrading the matrix [110]. Studies have shown that elevated levels of iron and ROS can promote the differentiation of osteoclasts, enhance bone breakdown, induce ferroptotic death in chondrocytes, and lead to the degradation of type II collagen in cartilage [111], 112]. FLSs, however, maintain normal survival and proliferation by resisting ferroptosis. Despite increased ROS levels in RA FLSs, several anti-ferroptosis proteins become upregulated while ACSL4 is reduced, resulting in balanced lipid ROS generation and detoxification through improved antioxidant capacity [113]. Research conducted by Wu and colleagues found that tumor necrosis factor boosts the expression of SLC7A11 along with increased GSH production in FLSs during the process of collagen-induced arthritis, thereby increasing resistance to ferroptosis [114]. Additionally, macrophages demonstrated a protective role against ferroptosis in FLSs upon RSL3 treatment. Another study by Cheng and team uncovered that raised levels of Semaphorin 5A (SEMA5A) synthesized by synovial macrophages enhanced the expression of GPX4 and stearoyl-CoA desaturase-1 (SCD1) through the PI3K/Akt/mTOR pathway. This enhancement inhibits lipid peroxidation and ferroptosis in FLSs [115]. The fact that FLSs are resistant to ferroptosis suggests that providing iron supplements to RA patients suffering from iron deficiency could potentially worsen RA symptoms [116]. Besides, Kong et al. found that exosomes derived from osteoarthritic FLS contain increased levels of microRNA-19b-3p, which targets and downregulates SLC7A11. This leads to a suppression of GSH synthesis and increased oxidative stress in chondrocytes, ultimately promoting ferroptosis and cartilage damage [117]. Recent efforts have been devoted to identifying potential ferroptosis-related genes implicated in RA progression [118]. Given the crucial role of FLSs in RA progression, therapeutic strategies targeting FLSs are needed to restore synovial homeostasis and improve disease outcomes in RA patients [119].
Osteoporosis
Osteoporosis is characterized by a disproportion between the bone resorption activities of osteoclasts and the bone-forming roles of osteoblasts. This condition causes a reduction in bone mass and strength, which elevates the risk of fragility fractures and impairs the healing process [120], 121]. Recent evidence suggests that ferroptosis induction in MSCs, osteoblasts and osteoclasts may be implicated in the pathogenesis of osteoporosis. In bone mesenchymal stem cells (BMSCs), Fanconi anemia complementation group D2 (FANCD2) was found to suppress erastin-induced ferroptosis by reducing iron overload and lipid peroxidation [122], indicating an anti-ferroptosis role of FANCD2 during differentiation of BMSCs under certain circumstances. In a rat model of type 2 diabetes osteoporosis (T2DOP), the overexpression of FtMt reduces oxidative stress and inhibits ferroptosis in osteoblasts. Conversely, knocking down FtMt triggers mitophagy and exacerbating cell death [86], implying that FtMt could be a significant therapeutic target for T2DOP. Furthermore, high glucose levels result in the induction of ferroptosis in the osteoblasts of T2DOP rats through the enhancement of ROS, lipid peroxidation, and GSH consumption. In contrast, melatonin mitigates ferroptosis by stimulating the Nrf2/HO-1 pathway, thereby boosting the osteogenic capacity of MC3T3-E1 cells [85]. Moreover, high doses of steroids can lead to osteoporosis and ferroptosis in osteoblasts, likely through the downregulation of GPX4 and the cystine/glutamate antiporter system Xc-. Interestingly, extracellular vesicles derived from endothelial progenitor cells can prevent osteoblastic ferroptosis by restoring the levels of GPX4 and system Xc-, thus slowing the progression of osteoporosis [88]. Endothelial cell-secreted exosomes could also reverse the detrimental impact of glucocorticoids on osteoblasts by repressing ferritinophagy-dependent ferroptosis [87].
Furthermore, RANKL stimulation has been reported to increase ferroptosis markers, such as malondialdehyde (MDA), PTGS2 and labile iron levels, in bone marrow-derived macrophages undergoing osteoclast differentiation, suggesting ferroptosis may occur during osteoclast differentiation [92]. However, this can be antagonized by hypoxia through the inhibition of RANKL-induced ferritinophagy, indicating an interplay of hypoxia signaling and the survival of osteoclasts. In ovariectomized mice, administration of an HIF-1α inhibitor prevented bone loss by inducing ferroptosis in osteoclasts by targeting HIF-1α and ferritin [92], providing a potential alternative treatment for postmenopausal osteoporosis. Moreover, the deficiency in estrogen levels after menopause can lead to a diminished inhibitory effect of estrogen on HIF-1α, resulting in enhanced osteoclast activity. This mechanism may contribute to the sustained activity of osteoclasts and the development of postmenopausal osteoporosis [123].
Regarding osteocytes, a recent study demonstrated that iron overload induced by hepcidin deficiency led to increased osteocyte cell death and bone degeneration in Hamp−/− mice. The bone loss was linked to heightened expression in osteocyte-derived proteins (sclerostin and RANKL), which inhibits osteogenesis and promotes bone resorption, respectively [124]. Nonetheless, limited research has been conducted to study the potential engagement of ferroptosis in osteocytes within osteoporosis. In summary, ferroptosis appears to occur in both osteoclasts and osteoblasts under various pathological conditions related to osteoporosis, including diabetes, glucocorticoid treatment, and estrogen deficiency. Key players such as GPX4, system Xc-, and iron homeostasis regulators play important roles. Modulating these pathways may serve as new intervention options for managing osteoporosis. Future research is required to fully decipher the underlying mechanisms.
Lumbar disc degeneration
Lumbar disc herniation is a widespread cause of persistent low back pain, impacting 70%–85 % of the global population. The main reason for lumbar disc herniation is intervertebral disc degeneration, influenced by multiple factors including age [125]. The causes of intervertebral disc degeneration encompass AF rupture, oxidative stress in NPC, and cartilage endplate degeneration [126]. Recent studies propose a potential role of ferroptosis in intervertebral disc degeneration. In individuals with intervertebral disc degeneration, the levels of GPX4 and FTH in disc tissue were decreased compared to those without the condition, while the levels of the ferroptosis marker PTGS2 were elevated, indicating the presence of ferroptosis in degenerative disc tissues [127]. Exposure to tert-butyl hydroperoxide (TBHP, a chemical that induces oxidative stress) led to an increase in PTGS2 and ACSL4 levels, accelerated lipid peroxidation, and reduced the expression of FTH and GPX4 in both AF cells and NPCs. The impact of these changes was counteracted by ferroptosis inhibitors Fer-1 and DFO [127], 128]. Additionally, inducing iron overload experimentally was found to exacerbate the deterioration of intervertebral discs and cartilage endplates in a manner dependent on the dosage [128]. Treatment with DFO delayed the progression of disc degeneration by suppressing ferroptosis, indicating that ferroptosis is involved in the pathophysiological process of intervertebral disc degeneration. Furthermore, the levels of hemoglobin signal and iron granules were found to be significantly elevated in herniated NPs compared to nonherniated NPs [129]. Higher expression of HO-1 was observed in the NP of herniated discs, and heme-mediated ferroptosis was associated with alterations in the Notch signaling pathway, which could be mitigated by co-treatment with DFO [129].
Single-cell RNA sequencing identified that various ferroptosis-associated genes exhibited differential expression in the nucleus pulposus cells (NPCs) of individuals with intervertebral disc degeneration (IDD) when compared to healthy controls [130]. This discovery suggests an alteration in the propensity for ferroptosis. Dysregulation of FPN has been implicated in causing intercellular iron accumulation in cases of IDD. When FPN expression was silenced, there was an elevation in iron levels within NPCs. Conversely, overexpression of FPN mitigated iron accumulation and reduced ferroptosis [131]. Additionally, nuclear translocation of the metal-regulated transcription factor 1 (MTF1) enhanced FPN transcription, thereby restoring its capacity to alleviate iron overload and safeguarding NPCs from ferroptosis. The deregulation of non-coding RNAs also plays a role in influencing ferroptosis in intervertebral disc cartilage. In comparison to normal tissue, the expression of miR-10a-5p was found to be diminished in degenerative intervertebral disc cartilage, whereas its target gene, IL-6R, was upregulated [132]. IL-6 induced chondrocyte ferroptosis by suppressing GPX4 expression in diseased cartilage, suggesting the involvement of IL-6-mediated inflammatory axis in chondrocyte loss during intervertebral disc degeneration [132]. Overall, recent studies have unveiled ferroptotic features in NPCs and the cartilage tissues of herniated intervertebral disc, and experimental evidence indicates the beneficial effect of targeting oxidative stress, iron overload and ferroptosis. This opens an avenue to mitigate the detrimental progression of lumbar disc degeneration.
Targeting oxidative imbalance, iron overload and ferroptosis in bone-degenerative conditions
Compelling evidence of oxidative stress, iron overload, and ferroptosis in bone-degenerative disorders has prompted preclinical endeavors to alleviate these conditions by targeting ferroptosis (Table 2), yielding promising experimental outcomes.
Targeting oxidative stress, iron overload and ferroptosis in bone-degenerative conditions.
Intervention approach or agent | Molecular targets | Effect on cell types | Effect on disease | Ref. |
---|---|---|---|---|
Melatonin | Nrf2/HO-1/GPX4, SLC7A11 | Inhibits ferroptosis in osteoblasts | Alleviates type 2 diabetes-induced osteoporosis | [85] |
Qing’e pill | DMT1, TFR1, FPN1, GPX4 | Reduces iron accumulation and ferroptosis in osteoblasts | Attenuates diabetic osteoporosis | [133] |
Artesunate (ART) | Intracellular iron, ROS, TRAP, CTR, CTSK | Inhibits osteoclast differentiation and bone resorption | Alleviates diabetic osteoporosis | [134] |
Ferrostatin-1, liproxstatin-1 | Iron accumulation, lipid peroxidation, Nrf2 | Inhibits ferroptosis in osteoblasts, osteocytes, chondrocytes | Prevents diabetic bone loss, mitigates osteoarthritis | [98], 104], 135] |
Deferoxamine (DFO) | Intracellular iron, ROS, Nrf2 | Inhibits ferroptosis in osteoblasts, MSCs, chondrocytes, annulus fibrosus cells, nucleus pulposus cells | Attenuates diabetic osteoporosis, osteoarthritis, intervertebral disc degeneration | [35], 94], 127], 128], 136] |
Mitochondrial ferritin (FtMt) overexpression | Oxidative stress, ROS/PINK1/Parkin | Inhibits ferroptosis in osteoblasts | Prevents bone loss in type 2 diabetic osteoporosis | [86] |
2-Methoxyestradiol (2ME2) | HIF-1α, ferritin | Induces ferroptosis in osteoclasts | Alternative treatment for postmenopausal osteoporosis | [92] |
Endothelial cell-derived extracellular vesicles (EPC-EVs) | GPX4, system Xc- | Inhibits ferroptosis in osteoblasts | Prevents glucocorticoid-induced osteoporosis | [88] |
Carbonyl cyanide-m-chlorophenyl-hydrazone (CCCP) | GPX4, ROS | Induces ferroptosis in osteoblasts | Contributes to osteoporosis | [86] |
GPX4 activation | GPX4 | Reduces chondrocyte sensitivity to oxidative stress | Slows osteoarthritis progression | [100] |
d-mannose, stigmasterol, icariin | GPX4, lipid peroxidation | Inhibits ferroptosis in chondrocytes | Delays osteoarthritis development | [95], 137], 138] |
BAPTA acetoxymethyl ester | Iron influx, ROS, mitochondrial dysfunction | Potentially attenuates ferroptosis in chondrocytes | Potential effect on osteoarthritis | [139] |
Rosiglitazone | ACSL4, lipid peroxidation | Inhibits ferroptosis in chondrocytes | Potential therapeutic effect on osteoarthritis | [140], 141] |
Ferroportin (FPN) regulation | FPN, iron overload | Attenuates iron overload and ferroptosis in nucleus pulposus cells | Potential effect on intervertebral disc degeneration | [131] |
Metal-regulated transcription factor 1 (MTF1) | FPN transcription | Eliminates iron overload and protects nucleus pulposus cells from ferroptosis | Potential effect on intervertebral disc degeneration | [131] |
Melatonin
Under high glucose conditions, melatonin triggers the pathways Nrf2/HO-1/GPX4 and SLC7A11 in osteoblasts. This action decreases ROS levels and lipid peroxidation, ultimately preventing ferroptosis. Consequently, osteoblasts exhibit enhanced osteogenic capability, alleviating osteoporosis induced by type 2 diabetes [85].
Qing’e pill
This traditional Chinese medicine preparation reduces DMT1 and TFR1 levels while promoting FPN1 expression in osteoblasts, reducing intracellular iron accumulation. It also upregulates GPX4 expression and inhibits ROS production, thus suppressing ferroptosis in osteoblasts and attenuating diabetic osteoporosis [133].
Artesunate (ART)
ART, a drug for malaria prevention, hinders the differentiation of osteoclasts and bone resorption in elevated glucose environments by controlling the iron levels and production of ROS inside cells. By reducing the activity of genes specific to osteoclasts such as TRAP, CTR, and CTSK, it helps mitigate diabetic osteoporosis [134].
Ferrostatin-1 and liproxstatin-1
These are ferroptosis inhibitors that reduce iron overload and lipid ROS levels in osteoblasts and osteocytes under diabetic conditions. They inhibit ROS production, preserve mitochondrial function, and prevent cell death, thereby maintaining bone homeostasis and preventing diabetic bone loss [98], 135]. These inhibitors also decrease lipid peroxidation and ferroptosis induced by IL-1β and ferric ammonium citrate in chondrocytes by enhancing Nrf2 system activation, reducing the advancement of osteoarthritis [104].
Deferoxamine (DFO)
DFO acts as an iron chelator to reduce intracellular iron levels and ROS production in osteoblasts and MSCs under high glucose conditions. It promotes osteogenic differentiation of MSCs and enhances osteoblast function, thus attenuating diabetic osteoporosis [35], 136]. Furthermore, DFO improves osteoarthritis by blocking chondrocyte ferroptosis by activating the Nrf2 pathway [94]. DFO has also been reported to delay disc deterioration by mitigating ferroptosis in annulus fibrosus cells and nucleus pulposus cells, demonstrating the therapeutic potential of iron chelation in intervertebral disc degeneration [127], 128].
Mitochondrial ferritin (FtMt) overexpression
Overexpression of FtMt in osteoblasts reduces high glucose induced oxidative stress by storing excess iron in mitochondria. This inhibits ferroptosis in osteoblasts and prevents bone loss in type 2 diabetic osteoporosis. Conversely, FtMt knockdown induces mitochondrial autophagy via the ROS/PINK1/Parkin pathway and promotes ferroptosis in osteoblasts [86].
2-Methoxyestradiol (2ME2)
The HIF-1α-specific inhibitor 2ME2 was found in in vivo experiments to protect against bone loss in ovariectomized mice, mimicking postmenopausal osteoporosis. By focusing on HIF-1α and ferritin, 2ME2 triggers ferroptosis in osteoclasts. The researchers suggest that triggering ferroptosis in osteoclasts via the targeting of HIF-1α and ferritin may offer a new approach to treating osteoporosis [92].
Endothelial cell-derived extracellular vesicles (EPC-EVs)
EPC-EVs hinder the initiation of the ferroptosis pathway in bone cells by reinstating GPX4 and system Xc-levels. The administration of EPC-EVs reverted alterations triggered by dexamethasone in cysteine and indicators of oxidative stress (MDA, GSH, GSSG) and enhanced bone characteristics in rodents. These findings propose that EPC-EVs have the potential to prevent glucocorticoid-triggered osteoporosis through the inhibition of ferroptotic pathway in osteoblasts [88].
Carbonyl cyanide-m-chlorophenyl-hydrazone (CCCP)
CCCP is a mitochondrial uncoupling agent that activates mitophagy. Treatment of osteoblasts with CCCP increased ferroptosis and disrupted cellular functions, as evidenced by reduced levels of GPX4, osteocalcin, alkaline phosphatase, and osteoprotegerin, impaired formation of mineralized nodules, and elevated production of ROS and lipid peroxidation. Ferroptosis inhibitors rescued these effects, suggesting that mitochondrial dysfunction and ferroptosis in osteoblasts may contribute to osteoporosis [86].
GPX4 activation
GPX4 expression was reported to be downregulated in the cartilage tissue of osteoarthritis patients. Upregulating GPX4 decreases the susceptibility of chondrocytes to oxidative damage, reduces cartilage degeneration, and slows osteoarthritis progression [100].
D-Mannose, stigmasterol, and icariin
By activating the GPX4 pathway or reducing lipid peroxidation, natural products with antioxidant or anti-inflammatory properties can slow down cartilage degradation and inhibit ferroptosis in chondrocytes. This, in turn, helps delay the development of osteoarthritis [95], 137], 138].
BAPTA acetoxymethyl ester
This chelator of calcium prevents the import of iron into chondrocytes, which hinders the production of ROS and the dysfunction of mitochondria caused by an excess of iron, thus potentially attenuating ferroptosis in osteoarthritis [139].
Rosiglitazone
This PPAR-γ activator inhibits the production of lipid ROS and ferroptosis in chondrocytes by suppressing ACSL4 activity, suggesting a promising beneficial impact on osteoarthritis [140], 141].
Ferroportin (FPN) regulation
FPN dysregulation causes an over-accumulation of intercellular iron in intervertebral disc degeneration. FPN overexpression alleviated iron overload and ferroptosis in nucleus pulposus cells [131].
Metal-regulated transcription factor 1 (MTF1)
Increased nuclear translocation of MTF1 promotes FPN transcription and restored its activity to expel excessive iron, preventing nucleus pulposus cells against ferroptosis [131].
miR-10a-5p/IL-6R axis
MiR-10a-5p levels were decreased in degenerative intervertebral disc cartilage, which causes an increase in IL-6R expression. The activation of IL-6 led to chondrocyte ferroptosis, implying a potential link between this pathway and intervertebral disc degeneration [132].
Together, these examples highlight different approaches to regulating iron metabolism and ferroptosis in key cell types involved in diabetic bone loss, osteoporosis, osteoarthritis and intervertebral disc degeneration, including MSCs, osteoblasts, osteoclasts, chondrocytes, annulus fibrosus cells, and nucleus pulposus cells. These interventions target crucial molecules and pathways such as GPX4, Nrf2, ACSL4, FPN, and MTF1. By modulating these targets, the primary goal is to inhibit excessive iron accumulation, lipid ROS generation, and ferroptosis, thereby maintaining joint and intervertebral disc homeostasis and preventing disease progression.
Conclusions and perspectives
Recent studies have presented compelling evidence for the involvement of oxidative stress, iron overload, and ferroptosis in various bone-degenerative conditions, such as osteoporosis, osteoarthritis, and intervertebral disc degeneration [142], [143], [144], [145], [146], [147], [148]. The complex interactions between iron metabolism, lipid peroxidation, and antioxidant defense systems have been found to play a crucial role in the development and progression of bone-degenerative disorders. Targeting these pathways offers insights into novel therapeutic interventions for the prevention and treatment of bone-degenerative diseases [149], 150].
However, several challenges and unanswered questions remain in this field of research. First, the precise molecular mechanisms underlying the regulation of ferroptosis in bone cells, such as osteocytes and chondrocytes, need to be further elucidated. It remains unclear whether ferroptosis occurs as a spontaneous event during cell differentiation, or if it is only induced under specific conditions. Furthermore, the factors contributing to iron overload in bone-degenerative conditions need to be further clarified. Second, the crosstalk between ferroptosis and other forms of cell death, such as apoptosis and autophagy, in the context of bone degeneration warrants further investigation [151], 152]. Importantly, the revelation of specific biomarkers and signaling pathways associated with ferroptosis in bone-degenerative conditions is crucial for the development of targeted therapies and diagnostic tools. Particularly, there is an increasing number of reports regarding the involvement of non-coding RNAs in ferroptosis regulation under pathophysiological conditions related to iron overload [153]. Characterization and validation of reliable non-coding RNA targets related to ferroptosis could provide useful targets for therapeutic interventions.
Future research should focus on unraveling the complex networks and molecular mechanisms governing ferroptosis in bone-degenerative diseases. The use of advanced techniques, such as single-cell RNA sequencing, proteomics, and metabolomics, may unravel a holistic picture of ferroptosis in different osteogenic lineages under various bone-regenerative conditions, offering valuable insights into dysregulated bone remodeling [154]. Furthermore, it is crucial to develop targeted inhibitors of ferroptosis and assess the efficacy of botanical compounds in animal models of bone degeneration to facilitate the translation of these results to practical medical treatments [155], 156]. By gaining a deeper comprehension of the functions and control mechanisms of ferroptosis in bone homeostasis and disease, we can lay the foundations for the formulation of novel and effective strategies to combat age-related orthopedic disorders.
Funding source: Yunnan Province technology innovation talent training object project
Award Identifier / Grant number: 202005AD160146
Funding source: General Project of Yunnan Province Kunming Medical Joint Special Basic Research Program
Award Identifier / Grant number: 202001AC070337
Funding source: Yunnan Orthopedics and sports rehabilitation Clinical Medical Research Center
Award Identifier / Grant number: 202102AA310068
-
Research ethics: Not applicable.
-
Informed consent: Not applicable.
-
Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission. All authors were involved in the conceptualization and design of the study. Junxiao Ren provided experimental ideas. CM completed the first draft of the manuscript. RL and YX performed the material preparation, data collection and analysis. CL was mainly responsible for reviewing the final manuscript. All authors participated in the editorial revision of the manuscript. All authors read and approved the final manuscript.
-
Use of Large Language Models, AI and Machine Learning Tools: None declared.
-
Conflict of interest: The authors state no conflict of interest.
-
Research funding: This work was supported by Yunnan Orthopedics and sports rehabilitation Clinical Medical Research Center (202102AA310068). General Project of Yunnan Province Kunming Medical Joint Special Basic Research Program (202001AC070337). Yunnan Province technology innovation talent training object project (202005AD160146).
-
Data availability: Not applicable.
References
1. Zhang, AS, Xu, A, Ansari, K, Hardacker, K, Anderson, G, Alsoof, D, et al.. Lumbar disc herniation: diagnosis and management. Am J Med 2023;136:645–51. https://doi.org/10.1016/j.amjmed.2023.03.024.Search in Google Scholar PubMed
2. Wu, B, Fu, Z, Wang, X, Zhou, P, Yang, Q, Jiang, Y, et al.. A narrative review of diabetic bone disease: characteristics, pathogenesis, and treatment. Front Endocrinol 2022;13:1052592. https://doi.org/10.3389/fendo.2022.1052592.Search in Google Scholar PubMed PubMed Central
3. Kim, D, Pirshahid, AA, Li, Y, Varghese, T, Pope, JE. Prevalence of osteoporosis in osteoarthritis: a systematic review and meta-analysis. Osteoporos Int 2022;33:1687–93. https://doi.org/10.1007/s00198-022-06376-0.Search in Google Scholar PubMed
4. Xiao, PL, Cui, AY, Hsu, CJ, Peng, R, Jiang, N, Xu, XH, et al.. Global, regional prevalence, and risk factors of osteoporosis according to the World Health Organization diagnostic criteria: a systematic review and meta-analysis. Osteoporos Int 2022;33:2137–53. https://doi.org/10.1007/s00198-022-06454-3.Search in Google Scholar PubMed
5. Finckh, A, Gilbert, B, Hodkinson, B, Bae, SC, Thomas, R, Deane, KD, et al.. Global epidemiology of rheumatoid arthritis. Nat Rev Rheumatol 2022;18:591–602. https://doi.org/10.1038/s41584-022-00827-y.Search in Google Scholar PubMed
6. Muhammad, T, Maurya, P, Sharma, P. Prevalence and correlates of bone and joint diseases and its association with falls among older adults in India: evidence from LASI, 2017-18. Geriatr Nurs 2021;42:1143–50. https://doi.org/10.1016/j.gerinurse.2021.07.007.Search in Google Scholar PubMed
7. Afzal, S, Abdul Manap, AS, Attiq, A, Albokhadaim, I, Kandeel, M, Alhojaily, SM. From imbalance to impairment: the central role of reactive oxygen species in oxidative stress-induced disorders and therapeutic exploration. Front Pharmacol 2023;14:1269581. https://doi.org/10.3389/fphar.2023.1269581.Search in Google Scholar PubMed PubMed Central
8. Cervellati, C, Bonaccorsi, G, Cremonini, E, Romani, A, Fila, E, Castaldini, MC, et al.. Oxidative stress and bone resorption interplay as a possible trigger for postmenopausal osteoporosis. BioMed Res Int 2014;2014:569563. https://doi.org/10.1155/2014/569563.Search in Google Scholar PubMed PubMed Central
9. Phull, AR, Nasir, B, Haq, IU, Kim, SJ. Oxidative stress, consequences and ROS mediated cellular signaling in rheumatoid arthritis. Chem Biol Interact 2018;281:121–36. https://doi.org/10.1016/j.cbi.2017.12.024.Search in Google Scholar PubMed
10. Jeney, V. Clinical impact and cellular mechanisms of iron overload-associated bone loss. Front Pharmacol 2017;8:77. https://doi.org/10.3389/fphar.2017.00077.Search in Google Scholar PubMed PubMed Central
11. Cai, C, Hu, W, Chu, T. Interplay between iron overload and osteoarthritis: clinical significance and cellular mechanisms. Front Cell Dev Biol 2022;9:817104. https://doi.org/10.3389/fcell.2021.817104.Search in Google Scholar PubMed PubMed Central
12. Chen, H, Han, Z, Wang, Y, Su, J, Lin, Y, Cheng, X, et al.. Targeting ferroptosis in bone-related diseases: facts and perspectives. J Inflamm Res 2023;16:4661–77. https://doi.org/10.2147/jir.s432111.Search in Google Scholar PubMed PubMed Central
13. Stockwell, BR, Jiang, X, Gu, W. Emerging mechanisms and disease relevance of ferroptosis. Trends Cell Biol 2020;30:478–90. https://doi.org/10.1016/j.tcb.2020.02.009.Search in Google Scholar PubMed PubMed Central
14. Jia, P, Xu, YJ, Zhang, ZL, Li, K, Li, B, Zhang, W, et al.. Ferric ion could facilitate osteoclast differentiation and bone resorption through the production of reactive oxygen species. J Orthop Res 2012;30:1843–52. https://doi.org/10.1002/jor.22133.Search in Google Scholar PubMed
15. Che, J, Lv, H, Yang, J, Zhao, B, Zhou, S, Yu, T, et al.. Iron overload induces apoptosis of osteoblast cells via eliciting ER stress-mediated mitochondrial dysfunction and p-eIF2α/ATF4/CHOP pathway in vitro. Cell Signal 2021;84:110024. https://doi.org/10.1016/j.cellsig.2021.110024.Search in Google Scholar PubMed
16. Jiang, Z, Wang, H, Qi, G, Jiang, C, Chen, K, Yan, Z. Iron overload-induced ferroptosis of osteoblasts inhibits osteogenesis and promotes osteoporosis: an in vitro and in vivo study. IUBMB Life 2022;74:1052–69. https://doi.org/10.1002/iub.2656.Search in Google Scholar PubMed
17. Jiang, Z, Qi, G, He, X, Yu, Y, Cao, Y, Zhang, C, et al.. Ferroptosis in osteocytes as a target for protection against postmenopausal osteoporosis. Adv Sci 2024;11:e2307388. https://doi.org/10.1002/advs.202307388.Search in Google Scholar PubMed PubMed Central
18. Callaway, DA, Jiang, JX. Reactive oxygen species and oxidative stress in osteoclastogenesis, skeletal aging and bone diseases. J Bone Miner Metabol 2015;33:359–70. https://doi.org/10.1007/s00774-015-0656-4.Search in Google Scholar PubMed
19. Lee, NK, Choi, YG, Baik, JY, Han, SY, Jeong, D, Bae, YS, et al.. A crucial role for reactive oxygen species in RANKL-induced osteoclast differentiation. Blood 2005;106:852–9. https://doi.org/10.1182/blood-2004-09-3662.Search in Google Scholar PubMed
20. Lean, JM, Jagger, CJ, Kirstein, B, Fuller, K, Chambers, TJ. Hydrogen peroxide is essential for estrogen-deficiency bone loss and osteoclast formation. Endocrinology 2005;146:728–35. https://doi.org/10.1210/en.2004-1021.Search in Google Scholar PubMed
21. Iantomasi, T, Romagnoli, C, Palmini, G, Donati, S, Falsetti, I, Miglietta, F, et al.. Oxidative stress and inflammation in osteoporosis: molecular mechanisms involved and the relationship with microRNAs. Int J Mol Sci 2023;24:3772. https://doi.org/10.3390/ijms24043772.Search in Google Scholar PubMed PubMed Central
22. Lepetsos, P, Papavassiliou, AG. ROS/oxidative stress signaling in osteoarthritis. Biochim Biophys Acta 2016;1862:576–91. https://doi.org/10.1016/j.bbadis.2016.01.003.Search in Google Scholar PubMed
23. Lin, CH, Li, NT, Cheng, HS, Yen, ML. Oxidative stress induces imbalance of adipogenic/osteoblastic lineage commitment in mesenchymal stem cells through decreasing SIRT1 functions. J Cell Mol Med 2018;22:786–96. https://doi.org/10.1111/jcmm.13356.Search in Google Scholar PubMed PubMed Central
24. Ishii, KA, Fumoto, T, Iwai, K, Takeshita, S, Itoa, M, Shimohata, N, et al.. Coordination of PGC-1beta and iron uptake in mitochondrial biogenesis and osteoclast activation. Nat Med 2009;15:259–66. https://doi.org/10.1038/nm.1910.Search in Google Scholar PubMed
25. Ryu, MS, Lichten, LA, Liuzzi, JP, Cousins, RJ. Zinc transporters ZnT1 (Slc30a1), Zip8 (Slc39a8), and Zip10 (Slc39a10) in mouse red blood cells are differentially regulated during erythroid development and by dietary zinc deficiency. J Nutr 2008;138:2076–83. https://doi.org/10.3945/jn.108.093575.Search in Google Scholar PubMed PubMed Central
26. Frade, BB, Dias, RB, Piperni, SG, Bonfim, DC. The role of macrophages in fracture healing: a narrative review of the recent updates and therapeutic perspectives. Stem Cell Investig 2023;10:4.10.21037/sci-2022-038Search in Google Scholar PubMed PubMed Central
27. Arnett, TR, Orriss, IR. Metabolic properties of the osteoclast. Bone 2018;115:25–30. https://doi.org/10.1016/j.bone.2017.12.021.Search in Google Scholar PubMed
28. Xiao, W, Beibei, F, Guangsi, S, Yu, J, Wen, Z, Xi, H, et al.. Iron overload increases osteoclastogenesis and aggravates the effects of ovariectomy on bone mass. J Endocrinol 2015;226:121–34. https://doi.org/10.1530/joe-14-0657.Search in Google Scholar
29. Jia, P, Xu, YJ, Zhang, ZL, Li, K, Li, B, Zhang, W, et al.. Ferric ion could facilitate osteoclast differentiation and bone resorption through the production of reactive oxygen species. J Orthop Res 2012;30:1843–52. https://doi.org/10.1002/jor.22133.Search in Google Scholar PubMed
30. Doyard, M, Fatih, N, Monnier, A, Island, ML, Aubry, M, Leroyer, P, et al.. Iron excess limits HHIPL-2 gene expression and decreases osteoblastic activity in human MG-63 cells. Osteoporos Int 2012;23:2435–45. https://doi.org/10.1007/s00198-011-1871-z.Search in Google Scholar PubMed
31. Guggenbuhl, P, Fergelot, P, Doyard, M, Libouban, H, Roth, MP, Gallois, Y, et al.. Bone status in a mouse model of genetic hemochromatosis. Osteoporos Int 2011;22:2313–19. https://doi.org/10.1007/s00198-010-1456-2.Search in Google Scholar PubMed
32. Zarjou, A, Jeney, V, Arosio, P, Poli, M, Zavaczki, E, Balla, G, et al.. Ferritin ferroxidase activity: a potent inhibitor of osteogenesis. J Bone Miner Res 2010;25:164–72. https://doi.org/10.1359/jbmr.091002.Search in Google Scholar PubMed
33. Zarjou, A, Jeney, V, Arosio, P, Poli, M, Antal-Szalma[Combining Acute Accent]s, PAA, Agarwal, A, et al.. Ferritin prevents calcification and osteoblastic differentiation of vascular smooth muscle cells. J Am Soc Nephrol 2009;20:1254–63. https://doi.org/10.1681/asn.2008070788.Search in Google Scholar
34. Yang, X, Jiang, T, Wang, Y, Guo, L. The role and mechanism of SIRT1 in resveratrol-regulated osteoblast autophagy in osteoporosis rats. Sci Rep 2019;9:18424. https://doi.org/10.1038/s41598-019-44766-3.Search in Google Scholar PubMed PubMed Central
35. Balogh, E, Tolnai, E, Nagy, BJr, Balla, G, Balla, J, Jeney, V. Iron overload inhibits osteogenic commitment and differentiation of mesenchymal stem cells via the induction of ferritin. Biochim Biophys Acta 2016;1862:1640–9. https://doi.org/10.1016/j.bbadis.2016.06.003.Search in Google Scholar PubMed
36. Chang, YK, Liu, YP, Ho, JH, Hsu, SC, Lee, OK. Amine-surface-modified superparamagnetic iron oxide nanoparticles interfere with differentiation of human mesenchymal stem cells. J Orthop Res 2012;30:1499–506. https://doi.org/10.1002/jor.22088.Search in Google Scholar PubMed
37. Liu, Y, Zhang, Z, Fang, Y, Liu, C, Zhang, H. Ferroptosis in osteoarthritis: current understanding. J Inflamm Res 2024;17:8471–86.10.2147/JIR.S493001Search in Google Scholar PubMed PubMed Central
38. Chen, Z, Zheng, S, Li, L, Jiang, H. Metabolism of flavonoids in human: a comprehensive review. Curr Drug Metabol 2014;15:48–61. https://doi.org/10.2174/138920021501140218125020.Search in Google Scholar PubMed
39. Yao, X, Cheng, X, Zhang, L, He, S, Wang, Y, Lin, Y, et al.. Punicalagin, a PTP1B inhibitor, induces M2c phenotype polarization via up-regulation of HO-1 in murine macrophages. Free Radic Biol Med 2017;110:408–20. https://doi.org/10.1016/j.freeradbiomed.2017.06.014.Search in Google Scholar PubMed
40. Abbaspour, N, Hurrell, R, Kelishadi, R. Review on iron and its importance for human health. J Res Med Sci 2014;19:164–74.Search in Google Scholar
41. Hirschberg, K, Miller, CM, Ellenberg, J, Presley, JF, Siggia, ED, Phair, RD, et al.. Kinetic analysis of secretory protein traffic and characterization of golgi to plasma membrane transport intermediates in living cells. J Cell Biol 1998;143:1485–503. https://doi.org/10.1083/jcb.143.6.1485.Search in Google Scholar PubMed PubMed Central
42. Arosio, P, Ingrassia, R, Cavadini, P. Ferritins: a family of molecules for iron storage, antioxidation and more. Biochim Biophys Acta 2009;1790:589–99. https://doi.org/10.1016/j.bbagen.2008.09.004.Search in Google Scholar PubMed
43. Ward, DM, Kaplan, J. Ferroportin-mediated iron transport: expression and regulation. Biochim Biophys Acta 2012;1823:1426–33. https://doi.org/10.1016/j.bbamcr.2012.03.004.Search in Google Scholar PubMed PubMed Central
44. Vulpe, CD, Kuo, YM, Murphy, TL, Cowley, L, Askwith, C, Libina, N, et al.. Hephaestin, a ceruloplasmin homologue implicated in intestinal iron transport, is defective in the sla mouse. Nat Genet 1999;21:195–9. https://doi.org/10.1038/5979.Search in Google Scholar PubMed
45. Gkouvatsos, K, Papanikolaou, G, Pantopoulos, K. Regulation of iron transport and the role of transferrin. Biochim Biophys Acta 2012;1820:188–202. https://doi.org/10.1016/j.bbagen.2011.10.013.Search in Google Scholar PubMed
46. Dautry-Varsat, A, Ciechanover, A, Lodish, HF. pH and the recycling of transferrin during receptor-mediated endocytosis. Proc Natl Acad Sci USA 1983;80:2258–62. https://doi.org/10.1073/pnas.80.8.2258.Search in Google Scholar PubMed PubMed Central
47. Ohgami, RS, Campagna, DR, McDonald, A, Fleming, MD. The Steap proteins are metalloreductases. Blood 2006;108:1388–94. https://doi.org/10.1182/blood-2006-02-003681.Search in Google Scholar PubMed PubMed Central
48. Liu, J, Kuang, F, Kroemer, G, Klionsky, DJ, Kang, R, Tang, D. Autophagy-dependent ferroptosis: machinery and regulation. Cell Chem Biol 2020;27:420–35. https://doi.org/10.1016/j.chembiol.2020.02.005.Search in Google Scholar PubMed PubMed Central
49. Fang, X, Wang, H, Han, D, Xie, E, Yang, X, Wei, J, et al.. Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci USA 2019;116:2672–80. https://doi.org/10.1073/pnas.1821022116.Search in Google Scholar PubMed PubMed Central
50. Shah, R, Shchepinov, MS, Pratt, DA. Resolving the role of lipoxygenases in the initiation and execution of ferroptosis. ACS Cent Sci 2018;4:387–96. https://doi.org/10.1021/acscentsci.7b00589.Search in Google Scholar PubMed PubMed Central
51. Yan, B, Ai, Y, Sun, Q, Ma, Y, Cao, Y, Wang, J, et al.. Membrane damage during ferroptosis is caused by oxidation of phospholipids catalyzed by the oxidoreductases POR and CYB5R1. Mol Cell 2021;81:355–69.e10. https://doi.org/10.1016/j.molcel.2020.11.024.Search in Google Scholar PubMed
52. Hentze, MW, Muckenthaler, MU, Galy, B, Camaschella, C. Two to tango: regulation of Mammalian iron metabolism. Cell 2010;142:24–38. https://doi.org/10.1016/j.cell.2010.06.028.Search in Google Scholar PubMed
53. Mancias, JD, Wang, X, Gygi, SP, Harper, JW, Kimmelman, AC. Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature 2014;509:105–9. https://doi.org/10.1038/nature13148.Search in Google Scholar PubMed PubMed Central
54. Ganz, T. Systemic iron homeostasis. Physiol Rev 2013;93:1721–41. https://doi.org/10.1152/physrev.00008.2013.Search in Google Scholar PubMed
55. Agmon, E, Solon, J, Bassereau, P, Stockwell, BR. Modeling the effects of lipid peroxidation during ferroptosis on membrane properties. Sci Rep 2018;8:5155. https://doi.org/10.1038/s41598-018-23408-0.Search in Google Scholar PubMed PubMed Central
56. Soupene, E, Kuypers, FA. Mammalian long-chain acyl-CoA synthetases. Exp Biol Med 2008;233:507–21. https://doi.org/10.3181/0710-mr-287.Search in Google Scholar PubMed PubMed Central
57. Dixon, SJ, Winter, GE, Musavi, LS, Lee, ED, Snijder, B, Rebsamen, M, et al.. Human haploid cell genetics reveals roles for lipid metabolism genes in nonapoptotic cell death. ACS Chem Biol 2015;10:1604–9. https://doi.org/10.1021/acschembio.5b00245.Search in Google Scholar PubMed PubMed Central
58. Hassannia, B, Wiernicki, B, Ingold, I, Qu, F, Van Herck, S, Tyurina, YY, et al.. Nano-targeted induction of dual ferroptotic mechanisms eradicates high-risk neuroblastoma. J Clin Invest 2018;128:3341–55. https://doi.org/10.1172/jci99032.Search in Google Scholar
59. Magtanong, L, Ko, PJ, To, M, Cao, JY, Forcina, GC, Tarangelo, A, et al.. Exogenous monounsaturated fatty acids promote a ferroptosis-resistant cell state. Cell Chem Biol 2019;26:420–32.e9. https://doi.org/10.1016/j.chembiol.2018.11.016.Search in Google Scholar PubMed PubMed Central
60. Gaschler, MM, Stockwell, BR. Lipid peroxidation in cell death. Biochem Biophys Res Commun 2017;482:419–25. https://doi.org/10.1016/j.bbrc.2016.10.086.Search in Google Scholar PubMed PubMed Central
61. Doll, S, Proneth, B, Tyurina, YY, Panzilius, E, Kobayashi, S, Ingold, I, et al.. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol 2017;13:91–8. https://doi.org/10.1038/nchembio.2239.Search in Google Scholar PubMed PubMed Central
62. Yang, WS, SriRamaratnam, R, Welsch, ME, Shimada, K, Skouta, R, Viswanathan, V, et al.. Regulation of ferroptotic cancer cell death by GPX4. Cell 2014;156:317–31. https://doi.org/10.1016/j.cell.2013.12.010.Search in Google Scholar PubMed PubMed Central
63. Shah, R, Shchepinov, MS, Pratt, DA. Resolving the role of lipoxygenases in the initiation and execution of ferroptosis. ACS Cent Sci 2018;4:387–96. https://doi.org/10.1021/acscentsci.7b00589.Search in Google Scholar PubMed PubMed Central
64. Brigelius-Flohé, R, Maiorino, M. Glutathione peroxidases. Biochim Biophys Acta 2013;1830:3289–303. https://doi.org/10.1016/j.bbagen.2012.11.020.Search in Google Scholar PubMed
65. Seibt, TM, Proneth, B, Conrad, M. Role of GPX4 in ferroptosis and its pharmacological implication. Free Radic Biol Med 2019;133:144–52. https://doi.org/10.1016/j.freeradbiomed.2018.09.014.Search in Google Scholar PubMed
66. Cao, JY, Dixon, SJ. Mechanisms of ferroptosis. Cell Mol Life Sci 2016;73:2195–209. https://doi.org/10.1007/s00018-016-2194-1.Search in Google Scholar PubMed PubMed Central
67. Conrad, M, Sato, H. The oxidative stress-inducible cystine/glutamate antiporter, system x (c) (-) : cystine supplier and beyond. Amino Acids 2012;42:231–46. https://doi.org/10.1007/s00726-011-0867-5.Search in Google Scholar PubMed
68. Bannai, S. Exchange of cystine and glutamate across plasma membrane of human fibroblasts. J Biol Chem 1986;261:2256–63. https://doi.org/10.1016/s0021-9258(17)35926-4.Search in Google Scholar
69. Dixon, SJ, Lemberg, KM, Lamprecht, MR, Skouta, R, Zaitsev, E, Gleason, C, et al.. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 2012;149:1060–72. https://doi.org/10.1016/j.cell.2012.03.042.Search in Google Scholar PubMed PubMed Central
70. Hidalgo-Gutiérrez, A, González-García, P, Díaz-Casado, ME, Barriocanal-Casado, E, López-Herrador, S, Quinzii, CM, et al.. Metabolic targets of coenzyme Q10 in mitochondria. Antioxidants 2021;10:520. https://doi.org/10.3390/antiox10040520.Search in Google Scholar PubMed PubMed Central
71. Bersuker, K, Hendricks, JM, Li, Z, Magtanong, L, Ford, B, Tang, PH, et al.. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 2019;575:688–92. https://doi.org/10.1038/s41586-019-1705-2.Search in Google Scholar PubMed PubMed Central
72. Mao, C, Liu, X, Zhang, Y, Lei, G, Yan, Y, Lee, H, et al.. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature 2021;593:586–90. https://doi.org/10.1038/s41586-021-03539-7.Search in Google Scholar PubMed PubMed Central
73. Bersuker, K, Hendricks, J, Lippincott-Schwartz, J. Measuring, visualizing, and manipulating the lipid mediators of ferroptosis. Biochemistry 2022;61:827–37.Search in Google Scholar
74. Doll, S, Freitas, FP, Shah, R, Aldrovandi, M, da Silva, MC, Ingold, I, et al.. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 2019;575:693–8. https://doi.org/10.1038/s41586-019-1707-0.Search in Google Scholar PubMed
75. Hu, Q, Wei, W, Wu, D, Huang, F, Li, M, Li, W, et al.. Blockade of GCH1/BH4 Axis activates ferritinophagy to mitigate the resistance of colorectal cancer to erastin-induced ferroptosis. Front Cell Dev Biol 2022;10:810327. https://doi.org/10.3389/fcell.2022.810327.Search in Google Scholar PubMed PubMed Central
76. Liu, Z, Kang, R, Yang, N, Pan, X, Yang, J, Yu, H, et al.. Tetrahydrobiopterin inhibitor-based antioxidant metabolic strategy for enhanced cancer ferroptosis-immunotherapy. J Colloid Interface Sci 2024;658:100–13. https://doi.org/10.1016/j.jcis.2023.12.042.Search in Google Scholar PubMed
77. Soula, M, Weber, RA, Zilka, O, Alwaseem, H, La, K, Yen, F, et al.. Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat Chem Biol 2020;16:1351–60. https://doi.org/10.1038/s41589-020-0613-y.Search in Google Scholar PubMed PubMed Central
78. Kraft, VAN, Bezjian, CT, Pfeiffer, S, Ringelstetter, L, Müller, C, Zandkarimi, F, et al.. GTP cyclohydrolase 1/tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Cent Sci 2020;6:41–53. https://doi.org/10.1021/acscentsci.9b01063.Search in Google Scholar PubMed PubMed Central
79. Florencio-Silva, R, Sasso, GR, Sasso-Cerri, E, Simões, MJ, Cerri, PS. Biology of bone tissue: structure, function, and factors that influence bone cells. BioMed Res Int 2015;2015:421746. https://doi.org/10.1155/2015/421746.Search in Google Scholar PubMed PubMed Central
80. Tang, Y, Su, S, Yu, R, Liao, C, Dong, Z, Jia, C, et al.. Unraveling ferroptosis in osteogenic lineages: implications for dysregulated bone remodeling during periodontitis progression. Cell Death Dis 2024;10:195. https://doi.org/10.1038/s41420-024-01969-6.Search in Google Scholar PubMed PubMed Central
81. Lin, F, Chen, W, Zhou, J, Zhu, J, Yao, Q, Feng, B, et al.. Mesenchymal stem cells protect against ferroptosis via exosome-mediated stabilization of SLC7A11 in acute liver injury. Cell Death Dis 2022;13:271. https://doi.org/10.1038/s41419-022-04708-w.Search in Google Scholar PubMed PubMed Central
82. Shao, C, Chen, Y, Yang, T, Zhao, H, Li, D. Mesenchymal stem cell derived exosomes suppress neuronal cell ferroptosis via lncGm36569/miR-5627-5p/FSP1 axis in acute spinal cord injury. Stem Cell Rev Rep 2022;18:1127–42. https://doi.org/10.1007/s12015-022-10327-x.Search in Google Scholar PubMed
83. Liu, J, Ren, Z, Yang, L, Zhu, L, li, Y, Bie, C, et al.. The NSUN5-FTH1/FTL pathway mediates ferroptosis in bone marrow-derived mesenchymal stem cells. Cell Death Dis 2022;8:99. https://doi.org/10.1038/s41420-022-00902-z.Search in Google Scholar PubMed PubMed Central
84. Ge, W, Jie, J, Yao, J, Li, W, Cheng, Y, Lu, W. Advanced glycation end products promote osteoporosis by inducing ferroptosis in osteoblasts. Mol Med Rep 2022;25. https://doi.org/10.3892/mmr.2022.12656.Search in Google Scholar PubMed PubMed Central
85. Ma, H, Wang, X, Zhang, W, Li, H, Zhao, W, Sun, J, et al.. Melatonin suppresses ferroptosis induced by high glucose via activation of the Nrf2/HO-1 signaling pathway in type 2 diabetic osteoporosis. Oxid Med Cell Longev 2020;2020:9067610. https://doi.org/10.1155/2020/9067610.Search in Google Scholar PubMed PubMed Central
86. Wang, X, Ma, H, Sun, J, Zheng, T, Zhao, P, Li, H, et al.. Mitochondrial ferritin deficiency promotes osteoblastic ferroptosis via mitophagy in type 2 diabetic osteoporosis. Biol Trace Elem Res 2022;200:298–307. https://doi.org/10.1007/s12011-021-02627-z.Search in Google Scholar PubMed
87. Yang, R, Xu, W, Zheng, H, Zheng, X, Li, B, Jiang, L, et al.. Exosomes derived from vascular endothelial cells antagonize glucocorticoid-induced osteoporosis by inhibiting ferritinophagy with resultant limited ferroptosis of osteoblasts. J Cell Physiol 2021;236:6691–705. https://doi.org/10.1002/jcp.30331.Search in Google Scholar PubMed
88. Lu, J, Yang, J, Zheng, Y, Chen, X, Fang, S. Extracellular vesicles from endothelial progenitor cells prevent steroid-induced osteoporosis by suppressing the ferroptotic pathway in mouse osteoblasts based on bioinformatics evidence. Sci Rep 2019;9:16130. https://doi.org/10.1038/s41598-019-52513-x.Search in Google Scholar PubMed PubMed Central
89. Zhang, J, Zhang, L, Yao, G, Zhao, H, Wu, S. NRF2 is essential for iron-overload stimulated osteoclast differentiation through regulation of redox and iron homeostasis. Cell Biol Toxicol 2023;39:3305–21. https://doi.org/10.1007/s10565-023-09834-5.Search in Google Scholar PubMed
90. Yang, J, Dong, D, Luo, X, Zhou, J, Shang, P, Zhang, H. Iron overload-induced osteocyte apoptosis stimulates osteoclast differentiation through increasing osteocytic RANKL production in vitro. Calcif Tissue Int 2020;107:499–509. https://doi.org/10.1007/s00223-020-00735-x.Search in Google Scholar PubMed
91. Qu, X, Sun, Z, Wang, Y, Ong, HS. Zoledronic acid promotes osteoclasts ferroptosis by inhibiting FBXO9-mediated p53 ubiquitination and degradation. PeerJ 2021;9:e12510. https://doi.org/10.7717/peerj.12510.Search in Google Scholar PubMed PubMed Central
92. Ni, S, Yuan, Y, Qian, Z, Zhong, Z, Lv, T, Kuang, Y, et al.. Hypoxia inhibits RANKL-induced ferritinophagy and protects osteoclasts from ferroptosis. Free Radic Biol Med 2021;169:271–82. https://doi.org/10.1016/j.freeradbiomed.2021.04.027.Search in Google Scholar PubMed
93. Sun, K, Guo, Z, Hou, L, Xu, J, Du, T, Xu, T, et al.. Iron homeostasis in arthropathies: from pathogenesis to therapeutic potential. Ageing Res Rev 2021;72:101481. https://doi.org/10.1016/j.arr.2021.101481.Search in Google Scholar PubMed
94. Guo, Z, Lin, J, Sun, K, Guo, J, Yao, X, Wang, G, et al.. Deferoxamine alleviates osteoarthritis by inhibiting chondrocyte ferroptosis and activating the Nrf2 pathway. Front Pharmacol 2022;13:791376. https://doi.org/10.3389/fphar.2022.791376.Search in Google Scholar PubMed PubMed Central
95. Zhou, X, Zheng, Y, Sun, W, Zhang, Z, Liu, J, Yang, W, et al.. D-mannose alleviates osteoarthritis progression by inhibiting chondrocyte ferroptosis in a HIF-2α-dependent manner. Cell Prolif 2021;54:e13134. https://doi.org/10.1111/cpr.13134.Search in Google Scholar PubMed PubMed Central
96. Bao, J, Yan, Y, Zuo, D, Zhuo, Z, Sun, T, Lin, H, et al.. Iron metabolism and ferroptosis in diabetic bone loss: from mechanism to therapy. Front Nutr 2023;10:1178573. https://doi.org/10.3389/fnut.2023.1178573.Search in Google Scholar PubMed PubMed Central
97. Ward, DM, Kaplan, J. Ferroportin-mediated iron transport: expression and regulation. Biochim Biophys Acta 2012;1823:1426–33. https://doi.org/10.1016/j.bbamcr.2012.03.004.Search in Google Scholar PubMed PubMed Central
98. Yang, Y, Lin, Y, Wang, M, Yuan, K, Wang, Q, Mu, P, et al.. Targeting Ferroptosis suppresses osteocyte glucolipotoxicity and alleviates diabetic osteoporosis. Bone Res. 2022;10:26. https://doi.org/10.1038/s41413-022-00198-w.Search in Google Scholar PubMed PubMed Central
99. Rauner, M, Baschant, U, Roetto, A, Pellegrino, RM, Rother, S, Salbach-Hirsch, J, et al.. Transferrin receptor 2 controls bone mass and pathological bone formation via BMP and Wnt signaling. Nat Metab 2019;1:111–24. https://doi.org/10.1038/s42255-018-0005-8.Search in Google Scholar PubMed PubMed Central
100. Miao, Y, Chen, Y, Xue, F, Liu, K, Zhu, B, Gao, J, et al.. Contribution of ferroptosis and GPX4’s dual functions to osteoarthritis progression. EBioMedicine 2022;76:103847. https://doi.org/10.1016/j.ebiom.2022.103847.Search in Google Scholar PubMed PubMed Central
101. Xu, C, Ni, S, Xu, N, Yin, G, Yu, Y, Zhou, B, et al.. Theaflavin-3,3’-digallate inhibits erastin-induced chondrocytes ferroptosis via the NRF2/GPX4 signaling pathway in osteoarthritis. Oxid Med Cell Longev 2022;2022:3531995. https://doi.org/10.1155/2022/3531995.Search in Google Scholar PubMed PubMed Central
102. Zhang, S, Xu, J, Si, H, Wu, Y, Zhou, S, Shen, B. The role played by ferroptosis in osteoarthritis: evidence based on iron dyshomeostasis and lipid peroxidation. Antioxidants 2022;11:1668. https://doi.org/10.3390/antiox11091668.Search in Google Scholar PubMed PubMed Central
103. Liu, H, Deng, Z, Yu, B, Liu, H, Yang, Z, Zeng, A, et al.. Identification of SLC3A2 as a potential therapeutic target of osteoarthritis involved in ferroptosis by integrating bioinformatics, clinical factors and experiments. Cells 2022;11:3430. https://doi.org/10.3390/cells11213430.Search in Google Scholar PubMed PubMed Central
104. Yao, X, Sun, K, Yu, S, Luo, J, Guo, J, Lin, J, et al.. Chondrocyte ferroptosis contributes to the progression of osteoarthritis. J Orthop Translat 2021;27:33–43. https://doi.org/10.1016/j.jot.2020.09.006.Search in Google Scholar PubMed PubMed Central
105. Yan, J, Feng, G, Ma, L, Chen, Z, Jin, Q. Metformin alleviates osteoarthritis in mice by inhibiting chondrocyte ferroptosis and improving subchondral osteosclerosis and angiogenesis. J Orthop Surg Res 2022;17:333. https://doi.org/10.1186/s13018-022-03225-y.Search in Google Scholar PubMed PubMed Central
106. Ma, S, Dubin, AE, Zhang, Y, Mousavi, SAR, Wang, Y, Coombs, AM, et al.. A role of Piezo1 in iron metabolism in mice and humans. Cell 2021;184:969–82.e13. https://doi.org/10.1016/j.cell.2021.01.024.Search in Google Scholar PubMed PubMed Central
107. Wang, S, Li, W, Zhang, P, Wang, Z, Ma, X, Liu, C, et al.. Mechanical overloading induces GPX4-regulated chondrocyte ferroptosis in osteoarthritis via Piezo1 channel facilitated calcium influx. J Adv Res 2022;41:63–75. https://doi.org/10.1016/j.jare.2022.01.004.Search in Google Scholar PubMed PubMed Central
108. Smolen, JS, Aletaha, D, McInnes, IB. Rheumatoid arthritis. Lancet 2016;388:2023–38. https://doi.org/10.1016/s0140-6736(16)30173-8.Search in Google Scholar
109. Ogilvie-Harris, DJ, Fornaiser, VL. Synovial iron deposition in osteoarthritis and rheumatoid arthritis. J Rheumatol 1980;7:30–6.Search in Google Scholar
110. Bustamante, MF, Garcia-Carbonell, R, Whisenant, KD, Guma, M. Fibroblast-like synoviocyte metabolism in the pathogenesis of rheumatoid arthritis. Arthritis Res Ther 2017;19:110. https://doi.org/10.1186/s13075-017-1303-3.Search in Google Scholar PubMed PubMed Central
111. Chang, S, Tang, M, Zhang, B, Xiang, D, Li, F. Ferroptosis in inflammatory arthritis: a promising future. Front Immunol 2022;13:955069. https://doi.org/10.3389/fimmu.2022.955069.Search in Google Scholar PubMed PubMed Central
112. Liu, P, Wang, W, Li, Z, Li, Y, Yu, X, Tu, J, et al.. Ferroptosis: a new regulatory mechanism in osteoporosis. Oxid Med Cell Longev 2022;2022:2634431. https://doi.org/10.1155/2022/2634431.Search in Google Scholar PubMed PubMed Central
113. Ling, H, Li, M, Yang, C, Sun, S, Zhang, W, Zhao, L, et al.. Glycine increased ferroptosis via SAM-mediated GPX4 promoter methylation in rheumatoid arthritis. Rheumatology 2022;61:4521–34. https://doi.org/10.1093/rheumatology/keac069.Search in Google Scholar PubMed
114. Wu, J, Feng, Z, Chen, L, Li, Y, Bian, H, Geng, J, et al.. TNF antagonist sensitizes synovial fibroblasts to ferroptotic cell death in collagen-induced arthritis mouse models. Nat Commun 2022;13:676. https://doi.org/10.1038/s41467-021-27948-4.Search in Google Scholar PubMed PubMed Central
115. Cheng, Q, Chen, M, Liu, M, Chen, X, Zhu, L, Xu, J, et al.. Semaphorin 5A suppresses ferroptosis through activation of PI3K-Akt-mTOR signaling in rheumatoid arthritis. Cell Death Dis 2022;13:608. https://doi.org/10.1038/s41419-022-05065-4.Search in Google Scholar PubMed PubMed Central
116. Ooi, M, Hibbs, S, Chen, FE. The safety of modern intravenous iron infusions in patients with rheumatoid arthritis - a review of the literature. Hematology 2020;25:108–11. https://doi.org/10.1080/16078454.2020.1730557.Search in Google Scholar PubMed
117. Kong, R, Ji, L, Pang, Y, Zhao, D, Gao, J. Exosomes from osteoarthritic fibroblast-like synoviocytes promote cartilage ferroptosis and damage via delivering microRNA-19b-3p to target SLC7A11 in osteoarthritis. Front Immunol 2023;14:1181156. https://doi.org/10.3389/fimmu.2023.1181156.Search in Google Scholar PubMed PubMed Central
118. Fan, Y, Li, Y, Fu, X, Peng, J, Chen, Y, Chen, T, et al.. Identification of potential ferroptosis key genes and immune infiltration in rheumatoid arthritis by integrated bioinformatics analysis. Heliyon 2023;9:e21167. https://doi.org/10.1016/j.heliyon.2023.e21167.Search in Google Scholar PubMed PubMed Central
119. Nygaard, G, Firestein, GS. Restoring synovial homeostasis in rheumatoid arthritis by targeting fibroblast-like synoviocytes. Nat Rev Rheumatol 2020;16:316–33. https://doi.org/10.1038/s41584-020-0413-5.Search in Google Scholar PubMed PubMed Central
120. Compston, JE, McClung, MR, Leslie, WD. Osteoporosis. Lancet 2019;393:364–76. https://doi.org/10.1016/s0140-6736(18)32112-3.Search in Google Scholar PubMed
121. Yamamoto, Y, Chiba, T, Dohmae, S, Higashi, K, Nakazawa, A. Osteoporosis medication after fracture in older adults: an administrative data analysis. Osteoporos Int 2021;32:1245–6. https://doi.org/10.1007/s00198-021-05973-9.Search in Google Scholar PubMed
122. Song, X, Xie, Y, Kang, R, Hou, W, Sun, X, Epperly, MW, et al.. FANCD2 protects against bone marrow injury from ferroptosis. Biochem Biophys Res Commun 2016;480:443–9. https://doi.org/10.1016/j.bbrc.2016.10.068.Search in Google Scholar PubMed PubMed Central
123. Miyamoto, T. Mechanism underlying post-menopausal osteoporosis: HIF1α is required for osteoclast activation by estrogen deficiency. Keio J Med 2015;64:44–7. https://doi.org/10.2302/kjm.2015-0003-re.Search in Google Scholar PubMed
124. Ma, J, Wang, A, Zhang, H, Liu, B, Geng, Y, Xu, Y, et al.. Iron overload induced osteocytes apoptosis and led to bone loss in hepcidin(-/-) mice through increasing sclerostin and RANKL/OPG. Bone 2022;164:116511. https://doi.org/10.1016/j.bone.2022.116511.Search in Google Scholar PubMed
125. Qiu, C, Cai, X, Zheng, Z, Ma, X, Zhang, G, Guo, Z, et al.. Differential proteomic analysis of fetal and geriatric lumbar nucleus pulposus: immunoinflammation and age-related intervertebral disc degeneration. BMC Muscoskel Disord 2020;21:339. https://doi.org/10.1186/s12891-020-03329-8.Search in Google Scholar PubMed PubMed Central
126. Shamji, MF, Setton, LA, Jarvis, W, So, S, Chen, J, Jing, L, et al.. Proinflammatory cytokine expression profile in degenerated and herniated human intervertebral disc tissues. Arthritis Rheum 2010;62:1974–82. https://doi.org/10.1002/art.27444.Search in Google Scholar PubMed PubMed Central
127. Yang, RZ, Xu, W, Huang, WY, Zheng, XF, Li, B, Jiang, LS, et al.. Involvement of oxidative stress-induced annulus fibrosus cell and nucleus pulposus cell ferroptosis in intervertebral disc degeneration pathogenesis. J Cell Physiol 2021;236:2725–39. https://doi.org/10.1002/jcp.30039.Search in Google Scholar PubMed PubMed Central
128. Wang, W, Xie, Z, Lin, X, Ren, J, Liu, X, Chen, F, et al.. Iron overload promotes intervertebral disc degeneration via inducing oxidative stress and ferroptosis in endplate chondrocytes. Free Radic Biol Med 2022;190:234–46. https://doi.org/10.1016/j.freeradbiomed.2022.08.018.Search in Google Scholar PubMed
129. Shan, L, Wang, X, Liu, J, Cai, P, Gao, H, Lu, Y, et al.. Increased hemoglobin and heme in MALDI-TOF MS analysis induce ferroptosis and promote degeneration of herniated human nucleus pulposus. Mol Med 2021;27:103. https://doi.org/10.1186/s10020-021-00368-2.Search in Google Scholar PubMed PubMed Central
130. Zhang, Y, Han, S, Kong, M, Tu, Q, Zhang, L, Ma, X. Single-cell RNA-seq analysis identifies unique chondrocyte subsets and reveals involvement of ferroptosis in human intervertebral disc degeneration. Osteoarthr Cartil 2021;29:1324–34.10.1016/j.joca.2021.06.010Search in Google Scholar PubMed
131. Lu, S, Song, Y, Luo, R, Li, S, Li, G, Wang, K, et al.. Ferroportin-dependent iron homeostasis protects against oxidative stress-induced nucleus pulposus cell ferroptosis and ameliorates intervertebral disc degeneration in vivo. Oxid Med Cell Longev 2021;2021:6670497. https://doi.org/10.1155/2021/6670497.Search in Google Scholar PubMed PubMed Central
132. Bin, S, Wang, Y, Zhang, L, Jinhua, Z, Rui, G, Xiang, Z. Targeting miR-10a-5p/IL-6R axis for reducing IL-6-induced cartilage cell ferroptosis. Exp Mol Pathol 2021;118:104570. https://doi.org/10.1016/j.yexmp.2020.104570.Search in Google Scholar PubMed
133. Hao, J, Bei, J, Li, Z, Han, M, Ma, B, Ma, P, et al.. Qing’E pill inhibits osteoblast ferroptosis via Atm serine/threonine kinase (Atm) and the Pi3k/Akt pathway in primary osteoporosis. Front Pharmacol 2022;13:902102. https://doi.org/10.3389/fphar.2022.902102.Search in Google Scholar PubMed PubMed Central
134. Zeng, HB, Dong, LQ, Xu, C, Zhao, XH, Wu, LG. Artesunate promotes osteoblast differentiation through Mir-34a/Dkk1 Axis. Acta Histochem 2020;122:151601. https://doi.org/10.1016/j.acthis.2020.151601.Search in Google Scholar PubMed
135. Zhao, Y, Du, Y, Gao, Y, Xu, Z, Zhao, D, Yang, M. Atf3 regulates osteogenic function by mediating osteoblast Ferroptosis in type 2 diabetic osteoporosis. Dis Markers 2022;2022:9872243. https://doi.org/10.1155/2022/9872243.Search in Google Scholar PubMed PubMed Central
136. Chen, YC, Hsiao, JK, Liu, HM, Lai, IY, Yao, M, Hsu, SC, et al.. The inhibitory effect of superparamagnetic iron oxide nanoparticle (ferucarbotran) on osteogenic differentiation and its signaling mechanism in human mesenchymal stem cells. Toxicol Appl Pharmacol 2010;245:272–9. https://doi.org/10.1016/j.taap.2010.03.011.Search in Google Scholar PubMed
137. Luo, H, Zhang, R. Icariin enhances cell survival in lipopolysaccharide-induced synoviocytes by suppressing ferroptosis via the Xc-/GPX4 axis. Exp Ther Med 2021;21:72. https://doi.org/10.3892/etm.2020.9504.Search in Google Scholar PubMed PubMed Central
138. Mo, Z, Xu, P, Li, H. Stigmasterol alleviates interleukin-1beta-induced chondrocyte injury by down-regulating sterol regulatory element binding transcription factor 2 to regulate ferroptosis. Bioengineered 2021;12:9332–40. https://doi.org/10.1080/21655979.2021.2000742.Search in Google Scholar PubMed PubMed Central
139. Jing, X, Wang, Z, Lin, Y, Zhang, W, Liu, X, Liu, Q, et al.. Calcium chelator BAPTA-AM protects against iron overload-induced chondrocyte mitochondrial dysfunction and cartilage degeneration. Int J Mol Med 2021;48:196. https://doi.org/10.3892/ijmm.2021.5029.Search in Google Scholar PubMed PubMed Central
140. Fahmi, H, Di Battista, JA, Pelletier, JP, Cheung, H, Fernandes, J, Martel-Pelletier, J. Peroxisome proliferator-activated receptor gamma activators inhibit MMP-1 production in human synovial fibroblasts likely by reducing the binding of the activator protein 1. Osteoarthr Cartil 2002;10:100–8. https://doi.org/10.1053/joca.2001.0485.Search in Google Scholar PubMed
141. Kim, DH, Kim, DH, Heck, BE, Shaffer, M, Yoo, KH, Hur, J. PPAR-delta agonist affects adipo-chondrogenic differentiation of human mesenchymal stem cells through the expression of PPAR-gamma. Regen Ther 2020;15:103–11.10.1016/j.reth.2020.07.003Search in Google Scholar PubMed PubMed Central
142. Chen, Y, Zhao, W, Hu, A, Lin, S, Chen, P, Yang, B, et al.. Type 2 diabetic mellitus related osteoporosis: focusing on ferroptosis. J Transl Med 2024;22:409. https://doi.org/10.1186/s12967-024-05191-x.Search in Google Scholar PubMed PubMed Central
143. Gao, Z, Chen, Z, Xiong, Z, Liu, X. Ferroptosis - a new target of osteoporosis. Exp Gerontol 2022;165:111836. https://doi.org/10.1016/j.exger.2022.111836.Search in Google Scholar PubMed
144. Fan, C, Chu, G, Yu, Z, Ji, Z, Kong, F, Yao, L, et al.. The role of ferroptosis in intervertebral disc degeneration. Front Cell Dev Biol 2023;11:1219840. https://doi.org/10.3389/fcell.2023.1219840.Search in Google Scholar PubMed PubMed Central
145. Zhang, H, Yang, F, Cao, Z, Xu, Y, Wang, M. The influence of iron on bone metabolism disorders. Osteoporos Int 2024;35:243–53. https://doi.org/10.1007/s00198-023-06937-x.Search in Google Scholar PubMed
146. Jing, X, Du, T, Li, T, Yang, X, Wang, G, Liu, X, et al.. The detrimental effect of iron on OA chondrocytes: importance of pro-inflammatory cytokines induced iron influx and oxidative stress. J Cell Mol Med 2021;25:5671–80. https://doi.org/10.1111/jcmm.16581.Search in Google Scholar PubMed PubMed Central
147. Karim, A, Bajbouj, K, Qaisar, R, Hall, AC, Hamad, M. The role of disrupted iron homeostasis in the development and progression of arthropathy. J Orthop Res 2022;40:1243–50. https://doi.org/10.1002/jor.25323.Search in Google Scholar PubMed
148. Iantomasi, T, Romagnoli, C, Palmini, G, Donati, S, Falsetti, I, Miglietta, F, et al.. Oxidative stress and inflammation in osteoporosis: molecular mechanisms involved and the relationship with microRNAs. Int J Mol Sci 2023;24:3772. https://doi.org/10.3390/ijms24043772.Search in Google Scholar PubMed PubMed Central
149. Poulet, B, Beier, F. Targeting oxidative stress to reduce osteoarthritis. Arthritis Res Ther 2016;18:32. https://doi.org/10.1186/s13075-015-0908-7.Search in Google Scholar PubMed PubMed Central
150. Cao, S, Wei, Y, Yue, Y, Chen, Y, Liao, S, Li, A, et al.. Targeting ferroptosis unveils a new era for traditional Chinese medicine: a scientific metrology study. Front Pharmacol 2024;15:1366852. https://doi.org/10.3389/fphar.2024.1366852.Search in Google Scholar PubMed PubMed Central
151. Li, J, Cao, F, Yin, HL, Huang, Z, Lin, Z, Mao, N, et al.. Ferroptosis: past, present and future. Cell Death Dis 2020;11:88. https://doi.org/10.1038/s41419-020-2298-2.Search in Google Scholar PubMed PubMed Central
152. Zeng, W, Long, X, Liu, PS, Xie, X. The interplay of oncogenic signaling, oxidative stress and ferroptosis in cancer. Int J Cancer 2023;153:918–31. https://doi.org/10.1002/ijc.34486.Search in Google Scholar PubMed
153. Jin, S, Liu, PS, Zheng, D, Xie, X. The interplay of miRNAs and ferroptosis in diseases related to iron overload. Apoptosis 2024;29:45–65. https://doi.org/10.1007/s10495-023-01890-w.Search in Google Scholar PubMed
154. Tang, Y, Su, S, Yu, R, Liao, C, Dong, Z, Jia, C, et al.. Unraveling ferroptosis in osteogenic lineages: implications for dysregulated bone remodeling during periodontitis progression. Cell Death Dis 2024;10:195. https://doi.org/10.1038/s41420-024-01969-6.Search in Google Scholar PubMed PubMed Central
155. Hassannia, B, Vandenabeele, P, Vanden Berghe, T. Targeting ferroptosis to iron out cancer. Cancer Cell 2019;35:830–49. https://doi.org/10.1016/j.ccell.2019.04.002.Search in Google Scholar PubMed
156. Zhang, Y, Qu, Z, Zhao, Y, Zhang, B, Gong, Y, Wang, X, et al.. The therapeutic effect of natural compounds on osteoporosis through ferroptosis. Curr Med Chem 2024;31:2629–48. https://doi.org/10.2174/0109298673258420230919103405.Search in Google Scholar PubMed
© 2024 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Frontmatter
- Review
- Targeting oxidative stress, iron overload and ferroptosis in bone-degenerative conditions
- Research Articles
- Assessing medical biochemistry professionals’ knowledge, attitudes, and behaviors regarding green and sustainable medical laboratory practices in Türkiye
- The efficacy of high pressure liquid chromatography (HPLC) in detecting congenital glycosylation disorders (CDG)
- Atypical cells parameter in sysmex UN automated urine analyzer: a single center study
- The frequency of single specific immunoglobulin E and allergen mixes with a MAST (multiple-antigen simultaneous test) technique
- Differences in second trimester risk estimates for trisomy 21 between Maglumi X3/Preaccu and Immulite/Prisca systems
- Comparison of classical and flowcytometric osmotic fragility and flowcytometric eosin-5-maleimide binding tests in diagnosis of hereditary spherocytosis
- Casticin inhibits the hedgehog signaling and leads to apoptosis in AML stem-like KG1a and mature KG1 cells
- Trimethylamine N-oxide, S-equol, and indoxyl sulfate inflammatory microbiota players in ocular Behçet’s disease
- Genomic profiling of interferon signaling pathway gene mutations in type 2 diabetic individuals with COVID-19
- CDR1as/miR-7-5p/IGF1R axis contributes to the suppression of cell viability in prostate cancer
- Role of interferon regulatory factors in predicting the prognosis of Crimean-Congo hemorrhagic fever
- The significance of taurine for patients with Crimean-Congo hemorrhagic fever and COVID-19 diseases: a cross-sectional study
- Gene mining, recombinant expression and enzymatic characterization of N-acetylglucosamine deacetylase
- Ethanol inhibited growth hormone receptor-mediated endocytosis in primary mouse hepatocytes
- Gypsophila eriocalyx roots inhibit proliferation, migration, and TGF-β signaling in melanoma cells
- The role of kynurenine and kynurenine metabolites in psoriasis
- Tobacco induces abnormal metabolism of tryptophan via the kynurenine pathway
- Effect of vitamin D and omega-3 on the expression of endoplasmic reticulum-associated protein degradation and autophagic proteins in rat brain
Articles in the same Issue
- Frontmatter
- Review
- Targeting oxidative stress, iron overload and ferroptosis in bone-degenerative conditions
- Research Articles
- Assessing medical biochemistry professionals’ knowledge, attitudes, and behaviors regarding green and sustainable medical laboratory practices in Türkiye
- The efficacy of high pressure liquid chromatography (HPLC) in detecting congenital glycosylation disorders (CDG)
- Atypical cells parameter in sysmex UN automated urine analyzer: a single center study
- The frequency of single specific immunoglobulin E and allergen mixes with a MAST (multiple-antigen simultaneous test) technique
- Differences in second trimester risk estimates for trisomy 21 between Maglumi X3/Preaccu and Immulite/Prisca systems
- Comparison of classical and flowcytometric osmotic fragility and flowcytometric eosin-5-maleimide binding tests in diagnosis of hereditary spherocytosis
- Casticin inhibits the hedgehog signaling and leads to apoptosis in AML stem-like KG1a and mature KG1 cells
- Trimethylamine N-oxide, S-equol, and indoxyl sulfate inflammatory microbiota players in ocular Behçet’s disease
- Genomic profiling of interferon signaling pathway gene mutations in type 2 diabetic individuals with COVID-19
- CDR1as/miR-7-5p/IGF1R axis contributes to the suppression of cell viability in prostate cancer
- Role of interferon regulatory factors in predicting the prognosis of Crimean-Congo hemorrhagic fever
- The significance of taurine for patients with Crimean-Congo hemorrhagic fever and COVID-19 diseases: a cross-sectional study
- Gene mining, recombinant expression and enzymatic characterization of N-acetylglucosamine deacetylase
- Ethanol inhibited growth hormone receptor-mediated endocytosis in primary mouse hepatocytes
- Gypsophila eriocalyx roots inhibit proliferation, migration, and TGF-β signaling in melanoma cells
- The role of kynurenine and kynurenine metabolites in psoriasis
- Tobacco induces abnormal metabolism of tryptophan via the kynurenine pathway
- Effect of vitamin D and omega-3 on the expression of endoplasmic reticulum-associated protein degradation and autophagic proteins in rat brain