Abstract Peri-prosthesis osteolysis (PPO) represents the most severe complication of total joint arthroplasty (TJA) surgery and imposes the primary cause of prosthesis failure and subsequent revision surgery. Antiresorptive therapies are usually prescribed to treat PPO, especially for elderly people. Nevertheless, the efficacy of anti-osteoporotic medications remains constrained. Recent therapeutic strategies to promote periprosthetic osseointegration by restoring osteoblast function are considered more effective approaches. However, the precise mechanism underlying the inhibition of osteogenesis triggered by wear particles remains enigmatic. Herein, we demonstrate that wear particles inhibit osteoblast function by inducing ferroptosis to sabotage extracellular mineralization and arouse periprosthetic osteolysis. The suppression of ferroptosis could significantly rescue osteogenesis thus alleviating PPO. Furthermore, Glutathione Peroxidase 4 (GPX4) has been identified as a key target in regulating osteoblastic ferroptosis. By utilizing virtual screening techniques, we have successfully conducted a comprehensive screening of a natural compound known as Urolithin A (UA), which exhibits remarkable inhibition of osteoblastic ferroptosis while simultaneously promoting the process of osteogenesis through its precise targeting mechanism on GPX4. Meanwhile, UA improves the osteolytic conditions significantly in vivo even when the adjunction of titanium (Ti) nanoparticles. This strategy has great potential in treating peri-prosthesis osteolysis and potentially broadens the scope of clinical therapy. Graphical Abstract [46]graphic file with name 12951_2024_3049_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12951-024-03049-4. Keywords: Peri-prosthesis osteolysis, Titanium nanoparticles, Osteoblastogenesis, Ferroptosis, GPX4, Urolithin A Introduction Total joint arthroplasty (TJA) is a surgical strategy widely used to treat degenerative, post-traumatic severe arthritis of the hip and knee [[47]1]. However, the wear particle-induced aseptic loosening around the prosthesis and subsequent osteolysis can lead to TJA failure and surgical revision [[48]2, [49]3]. Nanoparticles including titanium(Ti), chromium and polyethylene generated by prosthetic wear accumulate at the bone-implant interface and are engulfed by immune cells to induce an inflammatory response, thus leading to the further promotion of osteoclastic bone resorption and inhibition of osteoblast-mediated extracellular matrix mineralization as well as the eventual occurrence of PPO [[50]4]. Based on the underlying pathological mechanisms of PPO, endeavors have been undertaken to employ bone resorption inhibitors for the treatment of PPO [[51]5]. Despite some reduction in osteoclast activation and bone resorption, the outcomes remain unsatisfactory with frequent occurrence of side effects. Targeting osteoblastic function could alleviate the pathological process of PPO and extend the life of a prosthesis by improving the specific mechanism due to the direct detrimental impact of wear particles on osteoblasts [[52]6]. As a consequence, it is necessary to further explore the mechanism by which wear particles affect osteoblasts and extracellular matrix mineralization for the development of novel and effective therapies. Ferroptosis, a novel form of the pathway that regulates cell death, is characterized by iron-mediated accumulation of lipid peroxidation [[53]7]. Previous studies have shown that ferroptosis in osteoblasts could be induced by a variety of pathological conditions, which is manifested by a decrease in the osteogenic differentiation of mesenchymal stem cells, as well as the decreased cell activity of mature osteoblasts and the damaged function of extracellular matrix mineralization [[54]8, [55]9]. As a result of downregulation of Runt-related transcription factor 2 (Runx2) expression, ferroptosis inhibits osteogenic commitment and differentiation in mesenchymal stem cells and pre-osteoblasts, leading to a decrease in alkaline phosphatase and osteocalcin [[56]10]. Ferroptosis could also suppress osteoblast growth and impair osteoblast differentiation by downregulating PI3K/AKT/FOX3a/DUSP14 signaling pathway [[57]11, [58]12]. Moreover, Xu et al. discovered that the presence of CoCrMo nanoparticles facilitated osteoblastic ferroptosis, thereby exacerbating the pathological progression of aseptic loosening [[59]13]. However, the underlying mechanisms and optimal targets for intervention warrant further intricate investigation. Recent studies have clearly demonstrated the significant role of System Xc^−/GSH/GPX4 in regulating ferroptosis. GPX4 utilizes reduced glutathione (GSH) to efficiently convert phospholipid hydroperoxides into lipid alcohols, effectively preventing ferroptosis [[60]14]. Inhibiting the activity of system Xc^− (cystine/glutamate transporter) and GPX4 induces ferroptosis, highlighting the crucial role of GPX4 in regulating this process [[61]15]. Additionally, studies have shown that the NRF2/System Xc^−/GPX4 pathway can be activated to counteract ferroptosis and promote bone regeneration when there is an excess of reactive oxygen species (ROS) and lipid peroxidation [[62]16, [63]17]. However, the role of GPX4 in PPO and related mechanisms remain unclear. PPO is characterized by elevated levels of ROS and reduced osteogenic function, suggesting that GPX4 and ferroptosis may play a critical role in related pathological processes. Therefore, targeting GPX4 may be a potential therapeutic strategy for PPO. In the current investigation, we found that osteoblastic ferroptosis constituted a pivotal factor contributing to the pathogenesis of PPO. The induction of osteoblastic ferroptosis by Ti nanoparticles and the subsequent reduction in osteogenic function observed in clinical PPO samples, an osteolysis mouse model, and during osteoblast differentiation were all correlated with a significant decrease in GPX4 expression. The specific inhibitor of ferroptosis, known as ferrostatin-1 (Fer-1), exerts a suppressive effect on osteoblast susceptibility to ferroptosis by elevating the expression of GPX4, thereby rescuing their osteogenic function and effectively halting the progression of peri-prosthesis osteolysis. Taking into account the instability of Fer-1 and its implications for further clinical application, as well as the crucial anti-ferroptosis role of GPX4, we identified a small molecule urolithin A (UA) through virtual screening and molecular docking targeting GPX4, which possesses stable physicochemical properties, minimal toxicity, antioxidant and rejuvenating properties, significantly improving osteogenic function [[64]18, [65]19]. Mechanistically, the protective effects of UA against osteoblastic ferroptosis rely on the activation of GPX4 to mitigate Ti nanoparticle-induced osteoblastic ferroptosis. In vivo, UA exerts a therapeutic impact on peri-prosthesis osteolysis induced by Ti nanoparticles through its potent stimulation of bone formation. In conclusion, this study provides a new insight into Ti nanoparticle-induced peri-prosthesis osteolysis and a novel therapeutic approach for PPO. Method Drugs and reagents The acquisition of Ti nanoparticles was made from the Nanjing Emperor Nano Materials Company. Fer-1, UA, N-acetylcysteine (NAC)and Erastin were available from MedChem Express (MCE, USA). Minimum essential medium α (α-MEM, for culturing bone marrow-derived mesenchymal stem cells (BMSCs) and MC3T3-E1 cells). Fetal bovine serum (FBS) was purchased from VivaCell (China). Phosphate buffer saline (PBS) was purchased from Solarbio (China). RIPA lysis buffer and bovine serum albumin (BSA) were procured from Beyotime (China) for experimental use. Cell culture and viability assay The MC3T3-E1 cell line was procured from the Cell Bank affiliated with the Chinese Academy of Sciences. BMSCs were collected from 6-week-old SD rats, as previously described [[66]20]. The BMSCs and MC3T3-E1 cells were cultured in α-MEM supplemented with 10% FBS at a temperature of 37 °C under a CO[2] concentration of 5%. Osteogenesis of BMSCs or MC3T3-E1 cells was induced with Osteogenic medium (Sigma, USA). To mimic the osteolytic microenvironment, the cells were exposed to a medium supplemented with 5 µg/cm^2 or 10 µg/cm^2 Ti nanoparticles, which was refreshed every three days. The cells were subsequently exposed to 5µmol/L and 10 µmol/L of UA or 5µmol/L Fer-1 for treatment. Cell viability in MC3T3-E1 cells and BMSCs survivability were tested using CCK8 assay kit (Yeasen, China) according to the manufacture’s instruction. BMSCs and MC3T3-E1 cells were seeded at a density of 4 × 10^3 cells per well in 96-well plates. In order to evaluate the cytotoxicity of Ti nanoparticles, Fer-1, UA, and Erastin on these cells, different concentrations were added and incubated. The MC3T3-E1 cells and BMSCs inhibition were plotted using GraphPad Prism 8. Alkaline phosphatase (ALP) assay BMSCs were seeded at a density of 1 × 10^4 cells per well in 24-well plates and cultured in osteogenic medium for a duration of 7 days with indicated treatment. The cells were washed three times with PBS prior to being fixed in 4% paraformaldehyde at a temperature of 4°C for a duration of 20 min. After undergoing three times of PBS washing, the cells were subjected to incubation in a staining solution comprising 5-bromo-4-chloro-3-indolyl-phosphate/nitro blue tetrazolium (BCIP/NBT, Beyotime, China) for a duration of 30 min at room temperature under light-free conditions. Subsequently, the absorbance was measured at 530 nm. Alizarin red S (ARS) staining A mineralization evaluation was conducted using the alizarin red staining assay. 1 × 10^4 BMSCs were seeded in each well in a 24-well plate and subjected to a 21-day culture in an osteogenic medium. After undergoing three rounds of PBS washing, BMSCs were meticulously fixed in a 4% polyformaldehyde solution for a duration of 15 min. Subsequently, the cells were delicately incubated with alizarin red solution (Yeasen, China) at room temperature for an additional period of 15 min. Then BMSCs were washed 3 times with PBS before photographing under a microscope. The calcium nodules were dissolved in a solution containing 10% monohydrate of 1-Hexadecylpyridinium chloride, resulting in the dissolution of the alizarin red complex. Absorbance was recorded at 600 nm and analyzed using Image J software (National Institutes of Health, USA). Total RNA isolation and real-time PCR Total RNA was isolated from cells and tissues using TRIzol reagent (Beyotime, China) and NanoDrop (Thermo Fisher Scientific, USA) was utilized for RNA concentration measurement. Then, total RNA reverse transcribed with PrimeScriptTM RT Master Mix (Yeasen, China). Real-time PCR was performed in a CFX96™ thermal cycler (Bio-Rad Laboratories). The 2^−ΔΔCq formula was performed to quantify the gene expression. Human and Mouse GAPDH gene was used as endogenous control for sample normalization. Primers are listed in Table [67]S2 and [68]S3. Western blotting Proteins in samples were obtained by lysing with RIPA buffer (Beyotime, China). The BCA protein assay (Beyotime, China) was utilized to determine the protein concentrations. Afterwards, a careful transfer of the proteins onto a polyvinylidene fluoride membrane was conducted. The membranes were sealed with TBS containing 5% skimmed milk (Yeasen, China) for 1 h and incubated in primary antibody solution at 4 °C overnight. The following primary antibodies were put into use: COL1-a1 (ab260043, 1:1000), Runx2 (ab23981, 1:1000), Osterix (ab209484, 1:1000), GPX4 (ab125066, 1:1000), SLC7A11(ab307601, 1:1000), COX2 (ab1798001:1000), 4HNE (ab46545, 1:1000) all obtained from Abcam. After incubating with HRP-conjugated secondary antibodies (Beyotime, China) for 1 h at ambient temperature. Then, the ECL kit was utilized to observe the membranes. Protein quantification was performed using Image J. RNA sequencing Whole-transcriptome sequencing was performed using MC3T3-E1 cells induced by Ti nanoparticles (10 µg/cm^2) in osteogenic medium for 72 h and Relevant RNA was acquired using the conventional TRIzol extraction technique. Samples were sequenced using Illumina HiSeq 4000 instrument. We conducted comprehensive Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses to delve deeper into the subject matter. To further enhance our investigation, we employed OmicStudio tools for gene set enrichment analysis (GSEA) and bioinformatics analysis. Transmission electron microscope Transmission Electron Microscopy (TEM) was employed to examine changes in mitochondrial morphology. After the specified treatment, cells were subjected to fixation using a 2.5% solution of glutaraldehyde (Electron Microscope Grade) (Solarbio, China) for a duration of 16 h at a temperature of 4 °C. After fixation, the cells underwent dehydration, embedding, sectioning, and staining in a meticulous sequence. Finally, the TEM samples were elegantly visualized utilizing the capabilities of a state-of-the-art Hitachi Model H-7650 transmission electron microscope. Ferroptosis-related detection Mitochondrial membrane potential detection Mitochondrial membrane potential was measured by fluorescence microscopy of cells using Mitochondrial Membrane Potential Assay Kit with TMRE (Beyotime, China). Briefly, BMSCs were seeded in a 24-well plate at a density of 2 × 10^4 per well, cultured overnight, and treated as described above. The cells were then incubated with 1 TMRE staining solution for 30 min in the dark at 37 °C and analyzed with a fluorescent microscope (Carl Zeiss, German). ROS analysis To measure the ROS production, we used a 2,7-dichloro-dihydro-fluorescein diacetate (DCFH-DA) staining kit (Beyotime, China). Essentially, MC3T3-E1 cells were incubated for 30 min with DCFH-DA in FBS-free media in different groups in the dark for 20 min at 37 °C. The cells were meticulously fixed and captured through the lens of a fluorescence microscope, courtesy of Carl Zeiss (Germany). Lipid peroxidation assay and GSH assays The assessment of cellular lipid peroxidation was conducted using the MDA assay kit (Beyotime, China) to measure the concentration of malondialdehyde (MDA). MC3T3-E1 cells were treated as described above. After indicated treatment, the cells were thoroughly rinsed with phosphate-buffered saline (PBS) and subsequently lysed using a lysis buffer solution. The supernatant was collected and utilized for the detection of MDA after centrifuge. The MDA content is carried out in strict accordance with the manufacturer’s guidelines. A microplate reader was utilized to assess the absorbance of cellular MDA at a wavelength of 532 nm. The relative concentration of GSH in cell lysates was quantified using a glutathione assay kit (Sigma, USA) following the manufacturer’s instructions. FerroOrange staining Intracellular Fe^2+ level was analyzed by FerroOrange staining. 1 × 10^4 BMSCs per well were cultured in 24-well plates. After undergoing the specified treatments, osteocytes were thoroughly rinsed with HBSS solution and subsequently exposed to 1 µM FerroOrange probes (DOJINDO, Japan) for a duration of 30 min. The resulting stained cells were then captured using fluorescence microscopy (Carl Zeiss, Germany). Iron measurement To evaluate the Ti nanoparticle-induced ferroptosis, cellular iron was detected using the Iron Assay Kit (Solarbio, China) according to the manufacturer’s instructions. Briefly, 4 × 10^5 MC3T3-E1 cells per well were cultured in 6-well plates. After the specified treatments, mineralized osteocytes were collected and lysed in a buffer specifically designed for iron analysis. The resulting cell lysate was then subjected to centrifugation at 8000 × g for a duration of 10 min. Furthermore, the liquid portion above the sediment was utilized to determine the concentration of iron present. The experimental protocol strictly adhered to the guidelines provided by the manufacturers. A microplate reader was employed to measure absorbance at a wavelength of 510 nm. Immunofluorescence staining BMSCs were seeded on coverslips in 24-well plates in α-MEM for 24 h and then cultured in osteogenic differentiation medium with different interventions. Cells were seeded in 24-well plates at a density of 2 × 10^4 per well. After undergoing various treatment conditions, the BMSCs were subsequently exposed to 4% paraformaldehyde (PFA, NCM Biotech, China) at ambient temperature for a duration spanning from 20 to 30 min. The cells were subsequently treated with 0.5% Triton X-100 for a duration of 15 min to induce permeabilization. After being obstructed by a 1% BSA solution enriched with bovine serum albumin for a duration of 60 min, the cells were subjected to an overnight incubation at a chilly temperature of 4°C in the presence of the primary antibody. The OCN dilution ratio used for immunofluorescence staining was 1:400 (abcam, USA). Subsequently, the cells underwent three washes lasting for 10 min each using PBS. To facilitate visualization, a green-fluorescent Alexa Fluor 488 rabbit anti-mouse IgG (abcam, USA) (manufactured by Invitrogen) was utilized. After additional washing with PBS and subsequent staining with DAPI for a duration of 10 min, the cells were observed using a fluorescence microscope produced by Carl Zeiss (Germany). Cellular thermal shift assay (CETSA) As previously mentioned, the interaction between Urolithin A and GPX4 was investigated using CETSA [[69]21]. For the CETSA, the cell lysate was treated with DMSO or Urolithin A (10 µM) on a shaker at 4 °C for 3 h. The mixtures were then aliquoted into separate tubes and heated for 3 min at various temperatures. Following cooling, the cell lysates were centrifuged at 12,000 rpm for 15 min at 4 °C, and the supernatants were collected for subsequent Western blot analysis. Scanning electron microscope The morphology of Ti nanoparticles was observed with a FEI Quanta 600 field-emission scanning electron microscope. The acceleration voltage of 5 kV was used for the imaging. Samples for scanning electron microscopy were prepared from Ti nanoparticles powders after drying overnight and gold plating. The particle size distribution and average particle size of Ti nanoparticles were counted using image J. Bone tissue from clinical samples Bone tissues were procured from patients who underwent Total hip arthroplasty (THA). One group presented with developmental dysplasia of the hip (DDH) and underwent primary total joint arthroplasty (PTJA). The other group who underwent revised total joint arthroplasty (RTJA) for aseptic loosening. All patients underwent surgeries at the First Affiliated Hospital of Soochow University (Table [70]S1). This study has received approval from the esteemed Ethics Committee of the First Affiliated Hospital of Soochow University (2023107). Animals and osteolysis induced by wear particles Ti nanoparticle-induced mice calvarial osteolysis were established according to the literature and were generated as later described [[71]22]. All animal experiments were approved by the Ethics Committee of Soochow University (SUDA20221009A06). According to the random number table, the 8-week-old female C57BL/6 mice were randomly divided into five groups: Control group, Ti group (surgery), Fer-1 group (Ti + 0.1 mg/kg Fer-1), Low-UA group (Ti + 10 mg/kg UA), and High-UA group (Ti + 20 mg/kg UA). Doses of UA and Fer-1 have no physiological toxicity in vivo [[72]23, [73]24]. The size of Ti nanoparticles ranged from 25.39 to 121.67 nm (Fig [74]S1A and B). After surgical anesthesia, the mouse head was disinfected with 75% ethanol and covered with a sterile towel. A sterile scalpel was used to cut the skin of the mouse cranial roof to expose the cranium and peel the fascia of the cranium surface. According to previous studies, 40 mg of Ti nanoparticles (40 µL) were meticulously dispersed onto the calvariae surfaces, followed by the delicate application of sutures [[75]25]. Penicillin was administered 3 days after surgery to prevent infection. After surgery, all C57BL/6 mice were fed for 2 weeks in a specific, asymptomatic environment at room temperature and conventional humidity. The Fer-1 group received a fixed dose of Fer-1 by intraperitoneal injection daily after surgery. The UA group received a fixed dose of UA by gavage daily after surgery. The control group received physiological saline. All mice were sacrificed 14 days after the surgical procedure, and their crania were collected and then preserved in a solution containing 4% paraformaldehyde. Micro-computed tomography (micro-CT) analysis Cranial surface erosion was measured using micro-CT (SkyScan1176, Belgium). According to the manufacturer’s instructions, the scanning parameters for achieving isometric resolution are configured at 9 μm, while the X-ray energies are precisely set to 80 kV and 100 mA. 3D image reconstruction and quantitative analysis using SkyScan software. An equal area (3 mm in diameter) of area of interest (ROI) was found in the center of each cranium. We utilized SkyScan software to assess bone mineral density (BMD, mg/cm^3), bone volume (BV, mm^3), bone volume/tissue volume (BV/TV), and pores within the region of interest. Histological and immunohistochemical analysis After undergoing decalcification for a period of 28 days in a solution containing 10% ethylene diamine tetraacetic acid (EDTA) obtained from Sigma (USA), the crania were embedded in molten paraffin. Afterwards, the tissue samples were cut into 6 μm sections and processed following the guidelines provided by the manufacturer for staining with hematoxylin and eosin (H&E). The resulting visuals were captured using a Carl Zeiss microscope (Germany). The assessment of dynamic bone formation was conducted using the calcein double labeling assay. In brief, a solution of calcein (Sigma, USA) was administered intraperitoneally in phosphate buffered saline (PBS) at a dosage of 10 mg/kg on the 1st and 8th days prior before sacrifice. The fluorescence microscope (Germany) was utilized to capture images of undecalcified bone sections labeled with calcein, and the calculation of mineral apposition rate (MAR) was performed using image J. Immunohistochemistry (IHC) staining was employed to detect the expression levels of GPX4. In brief, tissue sections were deparaffinized and rehydrated using a gradient method for antigen retrieval. Subsequently, GPX4 primary antibody (ab125066) from Abcam (USA) was applied to the tissue sections and incubated overnight at 4 °C. Following that, corresponding secondary antibodies were used for blocking the sections for 30 min. The DAB Kit provided by Beyotime (China) was used to initiate the chromogenic reaction. Analysis was conducted on randomly selected areas throughout the entire field of view. Finally, fluorescence microscopy (Zeiss, Germany) was utilized to observe the tissue sections. Immunofluorescence staining was performed to ascertain the presence of GPX4, in accordance with established protocols and procedures. In brief, tissue sections were subjected to dewaxing, antigen retrieval, and blocking with 2% BSA for a duration of 60 min. Subsequently, primary antibodies targeting GPX4 (ab125066) from Abcam (USA) were added and incubated overnight at a temperature of 4 °C. Following this step, the corresponding secondary fluorescent antibodies 488 (Abcam, USA) were applied onto the sections in darkness for an hour. Afterwards, DAPI was utilized to counterstain the nuclei for a period of 10 min. Finally, observation of the tissue sections was performed using a fluorescence microscope manufactured by Zeiss (Germany). Quantification and statistical analysis The data represent the mean ± standard deviation (SD). The statistical analysis was conducted using GraphPad Prism version 8.0 software. A Student’s t-test was employed to ascertain the presence of statistically significant disparities between two groups. The significant difference among multiple groups was accessed by one-way ANOVA where appropriate (*P < 0.05; **P < 0.01). Results Ti nanoparticles suppressed osteogenic differentiation In order to investigate the impact of wear particles on the suppression of osteoblasts, we have opted to utilize Ti nanoparticles as an intervention in osteoblasts to replicate the complex microenvironment of osteolysis in vitro (Fig. [76]S2A). The results of ALP staining and ARS staining demonstrated a significant decrease in the osteogenic activity and extracellular mineralization when treated with both the 5 µg/cm^2 and 10 µg/cm^2 Ti nanoparticles (Fig. [77]1A-D). The expression level of osteogenic genes was found to be significantly reduced in both the low- and high-dose groups exposed to Ti nanoparticles (Fig. [78]1E). Similar results were also confirmed by western blot (Fig. [79]1F and G). These results showed that the presence of Ti nanoparticles had a significant inhibitory effect on osteogenic differentiation and the mineralization of the extracellular matrix. Fig. 1. [80]Fig. 1 [81]Open in a new tab Ti nanoparticles suppressed osteogenic differentiation. (A)-(B) Representative images of ALP staining and ARS staining. (scale bar = 100 μm). (C)-(D) Quantitative analysis of relative ALP activity and ARS recovery ratio. (E) The mRNA levels of osteoblast-specific genes with the intervention of Ti nanoparticles. (F) Protein levels of COL1-a1, Runx2 and Osterix. (G) Quantification of osteogenesis-related markers shown in (F). Data are mean ± SD, n = 3; *P < 0.05, **P < 0.01 vs. Control. ns indicates not significant Ferroptosis was involved in osteoblasts with the intervention of Ti nanoparticles In order to investigate the underlying mechanism through which Ti nanoparticles exert detrimental effects on osteoblasts, we performed RNA sequencing to identify genes that displayed differential expression between the control and Ti groups. Total mRNA was extracted from osteoblasts following exposure to 10 µg/cm^2 Ti nanoparticles for a duration of 72 h. The Illumina HiSeq 2500 platform was employed to conduct RNA-seq analysis and generate mRNA profiles (Fig. [82]2A and Fig. [83]S3A). The volcano plot revealed that a total of 1762 genes were upregulated, while 1008 genes were downregulated after Ti nanoparticles treatment (Fig. [84]2B). The aforementioned findings were represented graphically in the form of a heat map, and then GO categories were used to annotate the 2770 differentially expressed genes (DEGs) (Fig. [85]2C, D and Fig. [86]S3D). The GO enrichment analysis demonstrated a significant enrichment of pathways associated with the transport and homeostasis of iron ions, which may potentially be linked to the occurrence of ferroptosis [[87]26]. In addition, pathway enrichment analysis was conducted using the Kyoto Encyclopedia of Genes and Genomes (KEGG) on the dataset obtained from RNA sequencing. Interestingly, the genes that exhibited significant differential expression led to a notable alteration in the ferroptosis pathway, as depicted in Fig. [88]2E and Figure [89]S3B. The Ti group exhibited a significant enhancement and stimulation of the ferroptosis pathway, which was verified through gene set enrichment analysis (GSEA) (Fig. [90]2F). Additionally, the induction of ferroptosis resulted in a notable decrease in the level of GPX4 and the expression of osteogenesis-related genes, including Col1-a1, Alp, Runx2, and Sp7. This confirmation was supported by the visually striking heat map and cluster analysis conducted on the gene set linked to ferroptosis and differentially expressed genes (DEGs) (Fig. [91]2G and Fig. [92]S3C) [[93]27]. Likewise, we observed that Ti nanoparticles suppressed osteoblast function to a degree comparable to the classical inducer of ferroptosis called erastin (Fig. [94]S4A-C). In summary, this RNA-seq analysis demonstrated that the inclusion of Ti nanoparticles has the capacity to elicit a reduction in osteogenic activity by triggering the onset of ferroptosis. Fig. 2. [95]Fig. 2 [96]Open in a new tab RNA sequencing of osteoblasts under the intervention of Ti nanoparticles. (A) A schematic diagram depicting the experimental design and protocol for sample preparation in RNA sequencing. (B) The volcano plot revealed differentially expressed genes. (C) Heat map depicts genes that were differentially expressed between the Control and Ti groups. Red: upregulated genes. Blue: downregulated genes. (D) Significant pathway of GO Enrichment analysis. (E) KEGG pathway enrichment analysis of the differentially expressed genes. (F) GSEA enrichment plot for the ferroptosis pathway based on RNA sequencing. (G) The heat map revealed the genes significantly regulated in relation to osteogenesis and ferroptosis Subsequently, the impact of Ti nanoparticles on ferroptosis were investigated in osteoblasts. The TEM analysis demonstrated a reduction or absence of mitochondrial ridges and disruption of the outer mitochondrial membrane (OMM) in osteoblasts after a 72-hour intervention with Ti nanoparticles, which serve as a critical indicator of ferroptosis (Fig. [97]3A). Simultaneously, the inclusion of Ti nanoparticles led to a notable elevation in ROS levels and a decline in the mitochondrial membrane potential, indicating an impairment in mitochondrial functionality (Fig. [98]3B). Ti nanoparticles stimulation resulted in a sharp increase of FerroOrange in the fluorescence intensity, which suggested that iron homeostasis was disrupted in osteoblasts, thus inducing ferroptosis (Fig. [99]3B). Likewise, the generation of MDA and the concentration of total iron (Fe) exhibited the identical increased trends (Fig. [100]3 Cand D). The results of western blot and RT-PCR indicated that with the increase of the concentration of Ti nanoparticles, the protein levels and gene levels of critical regulators significantly changed (Fig. [101]3E-G). Consistently, Ti nanoparticles significantly suppressed the content of GSH and the subsequent impact on the activity of GPX4, which was consistent with the results of clinical samples (Fig. [102]S5A). Taken together, the obtained detection results provide conclusive evidence supporting the correlation between Ti nanoparticles and osteoblastic ferroptosis. Fig. 3. [103]Fig. 3 [104]Open in a new tab Ti nanoparticles promote osteoblastic ferroptosis. (A) TEM images of osteoblasts after treatment with Ti nanoparticles (5 and 10 µg/cm^2). Scale bar = 5 μm (upper) and 500 nm (lower). (B) Representative images of Ti nanoparticle-induced ROS generation (scale bar = 100 μm), Mitochondrial membrane potential (scale bar: 50 μm) and FerroOrange staining (scale bar = 50 μm) after Ti nanoparticles treatment. (C) The level of MDA. (D) The content of cellular total iron. (E) The mRNA levels of ferroptosis-related genes. (F) Protein levels of 4HNE, COX2, SLC7A11 and GPX4 after the intervention of Ti nanoparticles. (G) Quantification of ferroptosis-related markers in (F). Data are mean ± SD, n = 3; *P < 0.05, **P < 0.01 vs. Control. ns indicates not significant Osteoblastic ferroptosis was involved in peri-prosthesis osteolysis In order to investigate the potential relationship between PPO and ferroptosis, bone tissue samples were collected from the RTJA group, while bone tissue from the PTJA group was utilized as a control (Fig. [105]S6A). In the RTJA group, a notable decrease in the expression of markers associated with bone formation, such as COL1-a1, Runx2, and Osterix was observed when compared to the PTJA group (Fig. [106]4A and B). In the RT-PCR assay, a significant reduction in bone formation capacity was observed subsequent to the loosening of the prosthesis (Fig. [107]4E). The findings of our study aligned with prior research indicating a decline in bone formation capacity when a prosthesis becomes loose [[108]28]. Further investigations have uncovered notable alterations in the expression levels of crucial ferroptosis-related proteins, such as GPX4 and SLC7A11, in the RTJA group compared to the PTJA group. Specifically, the expression levels of GPX4 and SLC7A11 were significantly downregulated, while COX2 expression was observed to be increased (Fig. [109]4C and D). The progressive modification in the expression of genes linked to ferroptosis offers additional confirmation of its potential role in inhibiting the osteogenic function during peri-prosthesis osteolysis (Fig. [110]4F). Meanwhile, the results of immunofluorescence staining demonstrated a significant decrease in the number of GPX4-positive cells observed on the surface of bone tissue in the RTJA group compared to the PTJA group. This finding suggests that GPX4 may play a critical role in the suppression of ferroptosis induced by peri-prosthetic osteolysis (Fig. [111]S6B). Additionally, significant increases in MDA levels, a byproduct of lipid oxidation, and total iron concentrations were observed in the RTJA group. These findings suggest the presence of iron accumulation and lipid peroxidation in the bone tissue during peri-prosthetic osteolysis, as depicted in Fig. [112]4G and H. The assessment of the content of GSH, which acts as the reducing substrate for GPX4 activity, was also conducted. The results revealed a significant decrease in the levels of GSH in the RTJA group compared to the PTJA group (Fig. [113]4I). Taken together, the aforementioned findings suggest that osteogenic ferroptosis plays a crucial role in the suppression of osteogenic function induced by wear particles. Fig. 4. [114]Fig. 4 [115]Open in a new tab Osteoblastic ferroptosis is involved in peri-prosthesis osteolysis. (A) Protein levels of COL-a1, Runx2 and Osterix in the PTJA and RTJA group. (B) Relative protein quantification of osteogenic markers in (A). (C) Protein levels of COX2, SLC7A11 and GPX4. (D) Relative protein quantification of ferroptosis-related markers in (C). (E) The mRNA levels of osteoblast-specific genes. (F) The mRNA levels of ferroptosis- related genes. (G) The level of MDA. (H) The content of total iron. (I) The content of GSH. Data are mean ± SD, n = 3; *P < 0.05, **P < 0.01 vs. RTJA. ns indicates not significant Suppression of ferroptosis promoted osteogenic differentiation and alleviated PPO Given the significant role of ferroptosis in the suppression of osteogenesis and the promotion of PPO, it is postulated that the inhibition of ferroptosis could potentially restore osteogenesis and mitigate PPO. Fer-1 was identified as an early synthetic radical-trapping antioxidant (RTA) with documented efficacy in inhibiting ferroptosis so that it was employed for the subsequent study [[116]29]. The CCK8 analysis was utilized to assess the impact of Fer-1 concentration on osteoblast proliferation and viability (Fig. [117]S7A). Fer-1 was applied to osteogenic differentiated MC3T3-E1 cells with 10 g/cm^2 Ti nanoparticles for 3 days. The western blot results demonstrated that Fer-1 treatment effectively inhibited the degradation of SLC7A11 and GPX4 and decreased the expression of COX2, indicating the inhibition of osteoblastic ferroptosis (Fig. [118]5A and B). Hereafter, ALP staining and ARS staining were used to verify the effects of Fer-1 on osteogenic function. As anticipated, Fer-1 effectively restored the inhibited osteogenic cell function caused by Ti nanoparticles (Fig. [119]5C-F). The expression level of OCN was further investigated through immunofluorescence staining. Treatment with Ti (10 µg/cm^2) significantly reduced the average fluorescence intensity of OCN. However, Fer-1 exhibited a notable upregulation in OCN expression (Fig. [120]5G). In the meanwhile, the osteogenic-related markers were further detected by western blot and RT-PCR, and osteogenic-related markers such as COL1-a1, Runx2, and Osterix were significantly upregulated after the treatment of Fer-1 (Fig. [121]S7B-D). In conclusion, the application of Fer-1 demonstrated a significant ability to restore osteoblast differentiation by effectively suppressing ferroptosis in an in vitro setting. Fig. 5. [122]Fig. 5 [123]Open in a new tab Suppression of osteoblastic ferroptosis promoted osteogenic differentiation. (A) Protein levels of COX2, SLC7A11 and GPX4 after Ti nanoparticles (10 µg/cm^2) and Fer-1 (5µmol/L) treatment. (B) Quantification of ferroptosis-related markers in (A). (C)-(D) Representative images of ALP staining and ARS staining. (scale bar = 100 μm). (E)-(F) Quantitative analysis of relative ALP activity and ARS recovery ratio. (G) Illustrative images of immunofluorescence staining; green (OCN), red (F-actin) and blue (nuclei). (scale bar = 25 μm). Data are mean ± SD, n = 3; *P < 0.05, **P < 0.01 vs. Ti group. ns indicates not significant To provide additional clarity on the influence of Fer-1 on peri-prosthesis osteolysis, we established a calvarial osteolysis model by Ti nanoparticles (40 mg per mouse) and treated with Fer-1 (0.1 mg/kg). As shown by the micro-CT and 3D reconstructed images in Fig. [124]6A, the Ti group induced an extensive fibrous layer containing a high concentration of inflammatory cells and Fer-1 treatment significantly alleviated fibrous hyperplasia and bone resorption. Compared to the Ti group, the Fer-1 group showed higher bone morphometric parameters including BMD, BV as well as BV/TV (increased approximately 36.5%, 69.6% and 40.6%, respectively), indicating better osteogenesis after the Fer-1 treatment (Fig. [125]6B-D). A noticeable decrease in the number of pores was observed in the Fer-1 group, which was statistically significant (Fig. [126]6E). H&E staining confirmed the mitigation of osteolysis induced by Ti nanoparticles following the effective administration of Fer-1 (Fig. [127]6F). Additionally, the application of Fer-1 led to enhanced bone formation and a higher rate of mineral apposition, as demonstrated by the calcein double labeling (Fig. [128]6G). The changes were further confirmed by quantitatively analyzing the average thickness of mineral apposition (Fig. [129]S8A). Concomitantly, the Fer-1 group showed the increased number of GPX4-positive cells in the area of the osteolysis than the Ti group, indicating the suppression of osteoblastic ferroptosis (Fig. [130]6H and Fig. [131]S8B). The H&E staining of the major organs showed no obvious morphological abnormalities after Fer-1 treatment as shown in Fig. [132]S8C. These encouraging results indicate that the Fer-1 therapy is effective in alleviating the Ti nanoparticles induced osteolysis in vivo. Fig. 6. [133]Fig. 6 [134]Open in a new tab Fer-1 mitigated Ti nanoparticle-induced osteolysis. The concentration of Ti nanoparticles and Fer-1 in vivo were 40 mg per mouse and 0.1 mg/kg, respectively. (A) Exhibited 3D reconstruction images of Micro CT. (B) BMD (mg/cm^3). (C) BV (mm^3). (D) BV/TV (%). (E) Number of pores. (F) Representative images of H&E staining. (scale bar = 100 μm and 25 μm). (G) Calcein double labelling. (scale bar = 25 μm). (H) Immunohistochemical analysis of GPX4 protein. (scale bar = 100 μm and 25 μm). Data are mean ± SD, n = 5; *P < 0.05, **P < 0.01 vs. Ti group. ns indicates not significant Identification of natural small molecules targeting GPX4 to suppress ferroptosis Our study has revealed the significant role of GPX4 and ferroptosis in inhibiting bone formation caused by Ti nanoparticles. Additionally, Fer-1 showed significant effectiveness in suppressing osteoblastic ferroptosis, which reduced the osteolytic effects caused by Ti nanoparticles and promoted osteogenesis. However, despite its widespread use in experiments, the clinical application of Fer-1 has been limited due to its instability. This includes issues such as hydrophobicity, inadequate pharmacokinetics, and unknown toxicity with prolonged in vivo use [[135]30, [136]31]. In recent years, there has been a significant increase in the demand for pharmaceuticals, which has led to a greater emphasis on the use and potential of natural products in drug research [[137]32]. Natural products offer several advantages, including enhanced stability, a wide range of physiologically active components, and the potential to avoid adverse effects associated with conventional chemical synthesis medications [[138]33]. The active site of the GPX4 protein was identified using the SiteMap module within the Schrödinger software package. Relevant drugs were then screened in the natural product database using a model of the GPX4 protein binding (Fig. [139]7A) [[140]34, [141]35]. Subsequently, we utilized the Canvas 1.1 program to filter out pan-assay interference compounds (PAINS) from the broad screening of interfering compounds. We employed the QikProp 3.2 program within the Schrödinger software package to calculate the pharmacokinetic properties of the compounds selected. Additionally, molecules obtained through various pharmacophore screenings and docking were merged, duplicate molecules were removed, and the remaining small molecules were subjected to structural clustering analysis using FCFP_6 fingerprints. Finally, the optimal docking conditions using the Glide force field were selected, conducting docking studies with the GPX4 protein as the receptor. An active pocket was defined with a size of 10 Å × 10 Å × 10 Å. We conducted a screening of Urolithin A, a natural product, taking into account its physicochemical properties, docking scores, multiple interactions, preservation challenges, and economic factors. This compound is a natural metabolite of ellagitannin, a group of compounds found in pomegranate, as well as in other fruits and nuts. UA has been shown to have properties that can reverse mitochondrial and muscle aging, as shown in Fig. [142]7B [[143]36]. The validation of the GPX4 protein and UA was conducted using network pharmacology and molecular docking techniques. The appropriate conformation and spatial arrangement of UA demonstrated the steric complementarity between UA and the binding site of GPX4. The docking score between UA and GPX4 is -6.614. The binding mode revealed that UA binds the pocket of GPX4 by 4 H-bond bindings: ASP-21 (2.2 Å), PHE-100 (3.1 Å), VAL-98 (3.2 Å), LYS-90 (2.1 Å) (Fig. [144]7C). All of these interactions were beneficial for stabilizing the binding complexes. To validate the virtual screening and molecular docking results, we conducted cellular thermal shift assays (CETSA). As shown in Fig. [145]7D and E, compared to DMSO alone, GPX4 showed markedly enhanced thermal stability following treatment with UA. In conclusion, these findings suggest that UA is capable of effectively targeting GPX4 to exert its functions. Fig. 7. [146]Fig. 7 [147]Open in a new tab Identification of natural small molecules targeting GPX4 to suppress ferroptosis. (A) The active site of the GPX4 protein. (B) Screening by GPX4 protein to obtain a 2D map of urolithin A small molecules and forces of action. (C) The binding mode of GPX4 with UA. H-bond (yellow): ASP-21 (2.2 Å), PHE-100 (3.1 Å), VAL-98 (3.2 Å), LYS-90 (2.1 Å). (D) Cellular thermal shift assay between UA and GPX4. (E) The levels of GPX4 protein under different temperatures in the presence of UA and DMSO treatment. (F) Representative TEM images of osteoblasts after UA treatment. Scale bar = 5 μm (upper) and 500 nm (lower). (G) The content of MDA. (H) Representative images of Ti nanoparticles-induced ROS generation (scale bar = 100 μm), Mitochondrial membrane potential (scale bar = 50 μm) and FerroOrange staining (scale bar = 50 μm) after UA treatment. (I) The content of cellular total iron. (J) Protein levels of COX2, SLC7A11, GPX4. (K) The mRNA levels of ferroptosis-related genes. Data are mean ± SD, n = 3; *P < 0.05, **P < 0.01 vs. Ti group. ns indicates not significant The cytotoxicity and proliferative toxicity of UA were initially assessed using the CCK-8 assay. The results indicated that the concentration of 20 µmol/L UA did not exhibit significant cytotoxic effects, which was used in the following study to investigate the effects of UA on osteoblastic ferroptosis and osteogenesis (Fig. [148]S9A). Mitochondrial ultrastructure examined by TEM revealed that the mitochondrial morphology in the UA group showed a significant improvement compared to the group exposed to Ti nanoparticles, specifically characterized by a reduction in mitochondrial fragmentation (Fig. [149]7F). Similarly, UA treatment effectively suppressed the generation of MDA induced by Ti nanoparticles, exerting a potent inhibitory effect on ferroptosis (Fig. [150]7G). Furthermore, the staining of MMP, ROS, and FerroOrange was performed to provide additional evidence of the impact of UA on osteoblastic ferroptosis. These results indicated that UA treatment resulted in an elevation of MMP levels, while concurrently decreasing the production of Fe^2+ and ROS, which is comparable to the role of the ROS inhibitor NAC (Fig. [151]7H and Fig. [152]S9D). Similar patterns were also noted in the assessment of the overall iron content within the cells (Fig. [153]7I). The expression levels of ferroptosis-related markers were also examined following the treatment of UA. Western blot assays revealed that UA not only promoted protein level of GPX4 and SLC7A11, but also reduced the expression level of COX2 (Fig. [154]7J and [155]S9B). The mRNA levels of Cox2, Slc7a11 and Acsl4 and the quantification of GSH demonstrated the consistent expression as expected (Fig. [156]7K and Fig. [157]S9C). Likewise, we observed that UA suppressed osteoblastic ferroptosis following intervention with Ti nanoparticles to a degree comparable to the ROS inhibitor NAC (Fig. [158]S9E-G). Additionally, Erastin, the classical ferroptosis inducer, induced osteoblastic ferroptosis was also inhibited by UA treatment (Fig. [159]S10). To provide additional evidence regarding that UA inhibited ferroptosis rely on the upregulation of GPX4, specific siRNA was employed to effectively downregulate GPX4 expression (Fig. [160]8A). As expected, the protective effects of UA on Ti nanoparticles induced ferroptosis are markedly attenuated by GPX4 silence (Fig. [161]8B-D). The results showed that UA-activated GPX4 suppressed ferroptosis. Fig. 8. [162]Fig. 8 [163]Open in a new tab Silencing GPX4 reversed UA inhibition of ferroptosis. (A) The mRNA level of Gpx4 after GPX4 silence. (B) The mRNA levels of ferroptosis-specific genes after GPX4 silence in each group. (C) Protein levels of COX2, SLC7A11 and GPX4 after GPX4 silence. (D) Quantification of ferroptosis-related markers shown in (C). Data are mean ± SD, n = 3; *P < 0.05, **P < 0.01 vs. Ti + siGPX4 group. ns indicates not significant UA promoted osteoblast differentiation and attenuated Ti nanoparticle-induced osteolysis by the activation of GPX4 Based on the above results, we further evaluated the effect of UA on Ti nanoparticle-induced inhibition of osteogenic function. The ALP and ARS staining results provide direct evidence that UA had a significant impact on the restoration of osteoblast activity impaired by Ti nanoparticles, as well as the promotion of extracellular matrix mineralization (Fig. [164]9A-D). Subsequently, the expression of osteogenic indicators at the protein and gene levels was detected in the condition with or without UA treatment. We found that UA treatment significantly promoted the expression of osteogenic differentiation-related proteins, including COL1-a1, Runx2, and Osterix and the relative gene expression in a dose-dependent manner (Fig. [165]9E-G). Meanwhile, the results of cell immunofluorescence staining further supported the promoting effect of UA on osteogenesis (Fig. [166]9H). However, these positive effects were impaired by GPX4 silence (Fig. [167]9I-L). Taken together, the above results demonstrated that UA could alleviate the inhibition of osteogenesis induced by Ti nanoparticles, and this effect was dependent on the activation of GPX4. Fig. 9. [168]Fig. 9 [169]Open in a new tab UA promoted osteoblast differentiation by the activation of GPX4. (A)-(B) Representative images of ALP and ARS staining during Ti nanoparticles and UA treatment. (scale bar = 100 μm). (C)-(D) Quantitative analysis of ALP activity and ARS recovery ratio. (E) The mRNA levels of osteogenesis-related gene after UA treatment. (F) Protein levels of COL-a1, Runx2 and Osterix. (G) Quantification of osteogenic markers shown in (F). (H) Cellular immunofluorescence of OCN. (scale bar = 25 μm). Data are mean ± SD, n = 3; *P < 0.05, **P < 0.01 vs. Ti group. ns indicates not significant. (I)-(J) Representative images of ALP and ARS staining after GPX4 silence. (scale bar = 100 μm). (K)-(L) Quantitative analysis of ALP activity and ARS recovery ratio. Data are mean ± SD, n = 3; *P < 0.05, **P < 0.01 vs. Ti + UA + siGPX4 group. ns indicates not significant In view of the encouraging bone-promoting effect of UA in vitro, we then explored whether UA could reduce Ti nanoparticle-induced osteolysis. As shown by the micro-CT scanning and 3D reconstruction results in Fig. [170]10A, the bone destruction of osteolysis improved significantly after the UA treatment in a dose-dependent manner. The bone morphometric parameters observed in the UA group exhibit superiority over those of the Ti group (Fig. [171]10B-E). In addition to micro-CT, histological analysis serves to validate the effects of UA in mitigating osteolysis. As depicted in Fig. [172]10F, an extensive fibrous layer containing a high concentration of inflammatory cells is observed in the Ti group. In contrast, the UA group exhibited a significant increase in bone mass and a notable decrease in fibrous hyperplasia and bone destruction (Fig. [173]10F). The dynamic osteogenesis was found to be higher than that in the Ti group, and it was even comparable to the rate observed in the Control group (Fig. [174]10G and Fig. [175]S11A). On the other hand, Ti nanoparticle significantly reduced the level of GPX4 in bone destruction areas, while UA treatment effectively upregulated the expression of GPX4 to resist ferroptosis for osteoblasts (Fig. [176]10H and Fig. [177]S11B). The histological examination of major organs in the experimental mice revealed no significant morphological abnormalities among the different groups (Fig. [178]S11C). These results provide evidence that the UA treatment reduces further bone destruction induced by Ti nanoparticle via the suppression of ferroptosis and promotion of osteogenesis. Fig. 10. [179]Fig. 10 [180]Open in a new tab UA attenuated Ti nanoparticle-induced osteolysis. (A) Representative 3D reconstruction images of micro-CT. (B) BMD (mg/cm^3). (C) BV (mm^3). (D) BV/TV (%). (E) Number of pores. (F) Representative images of H&E staining. (scale bar = 100 μm and 50 μm). (G) Calcein double labelling. (scale bar = 25 μm). (H) Immunohistochemical analysis of GPX4 protein. (scale bar = 100 μm and 50 μm). Data are mean ± SD, n = 5; *P < 0.05, **P < 0.01 vs. Ti group. ns indicates not significant Discussion Total joint arthroplasty is widely recognized as the most advanced surgical treatment for trauma, degenerative arthritis, and other severe joint conditions [[181]37]. However, the presence of wear particles, such as Ti nanoparticles, can lead to peri-prosthesis osteolysis and subsequent aseptic loosening, which can cause total joint arthroplasty to fail. This not only creates a significant economic burden but also has social implications [[182]38, [183]39]. Previous studies have primarily focused on examining the effects of wear particles on osteoclast stimulation. However, these studies have overlooked the important role of osteoblasts in bone formation [[184]40, [185]41]. Ameliorating the inhibitory effects of wear particles on osteogenic differentiation may represent a crucial factor for the therapeutic potential of PPO. The study found that the presence of Ti nanoparticle-induced osteoblastic ferroptosis by the inhibition of GPX4. Fer-1 reversed this process and restored osteogenic function to protect against bone loss in vitro and vivo. Considering the limitations of Fer-1 in the clinic, a natural compound known as UA was screened using network pharmacology for its specific targeting of GPX4. Subsequently, it was further confirmed that UA alleviated Ti nanoparticle-induced osteoblastic ferroptosis and promoted osteoblast differentiation, but this effect was inhibited by silencing GPX4. In addition, UA effectively alleviated osteolysis-induced bone loss in the mouse model. This study validated the feasibility of osteolysis therapy via the suppression of osteoblastic ferroptosis and the results provide insights into drug development for future clinical applications. Ferroptosis is a form of regulated cell death that is mediated by iron, which occurs when lipid peroxides accumulate on cellular membranes, resulting in a toxic cellular environment [[186]42]. Several studies have suggested a potential link between ferroptosis and various diseases, including cancer, neurodegenerative diseases, acute kidney injury, and ischemia reperfusion [[187]43–[188]47]. Recent studies have shown that ferroptosis has an impact on orthopedic diseases, including osteoporosis and PPO, which leads to a decrease in osteogenic function and impairs the ability to form bone [[189]13, [190]48]. However, further investigation is necessary to understand the underlying mechanisms and identify the optimal targets for clinical applications. Our study analysed experimental data from clinical specimens and in vitro experiments, revealing a strong correlation between stimulation of osteoblastic ferroptosis by Ti nanoparticles and the development of periprosthetic osteolysis. RNA Seq analysis revealed that wear particles downregulated the expression of GPX4 and osteogenesis-related genes in osteoblasts, resulting in ferroptosis and a decrease in bone formation. Our findings demonstrate the effectiveness of Fer-1, a classic iron death inhibitor, in countering the induction of ferroptosis by Ti nanoparticles and promoting bone formation both in vivo and in vitro. The study provides evidence that wear particles induce osteoblast ferroptosis in PPO. The presence of Ti nanoparticles in peri-prosthetic osteolysis has been found to trigger osteoblastic ferroptosis, which reduces the ability of bone formation. The small molecule Fer-1 provides cellular protection against damage caused by lipid peroxides. It achieves this by effectively capturing free radicals and stabilizing lipid peroxides, which in turn prevents further oxidative reactions [[191]49]. Additionally, Fer-1 has been found to regulate intracellular iron ion metabolism. This compound effectively inhibits the accumulation and release of iron ions, preventing excessive free iron ions from participating in oxidative reactions and reducing the production of oxidative stress [[192]50]. In their research on ferroptosis in orthopedic diseases, Xu et al. found that Fer-1 can inhibit osteoblastic ferroptosis and improve peri-prosthetic osteolysis induced by CoCrMo nanoparticles [[193]13]. A recent study has shown that Fer-1 has the ability to slow down the progression of osteoporosis by inhibiting ferroptosis in osteoblasts and suppressing senescence markers associated with osteoblasts. This helps to prevent age-related osteoporosis [[194]16]. Our study has confirmed the important role of Fer-1 in promoting osteogenesis and inhibiting ferroptosis both in vitro and in vivo. However, despite the overwhelming evidence of its usefulness, Fer-1 has been limited in its clinical application because it is unstable in vivo [[195]30, [196]51]. We discovered a natural product called Urolithin A through virtual screening of the GPX4 protein, which is known as a key target of ferroptosis. UA, a component of the ellagitannin family, has shown its potential to improve metabolic health in humans and rodents. It has also been found to increase the lifespan of worms by supporting mitophagy and promoting optimal mitochondrial function [[197]52, [198]53]. Previous studies have shown that UA has a protective effect against ferroptosis in acute lung injury. It achieves this by increasing the expression of important proteins such as GPX4 and SLC7A11, as well as activating the Keap1-Nrf2/HO-1 signaling pathway [[199]54]. UA also demonstrates significant therapeutic potential in the prevention of diabetes mellitus (DM)-related Alzheimer’s disease (AD) pathology, which can be realized by inhibiting TGM2-mediated MAM assembly and maintaining mitochondrial calcium homeostasis as well as reactive oxygen species (ROS) stability [[200]18, [201]55]. Furthermore, under the potential of a functional polymer-lipid drug delivery system, this lays the groundwork for further investigation into the immunotherapeutic potential of UA for chemotherapy-induced acute kidney injury in cancer patients [[202]56–[203]58]. After examining the role of UA and GPX4 proteins in ferroptosis, we discovered that UA specifically binds to the GPX4 protein through molecular docking. Our study demonstrated the effectiveness of UA in enhancing the expression and activity of GPX4, as evidenced by western blot and RT-PCR results. This highlights the significant potential of UA. In recent years, UA has emerged as a crucial factor in the management of various orthopedic diseases. Our previous study showed that UA inhibits osteoclastogenesis and postmenopausal osteoporosis by suppressing inflammation and NF-κB-mediated pyroptotic pathways [[204]23]. Recent studies have found that UA effectively inhibits the harmful effects of inflammation-induced bone loss and specifically targets the PI3K/Akt/NF-κB pathway. This provides relief for individuals with osteoarthritis [[205]59]. In this study, we found that UA effectively suppressed ferroptosis in osteoblasts while also enhancing their osteogenic function in vitro. Additionally, our in vivo experiments provided evidence that UA promotes GPX4 expression and reduces periprosthetic osteolysis by suppressing ferroptosis. The compound demonstrates a therapeutic effect similar to Fer-1, making it a promising treatment option for PPO. Conclusion Osteoblastic ferroptosis was found to contribute to the pathogenesis of PPO. The specific inhibitor of ferroptosis, Fer-1 promoted the osteoblast susceptibility to ferroptosis by elevating the expression of GPX4 and rescue their osteogenic function thus halting the progression of peri-prosthesis osteolysis. A small molecule, UA, targeting GPX4 was found via network pharmacology and was confirmed to mitigate Ti nanoparticle-induced osteoblastic ferroptosis and impaired osteogenesis. In animal model, UA exerted a therapeutic impact on osteolysis induced by Ti nanoparticles. In conclusion, this study provides a new insight into osteolysis and a novel therapeutic approach for PPO. Electronic supplementary material Below is the link to the electronic supplementary material. [206]Supplementary Material 1^ (7.3MB, docx) Acknowledgements