International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Osteoporosis is a disease characterized by changes in bone microstructure, including thinning of trabeculae and an increased susceptibility to brittle fractures (1). Osteoporosis primarily results from an imbalance in bone metabolism, where bone formation is weakened while bone resorption increases, leading to a loss of bone mass (2). Factors such as aging, inflammation and hormonal changes lead to a reduction in bone formation (3); however, the specific reasons have not yet been fully elucidated.
Bone marrow mesenchymal stem cells (BMSCs) are a type of pluripotent stem cell with the ability to differentiate into three lineages: Osteoblasts, adipocytes and chondrocytes (4). It has previously been shown that the differentiation strength of BMSCs is markedly associated with bone changes in vivo (5). Research using tissue engineering functional scaffolds has demonstrated that increasing the osteogenic differentiation ability of BMSCs leads to a notable increase in bone formation in vivo (6). Therefore, BMSCs are considered an important target and functional cell for treating diseases characterized by reduced bone formation (7,8). However, the specific regulatory mechanism underlying the osteogenic differentiation of BMSCs has not yet been fully elucidated and requires further investigation.
Forkhead box (FOXJ3) possesses DNA-binding transcriptional activation activity, RNA polymerase II specificity and sequence-specific double-stranded DNA-binding activity. Notably, FOXJ3 participates in the positive regulation of RNA polymerase II transcription. Previous studies have shown that FOXJ3 is related to the progression of various diseases, such as rheumatoid arthritis (9), and spermatogenesis (10). Furthermore, it has been reported to serve an important role in the disease evolution process in cancer (11,12). At the metabolic level, it has been reported that FOXJ3 can promote the thermogenic effect of fat (13). In addition, FOXJ3 can promote the formation of osteoclasts (14). Fat metabolism and osteoclastogenesis in the bone marrow are associated with bone formation and other processes. Given the important role of BMSCs in bone formation, it is crucial to clarify whether FOXJ3 affects the osteogenic differentiation function of BMSCs and its role in bone metabolic diseases. However, to the best of our knowledge, no relevant studies have yet been published.
The Wnt/β-catenin pathway is a crucial pathway that serves important roles in various cell functions, including cell proliferation (15) and differentiation (16). It has previously been shown that this pathway can promote the osteogenic differentiation of BMSCs (17). After activation of the Wnt/β-catenin protein and its entry into the nucleus, it can activate the expression of osteogenic-related molecules, promote the secretion of extracellular matrix proteins, and the synthesis of alkaline phosphatase (ALP) and other substances by BMSCs, thereby promoting mineralization (18). Therefore, the present study aims to investigate whether FOXJ3 is involved in the osteogenic differentiation of BMSCs and whether it exerts its regulatory function through the Wnt/β-catenin pathway, thereby providing a new therapeutic target for bone metabolic diseases.
Rat BMSCs were obtained from Wuhan Servicebio Technology Co., Ltd. Since BMSCs from passages 3–6 exhibit a homogeneous population, consistent morphology and robust osteogenic differentiation functionality, all experiments were conducted using cells within this passage range. Adherent BMSCs were cultured in DMEM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (FBS; Zhejiang Tianhang Biotechnology Co., Ltd.) in a humidified incubator maintained at 37°C with 5% CO2.
BMSCs were seeded at a density of 1×105 cells/well in 12-well plates. Following medium renewal on day 2, the BMSCs were induced to differentiate by culturing them in low-glucose DMEM supplemented with 10% FBS, 10−8 M dexamethasone (Sigma-Aldrich; Merck KGaA; cat: D4902), 50 µg/ml ascorbic acid 2-phosphate (Sigma-Aldrich, cat: 49752) and 10 mM β-glycerophosphate (Sigma-Aldrich; Merck KGaA; cat. no. G9422). The culture medium was refreshed every 3 days. Osteogenic differentiation medium was supplemented with 5 µM SB216763 (19–21) or 10 µM XAV939 (22,23) (both from Shanghai Aladdin Biochemical Technology Co., Ltd.) to activate or inhibit the Wnt/β-catenin signaling pathway during BMSC culture in 37°C, respectively. The DMSO group was supplemented with the same volume of DMSO as the groups treated with SB216763 or XAV939.
BMSCs were seeded at a density of 1×10 cells/well in 12-well plates. ALP activity was assessed following 10 days of osteogenic differentiation, in accordance with the manufacturer's protocol, using an ALP activity assay kit (Beyotime Biotechnology, cat: C3206). Total protein concentrations in the lysates (Beyotime Biotechnology, cat: P0013) were determined using the bicinchoninic acid assay (Pierce; Thermo Fisher Scientific, Inc.). Results were normalized to total protein content and expressed relative to the control condition.
The degree of mineralization was determined by ARS staining. BMSCs were seeded at a density of 1×105 cells/well in 12-well plates. After osteogenic differentiation, the cells were fixed with 95% ethanol at 25°C for 30 min, followed by incubation with 0.1% ARS solution (pH 4.2; Beijing Solarbio Science & Technology Co., Ltd.) for 20 min at room temperature. To quantify mineralization, the calcium-bound dye was solubilized using 10% cetylpyridinium chloride (Sigma-Aldrich; Merck KGaA) for 1 h in 25°C, and the eluate was assayed spectrophotometrically at 562 nm. Staining intensity was captured in light microscope and normalized to total protein content and reported relative to the undifferentiated control.
Total RNA was extracted from BMSCs using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.), followed by cDNA synthesis via RT with random primers and an M-MLV Reverse Transcriptase kit (Invitrogen; Thermo Fisher Scientific, Inc.) according to manufacturer's protocol. Subsequently, qPCR analyses were performed using a SYBR Green PCR kit (Takara Biotechnology Co., Ltd.). Thermocycling conditions were as follows: 94°C, 30 sec; 55°C, 30 sec. Step 3: 72°C, 1 min). GAPDH expression was used for normalization. The ΔCq values were calculated relative to GAPDH, and relative quantification of gene expression was determined using the 2−ΔΔCq method (24). Each sample was assessed in triplicate. The primers used are shown in Table I.
Cell protein was obtained using lysis buffer (Beyotime Biotechnology, cat: P0013) and quantified by BCA method. Protein lysates (30 µg/lane) underwent electrophoretic separation on 10% SDS-polyacrylamide gels followed by wet transfer to PVDF membranes (Sigma-Aldrich; Merck KGaA). Membranes were then blocked for 1 h in 0.1% TBS-Tween (TBST) containing 5% non-fat dry milk at room temperature. Primary antibody (FOXJ3: Solarbio, Cat: K008825P. active β-catenin: Solarbio, Cat: K009589P. β-catenin: Solarbio, Cat: K008788P. AKT: Solarbio, Cat: K109232P. p-AKT: Solarbio, Cat: Cat:K000186M. ERK: Solarbio, Cat: K200062M. p-ERK: Solarbio, Cat: K009730P. GAPDH: Solarbio, cat. no. K200057M) incubation was performed overnight at 4°C (1:1,000). After washing, the blots were incubated for 1 h at room temperature with a HRP-linked goat anti-rabbit secondary antibody (1:1,000) (Solarbio, Cat. no. SE132 and SE134). Following three 5-min TBST washes, protein bands were treated by ECL kit (Beyotime Biotechnology, cat: P0018S) and detected by enhanced chemiluminescence after substrate application. GAPDH blotting served as the normalization control. ImageJ software (National Institutes of Health, V1.47) is used for protein quantification.
A lentiviral vector encoding FOXJ3 (lentiviral vector backbone: pCDH-EF1a-MCS-IRES-puro; OE-FOXJ3) was generated by Guangzhou iGene Biotechnology Co., Ltd. In short, 293T cells (Guangzhou iGene Biotechnology Co.) were co-transfected using a third-generation lentiviral system, with plasmid ratios of 4 µg (target plasmid): 3 µg (psPAX2): 1 µg (pMD2.G). Virus supernatants were collected in batches at 48 and 72 h after transfection and filtered through a 0.45 µm filter membrane. The virus particles were concentrated by ultracentrifugation (~70,000-100,000 × g, 2 h) in 4°C, and the precipitate was resuspended in a 500 µl of buffer. Finally, the samples were aliquoted and stored at −80°C. When cells reached 80–90% confluency, lentiviral transduction was performed for 24 h at 37°C. The viral supernatant was diluted in complete medium to achieve a multiplicity of infection of 10 and was applied to cells supplemented with polybrene (8 µg/ml; Sigma-Aldrich; Merck KGaA) in 24 h. The negative control was prepared by transducing the cells with the lentiviral vector backbone lacking the target gene. Transduction efficiency was assessed by RT-qPCR analysis of FOXJ3 mRNA levels. Subsequent experiments were performed in 24 h later.
Gene silencing was performed using FOXJ3-targeting siRNAs (Shanghai GenePharma Co., Ltd.), with a non-targeting scrambled siRNA (Shanghai GenePharma Co., Ltd.) used as a negative control. Transfection of 1×105 BMSCs was carried out using 50 nM siRNA with Lipofectamine® RNAiMAX (Invitrogen; Thermo Fisher Scientific, Inc.) according to standard procedures in 37°C in 6 h. Subsequent experiments were performed in 6 h later. The siRNA sequences are shown in Table II.
All data were obtained from experiments repeated at least three times. Results are presented as the mean ± standard deviation. All statistical analyses were conducted using SPSS 18.0 (IBM Corp.). Paired Student's t-test was used for two-group comparisons, whereas one-way ANOVA with Tukey's HSD post hoc multiple comparisons test applied for multi-group analyses. The Pearson correlation test was performed for correlation analyses. P<0.05 was considered to indicate a statistically significant difference.
Firstly, osteogenic differentiation of BMSCs was induced and dynamic changes in FOXJ3 expression were detected during this differentiation process. The results of ARS staining indicated that BMSCs were effectively differentiated into osteoblasts after a 14-day culture in osteogenic induction medium (Fig. 1A). RT-qPCR results demonstrated that FOXJ3 expression progressively increased with prolonged osteogenic induction time, reaching peak levels on day 10 of induction with a ~2-fold increase compared with that in the non-induced group (Fig. 1B). Furthermore, RT-qPCR analysis revealed a positive correlation between FOXJ3 expression and the expression of osteogenesis-related genes Runt-related transcription factor 2 (RUNX2) (Fig. 1C) and osteocalcin (OCN) (Fig. 1D). Additionally, western blotting demonstrated a progressive elevation in FOXJ3 protein expression with extended osteogenic induction, reaching a maximum on day 10 of induction (Fig. 1E and F). These findings collectively suggested that FOXJ3 may be positively associated with osteogenic differentiation of BMSCs and could serve a regulatory role in BMSCs osteogenic differentiation processes.
To investigate the regulatory role of FOXJ3 in BMSCs osteogenic differentiation, BMSCs were transfected with siRNA to knock down FOXJ3 expression. The results of RT-qPCR demonstrated that siFOXJ3-1 and siFOXJ3-2 exhibited significant knockdown efficiencies, whereas siFOXJ3-3 showed no substantial effect compared to the siControl group (Fig. 2A). Western blotting further confirmed that siFOXJ3-1 and siFOXJ3-2 effectively reduced FOXJ3 protein expression in BMSCs (Fig. 2B and C). Therefore, siFOXJ3-1 and siFOXJ3-2 for the subsequent experiments. Following FOXJ3 knockdown, osteogenic differentiation was induced in BMSCs. Quantitative ALP analysis revealed decreased ALP activity in both siFOXJ3-1 and siFOXJ3-2 groups compared with that in the control, indicating that FOXJ3 knockdown suppressed ALP activity in BMSCs (Fig. 2D). ARS staining showed that the numbers of osteogenic nodules in the siFOXJ3-1 and siFOXJ3-2 groups were markedly reduced compared with those in the control group (Fig. 2E). Quantitative analysis of ARS staining further confirmed that knockdown of FOXJ3 expression inhibited osteogenic differentiation of BMSCs (Fig. 2F). RT-qPCR demonstrated that the expression of osteogenic differentiation-related genes RUNX2 and OCN was suppressed following FOXJ3 knockdown (Fig. 2G). These findings collectively demonstrated that FOXJ3 knockdown may impair the osteogenic differentiation potential in BMSCs.
To further elucidate the regulatory role of FOXJ3 in osteogenic differentiation, lentiviral infection was used to overexpress FOXJ3 in BMSCs. The results of RT-qPCR showed that the expression levels of FOXJ3 in BMSCs were significantly increased after lentiviral infection (Fig. 3A). Western blotting also revealed that FOXJ3 protein expression was elevated in the OE-FOXJ3 group compared with that in the control group (Fig. 3B). Protein semi-quantification demonstrated a ~2-fold increase in protein expression in the OE-FOXJ3 group relative to the control group (Fig. 2C). Following FOXJ3 OE, osteogenic differentiation was further induced in BMSCs. Quantitative detection of ALP revealed that ALP activity in the OE-FOXJ3 group was significantly increased compared with that in the control group (Fig. 3D). ARS staining results demonstrated a marked increase in osteogenic nodules within the OE-FOXJ3 group relative to the control group (Fig. 3E), and quantitative analysis of ARS staining further confirmed that FOXJ3 overexpression enhanced osteogenic differentiation of BMSCs (Fig. 3F). Additionally, RT-qPCR revealed that the expression levels of the osteogenic differentiation-related genes RUNX2 and OCN were upregulated following FOXJ3 overexpression (Fig. 3G). These findings collectively demonstrated that FOXJ3 gain-of-function may promote osteogenic differentiation in BMSCs.
To investigate the mechanism by which FOXJ3 regulates BMSC osteogenic differentiation, osteogenic differentiation was induced after knocking down FOXJ3 expression, and the expression levels of proteins in common osteogenic differentiation pathways, including the Wnt/β-catenin, PI3K/AKT and MAPK/ERK pathways, were examined via western blotting. The results revealed that the expression levels of active β-catenin were decreased in the siFOXJ3-1 and siFOXJ3-2 groups compared with those in the control group, whereas p-AKT and p-ERK expression showed no significant differences (Fig. 4A and B). These results indicated that FOXJ3 may primarily promote BMSCs osteogenic differentiation by regulating the Wnt/β-catenin pathway.
To further elucidate the role of the Wnt/β-catenin pathway in FOXJ3-mediated regulation of BMSCs osteogenic differentiation, rescue experiments were performed using pathway-specific inhibitors or agonists. Western blotting initially confirmed alterations in the expression levels of proteins associated with the Wnt/β-catenin pathway following combined FOXJ3 knockdown and treatment with SB216763, a Wnt/β-catenin pathway agonist. The results showed that the expression of active β-catenin in the siFOXJ3 group was decreased compared with that in the siControl group; however, after the addition of the pathway agonist SB216763, levels of active β-catenin were increased to levels comparable with the control (Fig. S1A and B). Quantitative ALP analysis revealed that ALP activity was reduced in the siFOXJ3 group compared with that in the siControl group, whereas the addition of the pathway agonist SB216763 significantly enhanced ALP activity (Fig. 5A). Furthermore, ARS staining and quantification demonstrated fewer osteogenic nodules in the siFOXJ3 group compared with that in the siControl group, whereas the addition of the pathway agonist SB216763 restored the osteogenic differentiation capacity of BMSCs (Fig. 5B and C).
Furthermore, after overexpressing FOXJ3, the findings were further validated using the Wnt/β-catenin pathway inhibitor XAV939. Western blotting demonstrated that active β-catenin expression was elevated in the OE-FOXJ3 group compared with that in the vector group; however, this enhancement was reversed following treatment with the pathway inhibitor XAV939, restoring active β-catenin expression to control levels (Fig. S1C and D). ALP activity was significantly enhanced in the OE-FOXJ3 group relative to the vector group, whereas this effect was attenuated upon pathway inhibitor treatment (Fig. 5D). ARS staining and quantification showed increased osteogenic nodule formation in the OE-FOXJ3 group compared with that in the vector group, whereas this pro-osteogenic effect was abolished in the OE-FOXJ3 + XAV939 group, with nodule formation returning to baseline control levels (Fig. 5E and F). Therefore, these results indicated that FOXJ3 could regulate the osteogenic differentiation of BMSCs in a Wnt/β-catenin pathway-dependent manner.
The present study provided compelling evidence establishing the transcription factor FOXJ3 as a novel and important positive regulator of osteogenic differentiation in BMSCs and identified its crucial dependence on the Wnt/β-catenin signaling pathway. The findings suggested the potential role of FOXJ3 in in the development of osteoporosis, offering a promising novel target for therapeutic intervention.
The pivotal role of BMSCs in maintaining bone homeostasis and their dysfunction in osteoporosis is well-established (25). As multipotent progenitors residing in the bone marrow, BMSCs possess the capacity to differentiate into osteoblasts, the bone-forming cells essential for skeletal integrity and repair (26,27). In osteoporosis, an age-related imbalance occurs where the commitment of BMSCs shifts from osteogenesis towards adipogenesis, coupled with a general decline in their osteogenic potential and proliferative capacity (28). Previous studies have indicated that numerous transcription factors (including RUNX2 and Osterix/SP7) are master regulators of osteogenesis (29,30). FOXJ3, a member of the FOX family of transcription factors, which are characterized by a conserved winged-helix DNA-binding domain, represents a hitherto unrecognized regulator of cell differentiation (13). Although FOXJ3 has been implicated in other biological processes, such as spermatogenesis and cellular stress responses (12), its specific functions in bone metabolism and BMSC biology have remain unexplored. Notably, some other FOX members, such as FOXO1, have been implicated in the oxidative stress response in bone, and FOXC2 has been shown to be involved in BMP2 signaling (31), thus indicating that FOXJ3 may also be involved in osteogenesis.
The present study first revealed that FOXJ3 was upregulated during in vitro osteogenic differentiation, and further results indicated that a positive association existed between FOXJ3 and osteogenic differentiation of BMSCs. Furthermore, the siRNA-mediated knockdown of FOXJ3 resulted in a marked suppression of the osteogenic potential of BMSCs. Conversely, lentiviral overexpression of FOXJ3 robustly enhanced osteogenesis. These findings are important in identifying FOXJ3 as a novel modulator of BMSCs osteogenesis. While previous studies have explored factors such as microRNAs (32), long non-coding RNAs (33) and epigenetic regulators (34) in BMSC osteogenesis, the identification of the role of a transcription factor such as FOXJ3 may provide a novel mechanism and potential target. The present results demonstrated that manipulating FOXJ3 levels alone was sufficient to markedly alter the osteogenic differentiation trajectory of BMSCs, highlighting its potency as a regulator. FOXJ3, alongside other identified positive regulators of BMSCs osteogenesis, such as specific isoforms of Dlk1, may expand the known factors that potentially manipulate bone formation (35). Moreover, investigating FOXJ3 expression in well-characterized human osteoporosis cohorts, particularly its association with bone mineral density, fracture history or response to existing therapies, represents a critical next step to validate its clinical relevance. Such studies could further establish FOXJ3 as a potential diagnostic biomarker or therapeutic target in osteoporosis.
The canonical Wnt pathway is a well-established and powerful promoter of osteoblast differentiation and bone formation (36). Here, FOXJ3 knockdown specifically reduced the levels of active (non-phosphorylated) β-catenin, while leaving the PI3K/AKT and MAPK/ERK pathways unaffected. The selective impact of FOXJ3 on Wnt/β-catenin signaling suggests a focused regulatory mechanism. This finding is consistent with the results of previous studies emphasizing the critical role of precise Wnt pathway modulation in bone anabolism and its therapeutic exploitation (37,38). For example, romosozumab, an anti-sclerostin antibody that enhances Wnt signaling, has been shown to exert notable efficacy in treating patients with osteoporosis (39). The finding that FOXJ3 acts upstream of β-catenin activation adds a novel layer to this complex regulatory network. Previous studies have suggested that FOXJ3 can act as a recruited transcription factor to promote osteoclast formation (40,41). If both osteoclasts and osteoblasts exist in vivo, FOXJ3 may have regulatory effects on both types of cells. Whether it promotes or inhibits osteoporosis depends on whether its effect on bone formation is greater than that on bone resorption. This not only involves the quantity of osteoblasts and osteoclasts, but also the activity of the cells and their proportion of their roles in bone formation. The present study lacks animal experiments; therefore, whether FOXJ3 will aggravate osteoporosis remains unknown. To assess this, research using high-quality tools, such as gene knockout mice, is needed.
The rescue experiments in the present study demonstrated the pathway dependence and enhance the impact of the study. The use of the specific Wnt/β-catenin agonist SB216763 effectively reversed the inhibitory effects of FOXJ3 knockdown on β-catenin activation, ALP activity and mineralization. Conversely, the pro-osteogenic effects of FOXJ3 overexpression were negated by the Wnt pathway inhibitor XAV939. These experiments indicated that the ability of FOXJ3 to promote BMSC osteogenic differentiation requires a functional Wnt/β-catenin pathway, thus integrating FOXJ3 into a well-characterized and therapeutically relevant signaling axis. However, the exact molecular mechanism by which FOXJ3 regulates β-catenin activation remains to be fully determined, which is a promising direction for future research.
Placing the current findings within the broader context of osteoporosis research underscores their potential importance. Osteoporosis therapies have traditionally focused on anti-resorptive agents (such as bisphosphonates and denosumab) (42), however, while they are effective, these treatments primarily prevent bone loss rather than robustly rebuild bone. The development of true bone-forming (anabolic) agents, such as teriparatide [a parathyroid hormone (PTH) analogue], abaloparatide (a PTH-associated protein analogue) and the aforementioned romosozumab, represents a major advance (43). However, limitations remain, including cost, administration routes and potential side effects (44). Identifying novel upstream regulators such as FOXJ3, which positively drives osteogenesis through a fundamental anabolic pathway (Wnt/β-catenin), provides novel options for therapeutic development. Strategies may involve small molecules or biologics designed to enhance FOXJ3 expression or activity directly within BMSCs or osteoprogenitors, or gene therapy approaches. This approach aligns with the growing interest in stem cell-based therapies and targeting stem cell dysfunction in age-associated diseases such as osteoporosis (45–47). Enhancing the intrinsic osteogenic potential of endogenous BMSCs via FOXJ3 modulation could offer a powerful strategy for bone regeneration. Moreover, future studies should include in vivo models, such as FOXJ3-knockout mice or local injection of FOXJ3-modulating vectors in osteoporotic animal models, to further validate its role.
In conclusion, the present study advances the understanding of the molecular control of BMSC osteogenic differentiation and the pathogenesis of osteoporosis. Robust mechanistic evidence was provided demonstrating that FOXJ3 exerts its pro-osteogenic effects primarily, if not exclusively, through the potent Wnt/β-catenin signaling pathway. This dependency was conclusively proven through targeted pathway rescue experiments. The integration of functional cellular assays and mechanistic pathway analysis provided a strong foundation for considering FOXJ3 as a promising new molecular target for the development of novel anabolic therapies aimed at restoring bone formation in osteoporosis and other bone-deficit conditions. While the present study provided strong evidence for the role of FOXJ3 in vitro and its clinical association, certain limitations warrant mention and guide future research. First, the findings were based on in vitro models, which may not fully recapitulate the complex bone microenvironment. Second, the precise molecular mechanism by which FOXJ3 regulates β-catenin remains unclear. Third, clinical patient-derived data, to assess the association between FOXJ3 expression and osteoporosis severity or treatment outcomes, were not included. Thus, future research focusing on in vivo validation and detailed mechanistic assessment will be crucial to fully realize the therapeutic potential of targeting the FOXJ3-Wnt/β-catenin axis, and to confirm the role of FOXJ3 in osteoporosis and its translational potential.
Not applicable.
The present study was supported by the Zhuhai Xiangshan Talent Project (grant no. 2021XSYC-01) and the Supporting Project of Natural Science Foundation of China (grant no. PT8217140653).
The data generated in the present study may be requested from the corresponding author.
HX performed the cell experiments and wrote the initial manuscript and submitted the paper for publication. JL contributed to some cell experiments. WH conducted the statistical analysis of the data. YQ conceived the study, supervised the research and revised the manuscript. HX and JL confirm the authenticity of all the raw data. All authors read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Jin J: Screening for osteoporosis to prevent fractures. JAMA. 333:5472025. View Article : Google Scholar : PubMed/NCBI | |
|
Reid IR and Billington EO: Drug therapy for osteoporosis in older adults. Lancet. 399:1080–1092. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang B, He W, Pei Z, Guo Q, Wang J, Sun M, Yang X, Ariben J, Li S, Feng W, et al: Plasma proteins, circulating metabolites mediate causal inference studies on the effect of gut bacteria on the risk of osteoporosis development. Ageing Res Rev. 101:1024792024. View Article : Google Scholar : PubMed/NCBI | |
|
Yin JQ, Zhu J and Ankrum JA: Manufacturing of primed mesenchymal stromal cells for therapy. Nat Biomed Eng. 3:90–104. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Dalle Carbonare L, Cominacini M, Trabetti E, Bombieri C, Pessoa J, Romanelli MG and Valenti MT: The bone microenvironment: New insights into the role of stem cells and cell communication in bone regeneration. Stem Cell Res Ther. 16:1692025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang L, Yuan X, Song R, Yuan Z, Zhao Y and Zhang Y: Engineered 3D mesenchymal stem cell aggregates with multifunctional prowess for bone regeneration: Current status and future prospects. J Adv Res. Apr 11–2025.doi: 10.1016/j.jare.2025.04.008 (Epub ahead of print). | |
|
Wu KC, Chang YH, Ding DC and Lin SZ: Mesenchymal stromal cells for aging cartilage regeneration: A review. Int J Mol Sci. 25:129112024. View Article : Google Scholar : PubMed/NCBI | |
|
Artamonov MY and Sokov EL: Intraosseous delivery of mesenchymal stem cells for the treatment of bone and hematological diseases. Curr Issues Mol Biol. 46:12672–12693. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Ban JY, Park HJ, Kim SK, Kim JW, Lee YA, Choi IA, Chung JH and Hong SJ: Association of forkhead box J3 (FOXJ3) polymorphisms with rheumatoid arthritis. Mol Med Rep. 8:1235–1241. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Ni L, Xie H and Tan L: Multiple roles of FOXJ3 in spermatogenesis: A lesson from Foxj3 conditional knockout mouse models. Mol Reprod Deve. 83:1060–1069. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Jin J, Zhou S, Li C, Xu R, Zu L, You J and Zhang B: MiR-517a-3p accelerates lung cancer cell proliferation and invasion through inhibiting FOXJ3 expression. Life Sci. 108:48–53. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Challagundla KB, Pathania AS, Chava H, Kantem NM, Dronadula VM, Coulter DW and Clarke M: FOXJ3, a novel tumor suppressor in neuroblastoma. Mol Ther Oncol. 33:2009142025. View Article : Google Scholar : PubMed/NCBI | |
|
Huang J, Zhang Y, Zhou X, Song J, Feng Y, Qiu T, Sheng S, Zhang M, Zhang X, Hao J, et al: Foxj3 Regulates thermogenesis of brown and beige fat via induction of PGC-1α. Diabetes. 73:178–196. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Yuan L, Jiang N, Li Y, Wang X and Wang W: RGS1 Enhancer RNA promotes gene transcription by recruiting transcription factor FOXJ3 and facilitates osteoclastogenesis through PLC-IP3R-dependent Ca2+ response in rheumatoid arthritis. Inflammation. 48:447–463. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Ding Y and Chen Q: Wnt/beta-catenin signaling pathway: An attractive potential therapeutic target in osteosarcoma. Front Oncol. 14:14569592024. View Article : Google Scholar : PubMed/NCBI | |
|
Hosseini A, Dhall A, Ikonen N, Sikora N, Nguyen S, Shen Y, Amaral MLJ, Jiao A, Wallner F, Sergeev P, et al: Perturbing LSD1 and WNT rewires transcription to synergistically induce AML differentiation. Nature. 642:508–518. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Arya PN, Saranya I and Selvamurugan N: Crosstalk between Wnt and bone morphogenetic protein signaling during osteogenic differentiation. World J Stem Cells. 16:102–113. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Abhishek Shah A, Chand D, Ahamad S, Porwal K, Chourasia MK, Mohanan K, Srivastava KR and Chattopadhyay N: Therapeutic targeting of Wnt antagonists by small molecules for treatment of osteoporosis. Biochem Pharmacol. 230:1165872024. View Article : Google Scholar : PubMed/NCBI | |
|
Gong W, Li M, Zhao L, Wang P, Wang X, Wang B, Liu X and Tu X: Sustained release of a highly specific GSK3β inhibitor SB216763 in the PCL scaffold creates an osteogenic niche for osteogenesis, anti-adipogenesis, and potential angiogenesis. Front Bioeng Biotechnol. 11:12152332023. View Article : Google Scholar : PubMed/NCBI | |
|
Tanthaisong P, Imsoonthornruksa S, Ngernsoungnern A, Ngernsoungnern P, Ketudat-Cairns M and Parnpai R: Enhanced chondrogenic differentiation of human umbilical cord Wharton's jelly derived mesenchymal stem cells by GSK-3 inhibitors. PLoS One. 12:e01680592017. View Article : Google Scholar : PubMed/NCBI | |
|
Yao J, Wu X, Qiao X, Zhang D, Zhang L, Ma JA, Cai X, Boström KI and Yao Y: Shifting osteogenesis in vascular calcification. JCI Insight. 6:e1430232021. View Article : Google Scholar : PubMed/NCBI | |
|
Rong X, Kou Y, Zhang Y, Yang P, Tang R, Liu H and Li M: ED-71 Prevents Glucocorticoid-induced osteoporosis by regulating osteoblast differentiation via notch and Wnt/β-catenin pathways. Drug Des Devel Ther. 16:3929–3946. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yang N, Zhang X, Li L, Xu T, Li M, Zhao Q, Yu J, Wang J and Liu Z: Ginsenoside Rc promotes bone formation in Ovariectomy-induced osteoporosis in vivo and osteogenic differentiation in vitro. Int J Mol Sci. 23:61872022. View Article : Google Scholar : PubMed/NCBI | |
|
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Abdelbaset S, Mohamed Sob MA, Mutawa G, El-Dein MA and Abou-El-Naga AM: Therapeutic potential of different injection methods for bone marrow mesenchymal stem cell transplantation in Buslfan-induced male rat infertility. J Stem Cells Regen Med. 20:26–46. 2024.PubMed/NCBI | |
|
Pajarinen J, Lin T, Gibon E, Kohno Y, Maruyama M, Nathan K, Lu L, Yao Z and Goodman SB: Mesenchymal stem cell-macrophage crosstalk and bone healing. Biomaterials. 196:80–89. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Sun Y, Wan B, Wang R, Zhang B, Luo P, Wang D, Nie JJ, Chen D and Wu X: Mechanical stimulation on mesenchymal stem cells and surrounding microenvironments in bone regeneration: Regulations and applications. Front Cell Dev Biol. 10:8083032022. View Article : Google Scholar : PubMed/NCBI | |
|
Wu Y, Xie L, Wang M, Xiong Q, Guo Y, Liang Y, Li J, Sheng R, Deng P, Wang Y, et al: Mettl3-mediated m6A RNA methylation regulates the fate of bone marrow mesenchymal stem cells and osteoporosis. Nat Commun. 9:47722018. View Article : Google Scholar : PubMed/NCBI | |
|
Chan WCW, Tan Z, To MKT and Chan D: Regulation and role of transcription factors in osteogenesis. Int J Mol Sci. 22:54452021. View Article : Google Scholar : PubMed/NCBI | |
|
Komori T: Regulation of skeletal development and maintenance by Runx2 and Sp7. Int J Mol Sci. 25:101022024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang W, Zhang X, Li J, Zheng J, Hu X, Xu M, Mao X and Ling J: Foxc2 and BMP2 Induce Osteogenic/odontogenic differentiation and mineralization of human stem cells from apical papilla. Stem Cells Int. 2018:23639172018. View Article : Google Scholar : PubMed/NCBI | |
|
Hong L, Sun H and Amendt BA: MicroRNA function in craniofacial bone formation, regeneration and repair. Bone. 144:1157892021. View Article : Google Scholar : PubMed/NCBI | |
|
Jin C, Jia L, Huang Y, Zheng Y, Du N, Liu Y and Zhou Y: Inhibition of lncRNA MIR31HG promotes osteogenic differentiation of human Adipose-derived stem cells. Stem Cells. 34:2707–2720. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Liu S, Liu D, Chen C, Hamamura K, Moshaverinia A, Yang R, Liu Y, Jin Y and Shi S: MSC Transplantation improves osteopenia via epigenetic regulation of notch signaling in lupus. Cell Metab. 22:606–618. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Paradise CR, Galvan ML, Kubrova E, Bowden S, Liu E, Carstens MF, Thaler R, Stein GS, van Wijnen AJ and Dudakovic A: The epigenetic reader Brd4 is required for osteoblast differentiation. J Cell Physiol. 235:5293–5304. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Hu L, Chen W, Qian A and Li YP: Wnt/β-catenin signaling components and mechanisms in bone formation, homeostasis, and disease. Bone Res. 12:392024. View Article : Google Scholar : PubMed/NCBI | |
|
Huybrechts Y, Mortier G, Boudin E and Van Hul W: WNT signaling and bone: Lessons from skeletal dysplasias and disorders. Front Endocrinol (Lausanne). 11:1652020. View Article : Google Scholar : PubMed/NCBI | |
|
Maeda K, Kobayashi Y, Koide M, Uehara S, Okamoto M, Ishihara A, Kayama T, Saito M and Marumo K: The regulation of bone metabolism and disorders by Wnt signaling. Int J Mol Sci. 20:55252019. View Article : Google Scholar : PubMed/NCBI | |
|
Wu D, Li L, Wen Z and Wang G: Romosozumab in osteoporosis: Yesterday, today and tomorrow. J Transl Med. 21:6682023. View Article : Google Scholar : PubMed/NCBI | |
|
Alexander MS, Shi X, Voelker KA, Grange RW, Garcia JA, Hammer RE and Garry DJ: Foxj3 transcriptionally activates Mef2c and regulates adult skeletal muscle fiber type identity. Dev Biol. 337:396–404. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, Wang Z, Duan N, Zhu G, Schwarz EM and Xie C: Osteoblast-osteoclast interactions. Connect Tissue Res. 59:99–107. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Langdahl BL: Overview of treatment approaches to osteoporosis. Br J Pharmacol. 178:1891–1906. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Khosla S and Hofbauer LC: Osteoporosis treatment: Recent developments and ongoing challenges. Lancet Diabetes Endocrinol. 5:898–907. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Shimizu R, Sukegawa S, Sukegawa Y, Hasegawa K, Ono S, Nakamura T, Fujimura A, Fujisawa A, Nakano K, Takabatake K, et al: Incidence and risk of Anti-Resorptive Agent-related osteonecrosis of the jaw after tooth extraction: A retrospective study. Healthcare (Basel). 10:13322022. View Article : Google Scholar : PubMed/NCBI | |
|
Tong Y, Tu Y, Wang J, Liu X, Su Q, Wang Y and Wang W: Mechanisms and therapeutic strategies linking mesenchymal stem cells senescence to osteoporosis. Front Endocrinol (Lausanne). 16:16258062025. View Article : Google Scholar : PubMed/NCBI | |
|
Li H and Bai L: Advances in mesenchymal stem cell and Exosome-based therapies for aging and age-related diseases. Stem Cell Res Ther. 16:4012025. View Article : Google Scholar : PubMed/NCBI | |
|
Liang B, Burley G, Lin S and Shi YC: Osteoporosis pathogenesis and treatment: Existing and emerging avenues. Cell Mol Biol Lett. 27:722022. View Article : Google Scholar : PubMed/NCBI |