Patients’ BMD correlates negatively with their serum F4BP4 levels
Ovariectomy (OVX) in mice, leading to estrogen deficiency, is well-known for causing bone loss and increased marrow adiposity. However, the expression of FABP4 in the bone marrow of OVX mice is limited reported. Our study revealed significant trabecular bone loss 30 days post-ovariectomy, as shown by Micro-CT scans (Fig. 1A), and an increase in marrow adipose tissue, confirmed by paraffin sections and H&E staining (Fig. 1, B and D). Immunohistochemistry (IHC) demonstrated a marked upregulation of FABP4 in the bone marrow cavity (Fig. 1, C and E), suggesting changes in adipocyte function and possible effects on osteoblast activity.
Subsequent investigations included measuring serum levels of FABP4 and lipid profiles (total cholesterol (TC), triglycerides (TG), HDL, and LDL) in patients with OP (T-scores ≤ − 2.5) and non-OP (T-scores ≥ − 1) trauma fracture patients (Table S1). Our results showed that, among the lipid profile components, none of them were significantly abnormal in OP patients compared to non-OP individuals (Fig. S1A), and serum FABP4 levels were notably higher in OP patients (Fig. 1F). Correlation analysis revealed a significant negative relationship between BMD and serum FABP4 levels (Fig. 1G and S1B), indicating that higher FABP4 levels are associated with lower BMD. These findings provide compelling clinical evidence for FABP4's role in OP development.
FABP4 has no impact on the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs)
Both bone marrow adipocytes and osteoblasts originate from BMSCs (26). An increase in bone marrow adiposity is associated with a decrease in bone mass (27). Initially, we hypothesized that FABP4 might inhibit osteogenic differentiation by promoting BMSCs to differentiate into adipocytes. To test this, we supplemented recombinant human FABP4 protein (25ng/mL or 50 ng/mL) during the 21-day induction of human BMSCs towards osteogenic differentiation, along with the concurrent addition of FABP4 protein and its inhibitor BMS309403 (5 µM or 10 µM) (Fig. 2A). Surprisingly, Alizarin Red staining revealed that neither the FABP4 protein nor its inhibitor affected the osteogenic differentiation capacity of BMSCs (Fig. 2B).
This led us to consider whether increased bone marrow adiposity elevates endogenous FABP4 levels within BMSCs, thereby reducing their osteogenic differentiation potential. To investigate this, we established stable BMSC cell lines overexpressing FABP4 (Fig. 2, C and D), achieving nearly a 4000-fold increase in FABP4 gene expression (Fig. 2E). However, when inducing osteogenic differentiation in these FABP4-overexpressing BMSCs, with or without BMS309403 (10 µM), the Alizarin Red staining results remained unchanged, indicating that FABP4 overexpression did not impact the osteogenic differentiation of BMSCs (Fig. 2, F and G).
These findings demonstrate that both endogenous and exogenously elevated levels of FABP4 do not hinder osteogenic differentiation. While this result may be disheartening, it directs us towards further research, suggesting that FABP4 might influence OCs differentiation.
FABP4 plays a crucial role in enhancing OCs differentiation
To assess the impact of exogenous FABP4 on the differentiation of bone marrow-derived macrophages (BMMs) into OCs, we first induced primary mouse bone marrow single-nucleus cells (BMNCs) into BMMs using M-CSF (28). During the subsequent induction of BMMs into OCs with RANKL, we introduced recombinant mouse FABP4 and its inhibitor BMS309403. Trap staining results revealed that the addition of just 25 ng/mL FABP4 significantly promoted the formation of OCs, and this effect could be reversed by BMS309403 (Fig. 3, A and B).
To further investigate the effect of increased endogenous FABP4 on OCs formation, we added a mixture of free fatty acids (FFA) to the culture medium (29). Western blot results indicated a significant increase in intracellular FABP4 expression in BMMs with 200X and 400X diluted FFA (Fig. 3, C and D). The subsequent addition of FFA and FABP4 inhibitors during OCs differentiation showed that FFA alone significantly increased the number of differentiated OCs, an effect that could be reversed by BMS309403 (Fig. 3, E and F).
Confocal microscopy images of F-actin staining in OCs revealed a more regular and distinct bone skeleton structure with abundant F-actin when stimulated by additional FABP4 or FFA compared to the group with only RANKL and M-CSF (Fig. S2A). Furthermore, we confirmed the influence of FABP4 on the bone-resorptive capabilities of mature OCs on bovine bone slices. The addition of FFA or FABP4 enhanced bone resorption, whereas the FABP4 inhibitor effectively blocked this process (Fig. 3G and Fig. S2B). All of these experiments robustly demonstrate that FABP4 significantly promotes the differentiation of OCs and enhances bone resorption.
FABP4 inhibitor BMS309403 shows comparable OCs differentiation inhibition to ALD
Given the significant expression of FABP4 in BMMs cells (Fig. 3C), we postulated that an FABP4 inhibitor could inhibit OCs differentiation even without external stimuli. The initial CCK8 assay revealed that BMS309403, at concentrations up to 50 µM, did not hinder the proliferation of BMMs cells (Fig. S3A). Subsequently, the successful validation of our hypothesis through trap staining after exposure to different doses of BMS309403 underscores the intricate role of FABP4 in OCs differentiation (Fig. 3H and Fig. S3B). Remarkably, the in vitro efficacy of BMS309403 in inhibiting OCs, comparable to that of the established clinical therapy ALD (Fig. S3, C and D), reinforces its therapeutic promise. With an IC50 value of approximately 0.89 µM for BMS309403 and 0.44 µM for ALD (Fig. 3I), these results demonstrate the potency of BMS309403 in regulating OCs function.
FABP4 inhibitor suppresses OCs differentiation via Ca-Calcineurin-NFATc pathway
To further investigate the mechanism by which FABP4 inhibitors impede OCs differentiation, we performed transcriptome sequencing on three groups: BMMs cells, BMMs-induced OCs, and OCs treated with 1µM BMS309403 during induction (OCs + BMS). We initially analyzed differentially expressed genes using volcano plots for BMMs vs. OCs and OCs vs. OCs + BMS (Fig. 4A). A Venn diagram identified 107 genes upregulated 2-fold in BMMs-induced OCs and 284 genes downregulated 0.3-fold in OCs post-BMS309403 treatment, converging on 69 common genes for KEGG pathway analysis (Fig. 4B) and GO analysis (Fig. S4). The most affected KEGG pathways were OCs differentiation and calcium signaling (Fig. 4C). Cluster analysis of FPKM values (Fig. 4D) and subsequent qPCR validation (Fig. 4E) revealed significant downregulation of key genes (Oscar, Acp5, Fos, Nfatc1) following BMS309403 treatment. This was corroborated by Western Blotting results (Fig. 4F), which demonstrated suppression of TRAP, c-Fos, and NFATc1 protein levels (Fig. 4G), thus confirming that FABP4 inhibitors suppress OCs differentiation at both the transcriptional and translational levels. Regarding the critical role of the calcium signaling pathway highlighted by KEGG, we examined intracellular calcium levels in OCs, OCs + FABP4, OCs + FABP4 + BMS309403, and OCs + BMS309403 using the calcium fluorescent probe Fluo-4 AM via flow cytometry (Fig. 4H) and fluorescence microscopy (Fig. 4I). Results showed a significant increase in intracellular calcium levels in OCs with FABP4, which was reversed by BMS309403.
The calcium signaling pathway is crucial in OCs differentiation (30), whereby RANKL-RANK interaction activates downstream PLCγ, leading to elevated intracellular calcium concentrations and subsequent Calcineurin activation (31). This cascade dephosphorylates NFATc1, regulating transcription of key proteins such as TRAP, CTSK, and OSCAR in the nucleus (32). Our transcriptome analysis indicated no change in Plcγ gene expression post-BMS309403 treatment, supported by Western Blotting showing constant PLCγ levels, while the Calcineurin A and B subunits were significantly downregulated (Fig. 4, J and K). This suggests that FABP4 elevates calcium levels in OCs, thereby activating the Ca2+-Calcineurin-NFATc1 pathway to promote OCs differentiation, thus expanding our understanding of this cellular process.
Effects of FABP4 inhibitor on bone resorption in OVX mouse OP model
To further validate the efficacy of FABP4 inhibitors in combating OP in vivo, we established a bilateral ovariectomy-induced OP mouse model to simulate postmenopausal OP. Seven days post-surgery, mice were divided into four groups: Sham, Model, ALD (positive control), and BMS309403. The Sham group underwent minor fat tissue removal around the ovaries. After 28 days of daily oral gavage, mice were euthanized, and serum, uterus, tibia, and femur samples were collected for further analysis (Fig. 5A). Uterine atrophy confirmed the success of the surgical model (Fig. S5A). Dynamic weight changes showed a significant increase in body weight post-ovariectomy, with slight decreases observed in the ALD and BMS groups compared to the Model group, maintaining overall stability (Fig. S5B). ELISA analysis revealed a significant increase in serum FABP4 levels in the untreated Model group, while FABP4 levels notably decreased in the ALD and BMS groups (Fig. 5B). These results reaffirmed the close association between FABP4 levels and OP progression. Subsequently, we assessed serum levels of the bone formation marker PINP (Fig. 5C) and the bone resorption marker CTX1 (Fig. 5D). Similar to ALD, FABP4 inhibitors had no effect on bone formation but exhibited inhibitory effects on bone resorption. Micro-CT assessment of bone mass at the distal femur demonstrated significant inhibition of OP progression in ovariectomized mice treated with FABP4 inhibitors (Fig. 5E). 3D analysis indicated that oral administration of FABP4 inhibitors significantly increased BMD (Fig. 5F) and trabecular thickness (Tb.Th, Fig. 5G). Results from TRAP staining further confirmed the inhibitory effect of BMS309403 on bone resorption in vivo (Fig. 5, I and J). Additionally, HE staining (Fig. S5, C and D)and IHC analysis of FABP4 protein expression (Fig. S5, E and F) in bone marrow fat tissue revealed no significant difference between the BMS and ALD treatments, suggesting that the lack of impact on osteogenic and adipogenic differentiation of BMSCs may account for this.
Differential efficacy of FABP4 inhibitor and ALD in OVX mouse OP model
FABP4 protein is extensively distributed across various tissues in the body, including adipose tissue, liver, intestines, heart, and muscles (33). Consequently, developing FABP4 inhibitors presents targeting challenges, as these inhibitors may affect FABP4 in multiple tissues, leading to unpredictable systemic effects. Furthermore, the pharmacokinetic properties of these inhibitors could be influenced by the ubiquitous expression of FABP4, impacting their efficacy and safety.
In our in vivo study, the oral dose of ALD (1.3 mg/kg) followed clinical standards (7), whereas the dose of BMS309403 (10 mg/kg) was based on prior preclinical doses for diabetes treatment (34). Although BMS309403 demonstrated comparable activity to ALD in vitro, in vivo results showed that even at 7.7 times the dose of ALD, BMS309403 did not achieve the same efficacy. Specifically, compared to ALD, FABP4 inhibitors had no significant effect on trabecular number (Tb.N, Fig. 5H) and bone volume/tissue volume ratio (BV/TV, Fig. S5G) in OVX mice. Additionally, Western blot analysis of various OVX mice tissues revealed substantial FABP4 presence in muscles, white adipose tissue (WAT), brown adipose tissue (BAT), heart, and kidneys, while bone marrow showed minimal presence (Fig. S6). This highlights the critical need to enhance the bone-targeting capabilities of FABP4 inhibitors for their potential clinical application in treating OP.
Developing novel bone-targeted PLGA nanoparticles with click chemistry
The structural similarity of bisphosphonates to inorganic phosphates in bone allows them to establish strong bonds with hydroxyapatite (HA) crystals on bone surfaces. Studies have shown that bisphosphonate-modified PLGA-PEG nanoparticles (NPs) enable controlled drug release, proving effective in addressing diverse bone conditions (35). In this study, bisphosphonate-conjugated PLGA-PEG (PLGA-PEG-Ald) was synthesized via click chemistry using PLGA-PEG-N3 and DBCO-ALD (Fig. 6A). The structure was confirmed by FT-IR (Fig. S7) and 1H NMR (Fig. S8). Previous research has demonstrated that NPs assembled from polymers with 20% PLGA-PEG-Ald exhibit superior stability and bone-targeting ability (35). Building on these findings, incorporating the FABP4 inhibitor BMS309403 into Ald-BMS-NPs, composed of 20% PLGA-PEG-Ald and 80% PLGA-PEG, shows promise, with NPs made entirely of PLGA-PEG referred to as PLGA-BMS-NPs (Fig. 6B).
The encapsulation efficiency of BMS309403 in Ald-BMS-NPs and PLGA-BMS-NPs is 43.94% and 30.43%, respectively, with drug loading rates of 4.21% and 2.95%, significantly higher than reported in existing literature (35). The particle surface Zeta potential of Ald-BMS-NPs is approximately − 21.6 mV, while that of PLGA-BMS-NPs is around − 19.0 mV, indicating successful enrichment of bisphosphonate ions on Ald-BMS-NPs' surface (Fig. 6C). The particle size of Ald-BMS-NPs is uniform, with an average size of about 183.3 ± 2.8nm (Fig. 6D), slightly larger than that of PLGA-BMS-NPs at 183.3 ± 2.8nm (Fig. 6E).
Then in vitro CCK-8 experiments showed that different concentrations of Ald-BMS-NPs, PLGA-BMS-NPs, and empty Ald-NPs did not exhibit cytotoxicity towards BMMs cells (Fig. S9A). The BMS309403 encapsulated in Ald-BMS-NPs demonstrated slow release in physiological saline at pH 7.0 for at least 72 hours, with a slightly faster release rate compared to PLGA-BMS-NPs (Fig. 6F). By introducing a small amount of fluorescent molecule Cy5 into both NPs, Ald-BMS-Cy5-NPs and PLGA-BMS-Cy5-NPs were obtained. Under confocal microscopy, both types of nanoparticles were observed to be readily taken up by BMSCs after 30 minutes (Fig. 6G) of co-culturing and had increased accumulation within the cells after 6 hours (Fig. S9B).
Subsequently, the bone-targeting efficacy of Ald-BMS-NPs was confirmed through both in vitro and in vivo assessments. Initially, Ald-BMS-NPs and PLGA-BMS-NPs were incubated with nano-HA for 6 hours to assess their binding capability, as visualized with TEM imaging (Fig. 6H). This revealed strong adhesion of Ald-BMS-NPs to HA. Furthermore, Cy5-labeled NPs were co-cultured with bovine bone slices, illustrating increased binding of Ald-BMS-NPs (Fig. S10A). In vivo validation using IVIS revealed Ald-BMS-NPs exhibited significantly enhanced bone targeting compared to PLGA-BMS-NPs (Fig. S10B and C). IVIM imaging at 24- and 48-hours post-injection highlighted preferential accumulation of Ald-BMS-Cy5-NPs in the bone marrow, underscoring their potent bone-targeting capabilities (Fig. S10D).
In conclusion, our click-chemistry-derived PLGA nanoparticles demonstrate precise bone and bone marrow targeting, uniform size, efficient drug loading, controlled release, and are tailored for targeted FABP4 inhibitor delivery in vivo.
Bone-targeted NPs delivery of FABP4 inhibitor shows superior anti-OP effect in OVX mouse model
To further validate the in vivo therapeutic efficacy of bone-targeted Ald-BMS-NPs loaded with FABP4 inhibitors, we re-established the OVX-induced osteoporotic mouse model. Seven days post-bilateral ovariectomy, C57BL/6J mice were divided into groups: Sham (saline), Model (saline), ALD (10 mg/kg), Free BMS309403 (10 mg/kg), Ald-BMS-NPs (10 mg/kg of BMS), and empty Ald-NPs (equivalent PLGA content to Ald-BMS-NPs). All groups received bi-weekly intraperitoneal injections over four weeks (Fig. 7A). Body weight was monitored bi-daily throughout the treatment period (Fig. S11). At the conclusion of the experiment, tibia were subjected to Micro-CT analysis. BMD data revealed no improvement in the OVX group treated with empty Ald-NPs. In contrast, intraperitoneal BMS309403 significantly increased BMD. Notably, the Ald-BMS-NPs group exhibited significant BMD enhancement, with BMD values approximating those of the Sham group (Fig. 7B). Micro-CT reconstruction figures indicated reduced trabecular bone loss and increased bone mass in the Ald-BMS-NPs group compared to the ALD and Free BMS309403 groups (Fig. 7C). The 3D analysis data further demonstrated significant improvements in Tb.Sp (Fig. 7D), BV/TV (Fig. 7E), and Tb.N (Fig. 7F) in the Ald-BMS-NPs group. While statistically similar to the ALD group, the Ald-BMS-NPs group showed modestly superior values. Additionally, Ald-BMS-NPs significantly improved Tb.Sp (Fig. 7G), an effect not observed in the ALD group.
To assess the potential impact of NPs on liver and kidney functions, we analyzed serum markers of liver enzymes (ALT, AST, ALP, γ-GT) and kidney function indicators (UREA, CREA, UA). Results indicated no abnormalities in liver function markers across all groups, except for one mouse in the Ald-BMS-NPs group with elevated AST levels. Kidney function markers were also within normal ranges, except for increased UA levels in the ALD group (Fig. 7H). Histological examination of the heart, liver, spleen, lungs, and kidneys in all groups, using H&E staining, revealed no organic changes (Fig. S12).
These findings demonstrate that bone-targeted NPs loaded with FABP4 inhibitors not only provide excellent anti-OP effects but also exhibit high safety, offering strong evidence for the therapeutic potential of FABP4 inhibitors in OP treatment.