RESEARCH
Peer reviewed scientific video journal
Video encyclopedia of advanced research methods
Visualizing science through experiment videos
EDUCATION
Video textbooks for undergraduate courses
Visual demonstrations of key scientific experiments
BUSINESS
Video textbooks for business education
OTHERS
Interactive video based quizzes for formative assessments
Products
RESEARCH
JoVE Journal
Peer reviewed scientific video journal
JoVE Encyclopedia of Experiments
Video encyclopedia of advanced research methods
EDUCATION
JoVE Core
Video textbooks for undergraduates
JoVE Science Education
Visual demonstrations of key scientific experiments
JoVE Lab Manual
Videos of experiments for undergraduate lab courses
BUSINESS
JoVE Business
Video textbooks for business education
Solutions
Language
English
Menu
Menu
Menu
Menu
A subscription to JoVE is required to view this content. Sign in or start your free trial.
Research Article
Erratum Notice
Important: There has been an erratum issued for this article. View Erratum Notice
Retraction Notice
The article Assisted Selection of Biomarkers by Linear Discriminant Analysis Effect Size (LEfSe) in Microbiome Data (10.3791/61715) has been retracted by the journal upon the authors' request due to a conflict regarding the data and methodology. View Retraction Notice
Here, we present a protocol to evaluate the inhibitory effects of the γ-secretase inhibitor LY900009 on RANKL-induced osteoclastogenesis in bone marrow-derived macrophages. The protocol includes cytotoxicity assessment, functional assays, molecular analyses, and in vivo validation in a murine osteolytic model, with applications in the therapeutic development of osteolytic diseases.
Dysregulated osteoclast activity profoundly contributes to the progression of osteolytic disease, yet the extent of therapeutic intervention modalities is constrained by adverse effects. This study investigated LY900009, a γ-secretase inhibitor, and its capacity to suppress RANKL-induced osteoclastogenesis through modulation of Notch signaling. Using bone marrow-derived macrophages, we performed comprehensive in vitro evaluations of cytotoxicity, differentiation, and functional assays. Gene and protein expression analyses using qPCR and western blotting were used to elucidate the underlying molecular mechanisms. The in vivo efficacy was corroborated using a murine lipopolysaccharide-induced bone resorption model. LY900009 inhibited RANKL-induced osteoclast formation and resorptive function in a dose-dependent manner without cytotoxicity at therapeutic concentrations. This treatment significantly reduced the number of TRAP-positive osteoclasts, impaired actin belt formation, and suppressed bone matrix degradation. The expression of osteoclast markers, including NFATc1, c-Fos, cathepsin K, and TRAP, was markedly downregulated. Mechanistically, LY900009 disrupted Notch signaling and the downstream MAPK/Akt pathways. In vivo administration substantially reduced LPS-induced bone destruction, improving bone parameters and reducing osteoclast presence. These findings demonstrate that LY900009 effectively inhibits osteoclastogenesis and bone resorption by modulating the Notch/MAPK/Akt signaling axis, making it a promising therapeutic candidate for osteolytic disorders.
Bone homeostasis constitutes an exquisitely orchestrated physiological paradigm in vertebrates, harmonized by the delicate equilibrium between osteoblast-mediated bone formation and osteoclast-driven bone resorption1. This intricate balance maintains skeletal integrity, biomechanical resilience, and metabolic fidelity throughout one's lifespan, dynamically responding to mechanical demands, endocrine perturbations, and pathological challenges2. The bone remodeling cycle involves sequential phases of activation, resorption, transition, formation, and termination, each governed by intricate molecular cascades that ensure the spatiotemporal synchronization of cellular activities3.
Disruption of this homeostatic mechanism, particularly through excessive osteoclast activity or inadequate osteoblast function, precipitates pathological bone loss, characteristic of numerous osteolytic disorders4,5. These conditions encompass a broad spectrum of diseases, including postmenopausal and senescent osteoporosis, rheumatoid arthritis, inflammatory arthritis, neoplastic bone involvement, Paget's disease, and periodontal disorders6. The clinical manifestations of excessive bone resorption include increased fracture risk, skeletal deformity, chronic pain, functional disability, and a substantial healthcare burden6,7. Contemporary epidemiological metrics indicate that osteoporosis alone affects over 200 million individuals worldwide, with associated fractures resulting in significant morbidity, mortality, and economic costs exceeding billions of dollars annually7,8.
Osteoclasts manifest as highly specialized multinucleated giant cells derived from hematopoietic precursors of the mononuclear phagocyte lineage, specifically bone marrow-derived macrophages9. Their formation represents a complex multistep process that requires the precise coordination of extracellular signals, intracellular signaling cascades, and transcriptional programs10. The essential cytokines governing osteoclastogenesis include receptor activator of nuclear factor κB ligand (RANKL) and macrophage colony-stimulating factor (M-CSF), which bind to their respective receptors RANK and c-Fms to initiate differentiation programs11. RANKL-RANK interaction triggers the activation of multiple downstream signaling pathways, including mitogen-activated protein kinases (MAPKs), nuclear factor κB (NF-κB), phosphoinositide 3-kinase/protein kinase B (PI3K/Akt), and calcium/calcineurin pathways, which converge on nuclear factor of activated T cells cytoplasmic 1 (NFATc1), the master transcriptional regulator of osteoclastogenesis10,11,12.
Among the various regulatory mechanisms that control osteoclast biology, Notch signaling has emerged as a critical modulator with complex and context-dependent functions13. The Notch pathway is an evolutionarily conserved cell-cell communication system that governs cell fate determination, differentiation, proliferation, and apoptosis across diverse tissues and developmental stages14. In mammals, four Notch receptors (Notch1-4) interact with five canonical ligands (Delta-like 1, 3, and 4, and Jagged 1 and 2) to mediate intercellular signaling. Pathway activation involves ligand binding, conformational changes, and sequential proteolytic cleavage by ADAM metalloproteases and the γ-secretase complex, which ultimately releases the Notch intracellular domain (NICD) for the nuclear translocation and transcriptional regulation of target genes, including the Hes and Hey families14,15. The role of Notch signaling in osteoclastogenesis remains contentious and appears to be highly context-dependent, with studies reporting both stimulatory and inhibitory effects depending on the specific receptor subtypes, ligand availability, cellular microenvironment, and experimental conditions16,17,18.
Prevailing therapeutic approaches for osteolytic diseases primarily focus on anti-resorptive strategies, including bisphosphonates, denosumab (RANKL inhibitor), selective estrogen receptor modulators, and calcitonin7,8,19. Although these treatments effectively reduce bone resorption and fracture risk, they are associated with significant limitations, including gastrointestinal toxicity, osteonecrosis of the jaw, atypical fractures, increased infection risk, and concerns about long-term safety. Additionally, these agents may excessively suppress bone turnover, potentially compromising bone quality and repair mechanisms. Consequently, there is an urgent clinical need for novel therapeutic agents that can selectively inhibit pathological osteoclast activity while minimizing adverse effects and preserving physiological bone-remodeling capacity8,14.
γ-Secretase inhibitors represent a promising class of compounds with potential applications in osteolytic diseases through modulation of Notch signaling14,20. These agents have been extensively studied in oncology and neurodegenerative disease research, with several compounds advancing to clinical trials for Alzheimer's disease and various cancers. LY900009, a potent and selective γ-secretase inhibitor, has demonstrated favorable pharmacological properties, including high specificity, bioavailability, and a manageable safety profile in preliminary studies21,22. However, its specific effects on RANKL-induced osteoclastogenesis and potential therapeutic utility in osteolytic diseases remain incompletely characterized. This study was designed to evaluate whether LY900009 could inhibit osteoclastogenesis through the disruption of Notch signaling, potentially offering a novel therapeutic approach for osteolytic diseases. We developed comprehensive protocols to assess their effects on osteoclast formation, bone resorption activity, signaling pathways, and in vivo efficacy in inflammatory bone loss models.
All animal procedures were conducted in accordance with institutional guidelines and were approved by the Animal Care and Use Committee of the Central Hospital affiliated with Shandong First Medical University (approval number IACUC-2024-015). The study adhered to established ethical principles for animal research and ARRIVE guidelines, including animal housing in standard cages (5 mice/cage, 12-h light/dark cycle, ad libitum food/water) and randomization to groups
1. Bone marrow-derived macrophage isolation and culture
2. Cytotoxicity assessment
3. Osteoclast differentiation and TRAP staining
4. Functional bone resorption assay
5. Immunofluorescence microscopy
6. Quantitative real-time PCR analysis
7. Western blot analysis
8. In Vivo LPS-induced bone resorption model
9. Micro-CT and histological analysis
10. Statistical analysis
The successful implementation of this protocol revealed that LY900009 exerted potent inhibitory effects on osteoclast formation and function through multiple interconnected mechanisms. Initial cytotoxicity assessment in bone marrow-derived macrophages demonstrated time-dependent increases in toxicity, with calculated IC50 values of 4.44 µM, 3.66 µM, and 2.93 µM at 24 h, 48 h, and 96 h, respectively (Figure 1A,B). Importantly, these cytotoxic concentrations were substantially higher than the therapeutically effective doses, establishing a favorable therapeutic window. The molecular structure of LY900009 is shown in Figure 1C. TRAP staining analysis revealed dose-dependent suppression of RANKL-induced osteoclast formation, with significant reductions in both osteoclast number and size at concentrations ranging from 100 to 400 nM (Figure 1D-F). Functional assessment using hydroxyapatite-coated surfaces demonstrated a corresponding impairment of bone resorptive activity, with marked decreases in resorption pit formation and total resorbed areas (Figure 2A,B). Immunofluorescence examination revealed disrupted organization of the characteristic podosomal actin belts, indicating compromised cytoskeletal architecture essential for effective bone resorption (Figure 2C,D). These morphological changes were correlated with functional deficits, confirming that LY900009 affects both osteoclast differentiation and mature cell activity.
Molecular characterization elucidated the signaling mechanisms underlying LY900009's inhibitory effects on osteoclastogenesis. Quantitative PCR analysis demonstrated significant dose- and time-dependent downregulation of critical osteoclast-associated genes, including NFATc1, c-Fos, cathepsin K, and TRAP, indicating disruption of the transcriptional programs governing osteoclast differentiation (Figure 3A,B). Western blot analysis revealed that LY900009 markedly decreased cleaved Notch1 levels and downstream target Hes1 expression, accompanied by a substantial reduction in NFATc1 protein levels, confirming disruption of the Notch signaling cascade (Figure 4A,B). Short-term treatment studies demonstrated rapid suppression of phosphorylation of multiple key signaling molecules, including ERK1/2, p38 MAPK, Akt, and NF-κB p65, indicating interference with diverse pathways downstream of RANKL signaling (Figure 4C). In vivo validation using a murine LPS-induced bone resorption model provided compelling evidence of therapeutic efficacy, and micro-CT analysis revealed significant preservation of bone architecture, increased bone volume fraction, enhanced trabecular number, and improved bone mineral density in LY900009-treated groups compared with controls (Figure 5A,B). Histological examination confirmed these findings, demonstrating reduced osteolytic lesions and substantially decreased numbers of TRAP-positive osteoclasts in calvarial sections from treated animals (Figure 5C-E), thus establishing the translational relevance of the in vitro observations.

Figure 1: RANKL-induced osteoclast formation in vitro was downregulated by LY900009. (A) Cytotoxicity in LY900009 cells. (B) The results show the IC50 of LY900009 at different time points in BMMs. (C) Chemical structure of LY900009 (provided by Selleck). (D) BMMs were simulated with different concentrations of LY900009, M-CSF (33.3 ng/mL), and RANKL (100 ng/mL) for 5 days. Cells were fixed in 4% paraformaldehyde and stained with TRAP. (E, F) The area and number of TRAP-positive cells. Data are presented as mean ± SD (* p < 0.05, ** p < 0.01, *** p < 0.001), n=3 per group. Please click here to view a larger version of this figure.

Figure 2: Podosome actin belt formation and OC-mediated bone resorption were inhibited by LY900009. (A) Osteoclasts (OCL) were seeded onto Osteo Assay Stripwell Plates with RANKL (100 ng/mL), M-CSF (33.3 ng/mL), and LY900009 (0,100, 200, and 400 nM) for 4 days. (B) The resorption area in Osteo Assay Stripwell Plates. (C) BMMs were seeded onto 48-well plates with 0, 100, 200, or 400 nM LY900009 for 5 days. Cells were fixed and stained for immunofluorescence. (D) Area of OCs with an F-actin belt. The data are presented as mean ± SD (n = 3 per group). Please click here to view a larger version of this figure.

Figure 3: Osteoclastogenesis relative gene expression was depressed by LY900009. (A) BMMs were treated with M-CSF (33.3 ng/mL) and RANKL (100 ng/mL) in the presence of 0, 100, 200, or 400 nM LY900009 for 5 days. The expression of osteoclast-specific genes, including NFATc1, Cath-K, TRAP, and c-Fos, was analyzed by quantitative real-time PCR. (B) BMMs were treated with M-CSF (33.3 ng/mL) and RANKL (100 ng/mL) in the presence of 400 nM LY900009 for 1, 3, or 5 days. Osteoclast-specific gene expression was analyzed by quantitative real-time PCR. RNA expression levels were normalized to those of GAPDH. The data were presented as the mean ± SD (* p < 0.05, ** p < 0.01, *** p < 0.001), n=3 per group. Please click here to view a larger version of this figure.

Figure 4: LY900009 suppresses osteoclastogenesis by Notch inhibition and AKT phosphorylation regulation. (A) LY900009 treatment suppressed the expression of cleaved Notch1. (B) LY900009 treatment suppressed the expression of Hes1 and NFATc1. (C) LY900009 treatment suppressed AKT phosphorylation. The data were presented as the mean ± SD (* p < 0.05, ** p < 0.01, *** p < 0.001), n=3 per group. Please click here to view a larger version of this figure.

Figure 5: LY900009 attenuates LPS-induced bone resorption in vivo. (A) Images of three-dimensional reconstruction based on micro-CT scanning. (B) Results of BMD, bone BV/TV, and Tb.n. (C, D) HE and TRAP results. (E) Amount of TRAP-positive OCs. Data are presented as mean ± SD (* p < 0.05, ** p < 0.01, *** p < 0.001), n=6 per group. Please click here to view a larger version of this figure.
The present investigation provides compelling evidence that LY900009 represents a promising therapeutic agent for osteolytic diseases through its potent and selective inhibition of osteoclastogenesis, consistent with the growing interest in γ-secretase inhibitors for bone-related disorders14,20,21. The findings demonstrate that this γ-secretase inhibitor effectively suppresses RANKL-induced osteoclast formation and bone resorptive function through the coordinated disruption of multiple critical signaling pathways, including Notch, MAPK, PI3K/Akt, and NF-κB cascades. The therapeutic significance of these results extends beyond mechanistic insights, as evidenced by the compound's efficacy in preventing LPS-induced bone loss in vivo, suggesting substantial clinical potential for managing inflammatory and metabolic bone disorders7,8,19,25. The favorable therapeutic window observed between effective concentrations (0.1-0.4 µM) and cytotoxic doses (>2.9 µM) supports the feasibility of clinical development.
The molecular mechanisms underlying LY900009's anti-osteoclastic effects reveal complex interactions between Notch signaling and osteoclastogenic pathways16. Our demonstration that LY900009 reduces cleaved Notch1 levels and downstream Hes1 expression, coupled with subsequent NFATc1 downregulation, provides important clarification regarding the controversial role of Notch signaling in osteoclast biology13. Unlike bisphosphonates, which primarily affect mature osteoclast function and survival, or denosumab, which blocks RANKL-RANK interaction, LY900009 appears to interfere with fundamental differentiation processes while preserving the possibility for more physiological regulation of bone remodeling10,17,22. The simultaneous suppression of MAPK (ERK1/2, p38), Akt, and NF-κB phosphorylation suggests that LY900009's effects extend beyond direct Notch inhibition, potentially involving crosstalk between Notch and other signaling networks essential for osteoclast development. This multi-pathway targeting may contribute to the compound's potent inhibitory effects and could provide advantages over more selective therapeutic approaches, particularly in inflammatory conditions where conventional therapies show limited efficacy.15,18,21,26,27.
Several critical steps are required for reproducible results using this protocol. RANKL concentration optimization (50-100 ng/mL) is essential, as BMM sensitivity varies between isolations. The timing of LY900009 addition significantly affects outcomes; adding 24 h post-RANKL stimulation allows initial lineage commitment while maintaining inhibitory efficacy. Maintaining M-CSF activity through -80 °C storage in single-use aliquots and maintaining culture pH at 7.2-7.4 are crucial for consistent osteoclast formation. When performing TRAP staining, strict adherence to the three-nucleus threshold for osteoclast identification ensured consistency between experiments. For troubleshooting, if osteoclast formation is inconsistent, verify cell viability exceeds 95% by trypan blue exclusion, confirm reagent activity, and ensure medium osmolality remains within physiological range (280-300 mOsm/kg). Alternative cell sources, including RAW264.7 cells, can be used, though primary BMMs better recapitulate physiological responses.
However, several important limitations and considerations must be acknowledged when interpreting these results and planning future research. The pleiotropic roles of γ-secretase and Notch signaling across multiple organ systems raise concerns about their potential systemic effects and long-term safety implications. Notch signaling plays critical roles in cardiovascular, immune, gastrointestinal, and nervous system functions, necessitating the comprehensive evaluation of off-target effects and organ-specific toxicity14,28. Additionally, this study focused primarily on Notch1 and Hes1, whereas other Notch receptors and downstream targets may contribute to osteoclast regulation. The bone marrow microenvironment contains diverse cell populations regulated by Notch signaling, including hematopoietic stem cells and mesenchymal stromal cells, and LY900009's effects on these populations warrant further investigation29,30,31. While LY900009 showed minimal impact on osteoblast viability in these preliminary assessments, a comprehensive evaluation of bone formation is needed. The LPS model represents acute inflammatory bone loss, and chronic disease models may yield different results.
The clinical implications of our findings are substantial, particularly considering the growing burden of osteolytic diseases in the aging population worldwide32,33,34. LY900009 demonstrated the ability to preserve bone microarchitecture while reducing osteoclast activity, which could translate into reduced fracture risk and improved bone quality across diverse clinical scenarios34,35,36,37. Future applications of this protocol extend beyond LY900009 to screening other γ-secretase inhibitors, Notch modulators, or novel anti-resorptive compounds. The development of bone-targeted formulations could minimize systemic exposure while maximizing skeletal efficacy38,39. The integration of LY900009 into combination therapy regimens, potentially with anabolic agents or targeted anti-inflammatory treatments, could maximize therapeutic benefits while minimizing individual agent limitations40,41. Personalized medicine approaches for patients with specific Notch pathway variants or inflammation-driven osteolysis represent exciting future directions.
In conclusion, this protocol provides researchers with a comprehensive, reproducible methodology for evaluating γ-secretase inhibitors as anti-osteoclastic therapeutics. The demonstrated efficacy of LY900009 in both in vitro and in vivo models, combined with its favorable therapeutic window and novel mechanism of action, validates this approach for drug discovery in bone diseases. The standardized techniques, quality control measures, and troubleshooting guidance presented here address common challenges in osteoclast research where protocol variations often yield inconsistent results. While successful clinical translation will require careful evaluation of safety and optimization of dosing strategies, this protocol establishes a robust platform for advancing novel therapeutics from bench to bedside. These methods can be completed within 3-4 weeks and adapted for various compound classes, providing the bone research community with valuable tools for developing next-generation treatments for osteolytic diseases.
The authors declare no competing financial interests or conflicts of interest. No author has any financial relationship with the commercial entities mentioned in this work. All authors have completed the ICMJE disclosure forms.
The authors would like to thank the staff of the Central Hospital Affiliated to Shandong First Medical University and Feicheng People's Hospital for their technical support and assistance during the course of this study. We also appreciate the contributions of our laboratory colleagues for their valuable discussions and assistance with animal experiments.
| 24-well cell culture plates | Corning | 3524 | Immunofluorescence microscopy |
| 6-well cell culture plates | Corning | 3516 | Cell culture and western blot |
| 70% Ethanol | Sigma-Aldrich | E7023 | Sterilization and disinfection |
| 75% Ethanol | Sigma-Aldrich | E7148 | RNA washing buffer |
| 96-well cell culture plates | Corning | 3596 | Cell culture and assays |
| Aluminum filter (0.5 mm) | Bruker | N/A | Micro-CT scanning filter |
| Anti-CD11b-FITC antibody | BioLegend | 101206 | Flow cytometry - macrophage marker |
| Anti-cleaved Notch1 antibody (rabbit monoclonal) | Cell Signaling Technology | 4147S | Western blot - Notch signaling |
| Anti-F4/80-PE antibody | BioLegend | 123108 | Flow cytometry - macrophage marker |
| Anti-fade mounting medium | Vector Laboratories | H-1000 | Fluorescence microscopy mounting |
| Anti-Hes1 antibody (rabbit monoclonal) | Cell Signaling Technology | 11988S | Western blot - Notch downstream target |
| Anti-NFATc1 antibody (mouse monoclonal) | Santa Cruz Biotechnology | sc-7294 | Western blot - osteoclast master regulator |
| Anti-phospho-Akt (Ser473) antibody | Cell Signaling Technology | 4060S | Western blot - PI3K/Akt signaling |
| Anti-phospho-ERK1/2 (Thr202/Tyr204) antibody | Cell Signaling Technology | 4370S | Western blot - MAPK signaling |
| Anti-phospho-NF-κB p65 (Ser536) antibody | Cell Signaling Technology | 3033S | Western blot - NF-κB signaling |
| Anti-phospho-p38 MAPK (Thr180/Tyr182) antibody | Cell Signaling Technology | 4511S | Western blot - p38 MAPK signaling |
| Anti-β-actin antibody (mouse monoclonal) | Cell Signaling Technology | 4970S | Western blot loading control |
| Automated imaging reader | Molecular Devices | ImageXpress Micro | Resorption pit quantification |
| BCA Protein Assay Kit | Pierce/Thermo Fisher | 23225 | Protein concentration determination |
| Bleach (10% sodium hypochlorite) | Sigma-Aldrich | 425044 | Cell removal from resorption plates |
| BSA (Bovine Serum Albumin) | Sigma-Aldrich | A7906 | Blocking buffer for immunofluorescence |
| C57BL/6 mice (male, 5-8 weeks old) | Charles River Laboratories | 27 | In vitro BMM isolation and in vivo studies |
| Calcium oxalate test reagent | Sigma-Aldrich | C0350 | Decalcification completion testing |
| CCK-8 reagent (Cell Counting Kit-8) | Dojindo Molecular Technologies | CK04 | Cell viability assessment |
| Cell strainer (70 μm) | BD Falcon | 352350 | Bone marrow cell filtration |
| Centrifuge (refrigerated) | Eppendorf | 5424R | Cell and sample preparation |
| Chloroform | Sigma-Aldrich | C2432 | RNA extraction phase separation |
| CO2 chamber | Euthanex Corp | EX-075 | Animal euthanasia |
| CTAn software | Bruker | Version 1.20 | Micro-CT image analysis |
| DAPI (4',6-diamidino-2-phenylindole) | Sigma-Aldrich | D9542 | Nuclear counterstaining |
| Dimethylformamide (DMF) | Sigma-Aldrich | D4551 | TRAP staining solvent |
| DMSO (Dimethyl sulfoxide) | Sigma-Aldrich | D2650 | Vehicle control preparation |
| ECL Western Blotting Substrate | Pierce/Thermo Fisher | 32106 | Protein detection in western blot |
| EDTA (10%, pH 7.4) | Sigma-Aldrich | E6758 | Decalcification solution |
| Eosin Y | Sigma-Aldrich | E4009 | H&E histological staining |
| Fast Red Violet LB salt | Sigma-Aldrich | F3381 | TRAP staining chromogen |
| Fetal Bovine Serum (FBS) | Gibco/Thermo Fisher | 16140-071 | Cell culture supplement |
| Flow cytometer | BD Biosciences | FACSCalibur | BMM characterization |
| Fluorescence microscope | Olympus | IX73 | Immunofluorescence imaging |
| G*Power software | Heinrich Heine University | Version 3.1 | Statistical power analysis |
| Glass coverslips (22 × 22 mm) | Fisher Scientific | 12-541-B | Immunofluorescence microscopy |
| GraphPad Prism | GraphPad Software | Version 9.0 | IC50 calculation and graphing |
| Hematoxylin | Sigma-Aldrich | H3136 | H&E histological staining |
| Hemocytometer | Hausser Scientific | 3200 | Cell counting |
| High-Capacity cDNA Reverse Transcription Kit | Applied Biosystems | 4368814 | cDNA synthesis from RNA |
| HRP-conjugated anti-mouse IgG | Cell Signaling Technology | 7076S | Western blot secondary antibody |
| HRP-conjugated anti-rabbit IgG | Cell Signaling Technology | 7074S | Western blot secondary antibody |
| Hydroxyapatite-coated 96-well plates | Corning BioCoat | 354651 | Bone resorption functional assay |
| ImageJ | NIH | Version 1.53e | Image analysis and quantification |
| Incubator (CO2) | Thermo Fisher | 51033058 | Cell culture maintenance |
| Infrared imaging system | Bio-Rad | ChemiDoc MP | Western blot detection |
| Inverted microscope | Olympus | CKX41 | Cell culture monitoring, TRAP counting |
| Isopropanol | Sigma-Aldrich | I9516 | RNA precipitation |
| LPS (Lipopolysaccharide from E. coli) | Sigma-Aldrich | L4391 | In vivo inflammatory bone resorption |
| LY900009 (γ-secretase inhibitor) | Selleck Chemicals | S7999 | Test compound - osteoclast inhibitor |
| M-CSF (Macrophage Colony-Stimulating Factor) | R&D Systems | 416-ML-010 | Macrophage survival and osteoclast differentiation |
| Micro-CT scanner | Bruker | SkyScan 1272 | Bone microarchitecture analysis |
| Microplate reader | BioTek | Synergy H1 | Absorbance measurements (CCK-8) |
| Microtome | Leica | RM2255 | Histological sectioning |
| Naphthol AS-MX phosphate | Sigma-Aldrich | N5000 | TRAP staining substrate |
| Needles (25G, 27G) | BD | 305122, 305109 | Bone marrow flushing, injections |
| NRecon software | Bruker | Version 1.7 | Micro-CT image reconstruction |
| Paraffin wax | Sigma-Aldrich | P3683 | Tissue embedding |
| Paraformaldehyde (4%) | Sigma-Aldrich | P6148 | Cell and tissue fixation |
| PBS (Phosphate Buffered Saline) | Gibco/Thermo Fisher | 14190-144 | Cell washing and buffer preparation |
| Pentobarbital sodium | Sigma-Aldrich | P3761 | Animal anesthesia and euthanasia |
| Phosphatase inhibitor cocktail | Roche | 4906837001 | Phosphorylation preservation |
| PowerUp SYBR Green Master Mix | Applied Biosystems | A25742 | Quantitative real-time PCR |
| Protease inhibitor cocktail | Roche | 11697498001 | Protein degradation prevention |
| PVDF membrane (0.45 μm) | Bio-Rad | 1620177 | Western blot protein transfer |
| RANKL (Receptor Activator of NF-κB Ligand) | R&D Systems | 462-TEC-010 | Osteoclast differentiation induction |
| Real-time PCR system | Applied Biosystems | 7500 Fast | Quantitative gene expression analysis |
| Rhodamine-phalloidin | Invitrogen/Thermo Fisher | R415 | F-actin visualization |
| RIPA buffer | Cell Signaling Technology | 9806S | Protein extraction |
| RNase-free water | Ambion/Thermo Fisher | AM9937 | RNA dissolution and PCR |
| SDS-PAGE gels (10%) | Bio-Rad | 4561036 | Western blot protein separation |
| Skim milk powder | Bio-Rad | 1706404 | Western blot blocking |
| Sodium acetate buffer (0.1 M, pH 5.0) | Sigma-Aldrich | S2889 | TRAP staining buffer |
| Sodium tartrate (50 mM) | Sigma-Aldrich | S4297 | TRAP staining specificity |
| Spectrophotometer | Thermo Fisher | NanoDrop 2000 | RNA/protein concentration measurement |
| SPSS | IBM Corporation | Version 28.0 | Statistical analysis |
| Syringes (1 mL, 10 mL) | BD | 309659, 309604 | Bone marrow flushing, injections |
| T75 cell culture flasks | Corning | 430641 | Primary BMM culture |
| TBST (Tris-Buffered Saline with 0.1% Tween-20) | Bio-Rad | 1706531 | Western blot washing buffer |
| Transfer buffer | Bio-Rad | 1704270 | Western blot protein transfer |
| Triton X-100 | Sigma-Aldrich | T8787 | Cell permeabilization |
| TRIzol reagent | Invitrogen/Thermo Fisher | 15596026 | Total RNA extraction |
| Trypan blue (0.4%) | Sigma-Aldrich | T8154 | Cell viability assessment |
| Trypsin-EDTA (0.25%) | Gibco/Thermo Fisher | 25200-056 | Cell harvesting and detachment |
| Western blot transfer system | Bio-Rad | Trans-Blot Turbo | Protein transfer to membrane |
| Xylene | Sigma-Aldrich | X3750 | Tissue processing and clearing |
| α-MEM (Alpha Minimum Essential Medium) | Gibco/Thermo Fisher | 12561-056 | Cell culture medium |