INTRODUCTION
Mesenchymal stem cells (MSCs) are multipotent stem cells capable of differentiating into various cell types, including osteoblasts, chondrocytes, and adipocytes.[
1] Owing to their regenerative capacity, MSCs have been extensively utilized in tissue engineering and regenerative medicine, particularly for bone regeneration.[
2-
4] Among their sources, bone marrow-derived MSCs (BMSCs) are the most widely investigated.[
5,
6] However, BMSCs are often contaminated with macrophages and hematopoietic cells, necessitating additional purification steps such as liposomal clodronate treatment.[
7,
8] These added procedures increase the complexity and cost of their clinical application.
Macrophage contamination diversely affects the osteogenic potential of BMSCs, particularly under inflammatory conditions. Macrophages modulate bone remodeling through polarization into either M1 (pro-inflammatory) or M2 (anti-inflammatory) phenotypes. M1 macrophages, activated by interferon-γ or lipopolysaccharide, inhibit osteogenesis via inflammatory cytokines and inducible nitric oxide synthase expression.[
9-
11] In contrast, M2 macrophages promote osteogenic differentiation by secreting regenerative factors such as transforming growth factor-β , bone morphogenetic protein (BMP)-2, and vascular endothelial growth factor.[
12,
13] Uncontrolled macrophage influence within MSC cultures hampers reproducibility and complicates mechanistic studies, especially those focused on lineage commitment, immunomodulation, and extracellular vesicle profiling.[
8,
14,
15]
Alveolar bone-derived MSCs (aBMSCs) have recently emerged as a promising alternative due to their higher osteogenic potential compared to other dental MSCs. Among various types, aBMSCs and periodontal ligament stem cells exhibit the greatest osteogenic capacity, while dental follicle progenitor cells rank lowest.[
16] In comparisons with iliac bone-derived MSCs, aBMSCs demonstrated superior mineral deposition and osteogenic gene expression.[
17] Moreover, single-cell RNA sequencing (scRNA-seq) has revealed a distinct transcriptional signature in aBMSCs, including high expression of FAT4, a potential regulator of osteogenic differentiation not prominently expressed in BMSCs.[
18,
19] In addition, Oncostatin M (OSM), secreted by classically activated macrophages, was enriched in alveolar bone compared to long bone, where it regulates the osteogenic and adipogenic differentiation of MSCs.[
19] Differences in OSM responsiveness between aBMSCs and BMSCs may underlie their contrasting behavior under inflammatory stimuli. However, the molecular mechanisms by which these pathways govern aBMSCs-mediated bone regeneration remain largely undefined.
The present study aims to obtain BMSCs with minimal macrophage contamination and to evaluate their osteogenic potential. Specifically, the objectives are to (1) quantify macrophage contamination in aBMSCs and BMSCs using flow cytometry; (2) evaluate the osteogenic differentiation capacity of aBMSCs; and (3) compare the osteogenic responses of aBMSCs and BMSCs under interleukin-1β-induced inflammatory stress, including analysis of OSM, FAT4, and another osteogenic regulatory gene expression. These are expected to elucidate the molecular basis underlying the osteogenic properties of aBMSCs and support their application in regenerative therapies, particularly for inflammation-associated bone loss.
METHODS
1. Isolation and culture of murine BMSCs and aBMSCs
The animal study was approved by the Institutional Animal Care and Use Committee of Chonnam National University (Approval No. CNU IACUC-YB-2023-12) and conducted in accordance with its guidelines. Murine BMSCs and aBMSCs were obtained from C57BL/6 mice aged 7 to 8 weeks (Damool Science, Daejeon, Korea). For the isolation of BMSCs, mice were sacrificed by CO2 inhalation, and their hind limbs were dissected. The tibias and femurs were meticulously cleaned to remove surrounding soft tissues and were subsequently placed in minimum essential medium-α (MEM-α) (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS) (Gibco; Thermo Fisher Scientific) and 1% penicillin-streptomycin. The bone marrow was extracted by flushing and passed through a 70 μm strainer (SPL, Daejeon, Korea) to eliminate any remaining bone debris.
The isolated cells from each mouse were suspended in 40 mL of complete culture medium and distributed evenly into four 10 cm culture plates at a density of 2.5×106 cells/mL. The cultures were incubated at 37°C with 5% CO2, and fresh media was provided every three days to remove unattached cells. After eight days, once the cells reached sub-confluency, adherent cells (passage 0, P0) were rinsed with phosphate-buffered saline (PBS) and detached by treating them with 0.25% trypsin-ethylenediaminetetraacetic acid (EDTA) (Gibco; Thermo Fisher Scientific) for 2 min.
For the isolation of aBMSCs, mice were euthanized by carbon dioxide inhalation, and the upper and lower jaws were carefully dissected and transferred to a 60 mm Petri dish. Soft tissues, including gingival tissue, were meticulously removed under sterile conditions. The molars were loosened by inserting a 1 mL needle between the crowns and subsequently extracted. Surrounding soft and non-alveolar bone tissues were carefully excised, leaving only the alveolar bone for further processing.
The isolated alveolar bone fragments were collected and placed in 2 mL of PBS containing 2% FBS. They were then finely minced using sterile scissors before being transferred to a 15 mL centrifuge tube. A solution containing 1 mg/mL type II collagenase (Gibco; Thermo Fisher Scientific) and 0.5 mg/mL DNase I (Sigma-Aldrich, St. Louis, MO, USA) was added for digestion. The sample was then incubated in a shaking incubator at 37°C, 1,500 rpm, for 30 min to break down the tissue enzymatically.
Following digestion, the resulting cell suspension was passed through a 70 μm cell strainer (SPL) to eliminate undigested tissue debris and then transferred into a 50 mL centrifuge tube. The filtered sample was centrifugated, after which the supernatant was discarded. The cell pellet was resuspended in 3 mL of PBS and centrifuged again under identical conditions. This cycle of digestion and centrifugation was repeated three times to enhance cell recovery.
After the final centrifugation step, the supernatant was carefully removed, and the remaining cell pellet was resuspended in 12 mL of complete culture medium (MEM-α supplemented with 10% FBS and 1% penicillin-streptomycin). The cells were then plated in a 10 cm culture dish and incubated at 37°C in a 5% CO2 environment. The culture medium was replenished every three days until the cells attained confluency.
2. Flow cytometric analysis
To assess cell surface marker expression through flow cytometry, BMSCs, and aBMSCs were first detached using 0.25% trypsin-EDTA, washed with pre-chilled PBS, and then resuspended in binding buffer (PBS containing 0.5% FBS and 0.1% sodium azide). The cells were then incubated at room temperature for 20 min with allophycocyanin (APC)-conjugated monoclonal antibody targeting CD11b marker and phycoerythrin (PE)-conjugated monoclonal antibodies targeting the following markers: Sca-1, CD44, CD105 (Endoglin), CD90.2 (Thy1.2), and CD140a (PDGFRα). These antibodies were procured from BioLegend (San Diego, CA, USA), except for CD105, obtained from eBioscience (San Diego, CA, USA). To ensure proper gating, isotype control antibodies were used, including APC-conjugated Rat IgG2b, κ, PE-conjugated Rat IgG2a, κ, and Rat Ig-G2b, κ (BioLegend).
Following incubation, the cells underwent a single wash with binding buffer before being promptly analyzed using an Accuri C6 Plus flow cytometer (BD Biosciences, San Jose, CA, USA) equipped with CSampler software. The specificity of antibody binding to target markers was validated by comparing fluorescence intensity levels with those of the respective isotype controls.
3. Differentiation induction
BMSCs and aBMSCs were seeded at a 5×104 cells/mL density in a complete culture medium to induce osteogenic differentiation. The cells were allowed to adhere overnight, after which the culture medium was replaced with an osteogenic induction medium. This specialized medium contained 50 μg/mL ascorbic acid (Sigma-Aldrich), 10 mM β-glycerophosphate (Sigma-Aldrich), and 100 ng/mL recombinant human BMP-2 (Cowellmedi Co., Busan, Korea) in MEM-α. For optimal differentiation conditions, the medium was replenished every three days.
For the control group, cells were cultured in a standard growth medium (GM) without osteogenic supplements. After the differentiation period, cells were fixed with 4% paraformaldehyde and stained with Alizarin Red S (ARS) (Sigma-Aldrich) to evaluate mineralization levels. The staining assessment was conducted on day 14 for BMSCs and day 10 for aBMSCs to monitor calcium deposition.
4. RNA extraction and quantitative real-time polymerase chain reaction (qRT-PCR)
Total RNA was extracted from cultured cells using TRIzol reagent (Ambion Inc., Austin, TX, USA) according to the manufacturer’s protocol. Approximately 2 μg of RNA was reverse-transcribed into complementary DNA (cDNA) using M-MLV reverse transcriptase (Promega, San Luis Obispo, CA, USA). qRT-PCR was performed using a QuantStudio cycler (Applied Biosystems, Foster City, CA, USA) with Power SYBR Green PCR Master Mix (Applied Biosystems) to analyze the expression levels of target genes. The primer sequences used for each target gene were as follows: Runx2 (Forward: 5′-TTCAAGGATGGAATTTGTGGG-3′, Reverse: 5′-GGATGAGGAATGGGCCCTA-3′); Osterix (Osx, Forward:5′-AGGCCACATTGGTAGCAAA-3′, Reverse: 5′-GCGGCTGATTGGCTCTTCT-3′); Bone sialoprotein (Bsp, Forward: 5′-AAGGCACCGTGTGAGTTAGGT-3′, Reverse: 5′-CCTTGAGTAGTGATTCATCCTC-3′); Osteocalcin (Ocn, Forward: 5′-GCAATAGGTAGTGAAGAACATCC-3′, Reverse: 5′-GTTTGTAGGCGGTCTTCAAGC-3′); Osm (Forward: 5′-TCCCCTCCAAAACCTGACACAC-3′, Reverse: 5′-ATGGTATCCCACTGAAAGGC-3′); FAT4 (Forward: 5′-GTTCCCTAGCAGGACTAACGGG-3′, Reverse: 5′-TAACATGACGTTGCTGGGG-3′); 18S rRNA (Forward: 5′-GGCCCTTCTTAGAGGGACA-3′, Reverse: 5′-CCCGGAACATCTCACGAC-3′).
Relative gene expression levels were calculated using the ΔΔCt method with normalization to 18S rRNA as an internal control. All data were analyzed using QuantStudio Design & Analysis software (Applied Biosystems).
5. Statistical analysis
All experiments were conducted in at least three independent replicates. Statistical comparisons between groups were performed using a paired Student’s t-test to assess significance. A P-value of less than 0.05 was considered statistically significant. All statistical analyses were performed using GraphPad Prism version 9.0 (GraphPad Software Inc., San Diego, CA, USA).
DISCUSSION
The present study demonstrates that aBMSCs exhibit markedly lower macrophage population, maintain stable osteogenic differentiation under IL-1β-induced inflammatory conditions, and display distinct gene expression profiles contributing to their enhanced osteogenic capacity compared to BMSCs derived from the long bones of the hindlimb.
For effective bone tissue engineering, securing a large quantity of high-quality stem cells is essential. In this study, BMSC cultures derived from long bones showed approximately 27% CD11b
+ macrophage contamination, whereas aBMSC cultures showed less than 1%. These findings suggest that aBMSCs are more homogeneous, which may result in more consistent regenerative outcomes. This difference likely stems from the anatomical origin of the cells. The alveolar bone, particularly in the anterior region, is composed of dense cortical bone with limited marrow space and a sparse vascular pore network. This contrasts with the richly vascularized and hematopoietic marrow environment of long bones.[
20,
21] These structural features of the alveolar bone naturally restrict the presence of hematopoietic and immune cells, which likely contributes to the reduced immune cell content observed in aBMSCs cultures.
In terms of osteogenic characteristics, aBMSCs isolated in this study exhibited robust osteogenic differentiation in osteogenic induction medium and maintained their bone-forming ability under IL-1β stimulation. In contrast, BMSCs showed a marked reduction in osteogenic capacity when exposed to the same inflammatory conditions. Specifically, in IL-1β-treated BMSCs, the expression of osteogenic genes (Runx2, Osx, Bsp, and Ocn) and calcium deposition were significantly diminished, whereas aBMSCs remained largely unaffected. These findings suggest that aBMSCs may possess unique anti-inflammatory tolerance, enabling them to sustain bone-regenerative function even in chronically inflamed environments. Given that alveolar bone is constantly exposed to microbial and mechanical stimuli, such adaptability may have emerged through evolutionary selection as a form of environmental resilience.
Moreover, under inflammatory stimulation, we observed contrasting expression patterns of FAT4 and OSM—both known to promote osteogenic differentiation [
22-
25]—between aBMSCs and BMSCs. OSM, a cytokine belonging to the IL-6 family, is primarily secreted by T cells and macrophages and promotes osteogenesis via activation of the JAK-STAT signaling pathway.[
22,
23] FAT4 is a membrane-bound protocadherin that functions as an upstream regulator of the Hippo pathway, contributing to osteogenic differentiation.[
24,
25] In our study, IL-1β stimulation in BMSCs resulted in a marked increase in OSM expression, which was accompanied by a reduction in mineral deposition. This likely reflects macrophage contamination inherent to conventionally isolated BMSCs, leading to enhanced OSM expression and the establishment of a pro-inflammatory microenvironment. In contrast, aBMSCs displayed minimal changes in OSM expression under the same inflammatory conditions, while maintaining stable osteogenic potential.
Notably, Fat4
+ osteogenic progenitors were identified as a cell population uniquely enriched in alveolar bone, underscoring the tissue-specific expression of FAT4.[
18,
24,
25] Consistent with this, our data showed that FAT4 expression increased in aBMSCs upon osteogenic induction and remained elevated following IL-1β stimulation, whereas such changes were negligible in BMSCs. These findings suggest that FAT4 may serve as a molecular mechanism supporting the osteogenic capacity of aBMSCs even under inflammatory stress, further indicating that aBMSCs and BMSCs exhibit subtle but functionally relevant differences. Nevertheless, these observations are correlative and warrant further mechanistic studies to determine causality.
Overall, aBMSCs exhibit low macrophage contamination and an inflammation-resistant osteogenic capacity, distinguishing them from conventionally isolated BMSCs. Their ability to sustain osteogenic differentiation even under pro-inflammatory conditions is particularly significant for regenerative strategies targeting periodontal disease and peri-implantitis, both characterized by chronic inflammation and progressive alveolar bone loss. Given these properties, aBMSCs represent a promising cell-based therapeutic approach for treating bone loss in inflammatory microenvironments.