Isolation and characterization of DPSC
The present study was approved by the Medical Ethical Committee of the Affiliated Stomatology Hospital of Guangzhou Medical University (No. JCYJ2022021), and all patients signed an informed consent form. The DPSC were all obtained from the human pulp tissue of premolars that were removed for orthodontic treatment in the Affiliated Stomatology Hospital of Guangzhou Medical University. Total 30 teeth from 20 patients (10 male, 10, female, 1-2 tooth/patient) were used for DPSC isolation. The inclusion criteria for teeth collection were the permanent teeth from 18 to 25-year-old patients without caries, periodontal disease, periapical lesion, and systemic inflammatory diseases. After extraction, the teeth' surface was washed with phosphate-buffered saline (PBS) (Gibco, USA) containing 4% penicillin/streptomycin (Gibco, USA) 2 times. The pulp tissues were gently separated from the root and crown within 4 h, minced with sterile scissors into 1-2 mm fragments, then digested with 3 mg/mL collagenase type I (Gibco, USA) and 4 mg/mL dispase (Gibco, USA) for 45 min at 37℃, and centrifuged at 1000 rpm for 5 min. The cells’ precipitation was resuspended using alpha-modified minimal essential media (α-MEM; Gibco, USA) containing 15% fetal bovine serum (FBS) (Gibco, USA) and 1% of penicillin/streptomycin, and cultured at 37℃ in an environment with 5% CO2 with the medium replacement every 3 days. Upon reaching 80‑90% confluency, cells were collected with 0.25% trypsin-EDTA (Gibco, USA) and subcultured at a 1:3 ratio. DPSC between passages 2 to 5 were used in subsequent experiments.
Flow cytometry analysis of DPSC’ surface markers
The DPSC of P3 in T75 flasks were digested with 0.25% trypsin-EDTA, collected, and incubated for 30 min with monoclonal antibodies against human CD29, CD34, CD44, CD45, CD73, CD90, and CD105 (BD Pharmingen, USA; Abcam, USA), as well as isotype-matched control IgG1. CD44 and CD105 were labeled with phycoerythrin (PE). CD45 and CD90 were labeled with PE-CY5 conjugate. CD29, CD34, and CD73 were labeled with fluorescein isothiocyanate (FITC). The expression profiles for the cell surface markers were analyzed using a flow cytometer and Cell Quest software (Beckman Coulter, USA).
Colony forming unit assay
Using the low-density cell seeding method, the capacity of colony-forming units was assessed. DPSC (P3) were seeded at a density of 500 cells/well into 6-well plates (Corning Incorporated, Corning, NY, USA) and maintained in the normal growth medium. After 7 days, cells were fixed with 4% paraformaldehyde for 10 min, washed twice with PBS, and stained with crystal violet staining (Sigma, USA).
Multilineage differentiation of DPSC
Alizarin red staining: DPSC (P3) were seeded in 6‑well culture plates at a density of 105 cells/well and cultured in α-MEM supplemented with 10% FBS and 1% of penicillin/streptomycin at 37˚C with 5% CO2 in a humidified incubator. When the cells become 80% confluence, the culture medium was replaced with osteogenic medium (growth medium supplemented with 10 mM beta-glycerophosphate, 50 mg/L ascorbic acid, and 10 nM dexamethasone). The osteogenic medium was replenished every 3 days. At day 21, the cells were fixed with 4% paraformaldehyde for 20 min at room temperature and washed with PBS. Osteogenic cultures were stained with 2% alizarin red solution, pH 4.2 (500 μl/well, Sigma-Aldrich), for 5-10 min to assess for mineral nodule deposition detection by light microscopy.
Oil red O staining: DPSC (P3) were seeded in 6‑well culture plates at a density of 105 cells/well and cultured in α-MEM supplemented with 10% FBS and 1% of penicillin/streptomycin at 37˚C with 5% CO2 in a humidified incubator. When the cells become 90% confluence, the culture medium was replaced with adipogenic medium (growth medium containing 10 mg/L insulin, 1 μM dexamethasone, 0.5 mM 3-Isobutyl-1-methylxanthine and 0.2 mM indomethacin). The adipogenic medium was replenished every 3 days. At day 28, the cells were fixed with 4% paraformaldehyde for 30 min at room temperature and washed with PBS. Adipogenic cultures were stained with a working solution of the Oil Red O staining (Solarbio, China) for 30 min to observe the formation of intracellular lipid vacuoles by light microscopy.
Alcian blue staining: DPSC (P3) were resuspended in a growth medium at a density of 4×106 cells/ml. Ten microliters of this cell suspension containing 4×104 cells were pipetted in the center of each well of a 48-well plate (Corning Incorporated, Corning, NY, USA). Then, micro-masses were given 2 h to form. After that, maintenance medium (0.5 ml/well) was added for 1 day, followed by the addition of chondrogenic medium (growth medium supplemented with 50 mg/L ascorbic acid, 0.1 μM dexamethasone, 40 mg/L L-Proline, 10 μg/L TGF-β3 and 1% Insulin-Transferrin-Selenium). The chondrogenic medium was refreshed every 3 days. At day 21, the micro-masses were fixed with 4% paraformaldehyde for 30 min at room temperature and washed with PBS. Chondrogenic cultures were stained with a working solution of Alcian blue staining (Solarbio, China) for 30 min to assess the chondrogenic differentiation.
Cell proliferation assay
To determine the effect of CBD on DPSC proliferation ability, we used cell counting kit-8 (CCK-8; Dojindo, Japan). DPSC were seeded at a density of 2×103 cells/well in 96‑well plates (Corning Incorporated, Corning, NY, USA) in triplicate. After 24 h of culture, the old medium was replaced with the medium containing 0, 0.1, 0.5, 2.5, and 12.5 µM CBD (Sigma, USA) respectively (six repeats per concentration). The medium was replaced every 3 days. At 1, 3, or 5 days, the culture medium was discarded. Then, 90 µl medium and 10 µl CCK8 reagent was added to each well and incubated in the incubator at 37 ℃ for 1 h. The results were recorded by a microplate reader (Thermofisher, USA) at an absorbance of 450 nm.
Alkaline phosphatase (ALP) staining and activity
DPSC (2 × l04 cells/well) were seeded in 48-well culture plates, and cultured in a complete medium for 24 h. Then, the old medium was replaced with the osteogenic medium containing 0, 0.1, 0.5, and 2.5 µM CBD respectively (four repeats per concentration). The medium was replenished every 3 days. On days 4 and 7, the original medium was removed. For ALP staining, the cells were fixed with 4% paraformaldehyde for 30 min. Then, the cells were washed twice with PBS and stained using the BCIP/NBT ALP color development kit (Beyotime, China). Staining was observed under a bright field microscope. (Leica, Germany).
For the measurement of ALP activity, the cells were washed twice with PBS, and lysate was extracted in a lysis buffer containing 0.1% Triton x-100. The total protein concentration in the cell lysate was analyzed by the BCA protein assay kit (Thermo Scientific, USA). The activity of ALP was performed using the ALP assay kit (Nanjing Jiancheng Chemical Industrial Ltd, China). The results were measured by a microplate reader at an absorbance of 520 nm. The value of ALP activity was normalized to total protein content.
Matrix mineralization assays
DPSC (2 × l04 cells/well) were seeded in 48-well culture plates, and cultured in a complete medium for 24 h. Then, the old medium was replaced with the osteogenic medium containing 0, 0.1, 0.5, and 2.5 µM CBD respectively. The medium was replenished every 3 days. On day 21, the original medium was removed. The mineralized matrix was stained by alizarin red staining. For semiquantitative analysis of 150 µl of 10% cetylpyridinium chloride (CPC) solution (Sigma, USA), the solution was added to each well of 48-well culture plates to dissolve the mineralized nodules. The eluted liquid was transferred to the 96-well plates. The results were measured by a microplate reader at an absorbance of 562 nm.
Development of micro-spheroids and characterization
The polydimethylsiloxane (PDMS) master mold with 200 µm pillar diameter, 150 µm pillar height, and 100 µm gap between two neighboring pillars was fabricated and used to develop agarose gel microwells for micro-spheroids culture. The PDMS mold was sterilized with 75% alcohol or under an ultraviolet lamp for 30 min. Agarose gel (Baygene, China; 3% w/v, agarose/deionized water) was sterilized using an autoclave and poured onto a PDMS mold containing pillars with a diameter of 200 µm. After allowing the agarose to solidify, microwell inserts with an area of ≈1.8 cm2 were punched out and inserted in 24-well plates. PBS (1 mL) was then added, and the wells were sterilized using a UV light for 30 min. Each 24-well insert contained ≈2000 microwells. To form a micro-spheroid structure, DPSC were trypsinized, and 5x105 cells were diluted in 500 µL of serum-free chemically defined medium (icell bioscience, China) and seeded in 24-well plates. Around 250 cells homed in each well and self-aggregated to form micro-spheroid. Micro-spheroids were further cultured in a serum-free osteogenic medium with or without CBD (2.5 µM). After that, images of micro-spheroids were captured at each time point, and the medium was changed every 48 h. For mechanism-related rescue experiments, CBD-treated micro-spheroids were incubated with an additional 100 ng/ml recombinant dickkopf-related protein 1 (DKK1, Sino biological, China) for 7 days. The half medium was replaced with fresh conditioned medium containing DKK1 every 24 h.
Live/dead staining
Cell viability in microspheres was assessed qualitatively with the Live/Dead viability/cytotoxicity kit (Solarbio, China). Briefly, micro-spheroids were rinsed with PBS, where after they were incubated in 2×10−6 M Calcein-AM and 4×10−6 M Propidium Iodide for 30 min at 37 °C, 5% CO2, and 95% humidity. Images of each stained micro-spheroids were captured on days 1, 3, 7, and 14 using confocal laser scanning microscopy (CLSM, Leica TCS SP8). Live cells stain green and dead cells stain red.
Staining for cytoskeleton
Micro-spheroids were fixed for 30 min at room temperature with 4% paraformaldehyde, rinsed with PBS, permeabilized for 30 min with PBS containing 0.5% triton X-100, and rinsed with PBS. Cell nucleus and F-actin distribution within micro-spheroids were visualized by staining with 4′,6-diamidino-2-phenylindole (DAPI) (Solarbio, China) and 0.8 U/mL tetramethyl rhodamine isothiocynate (TRITC) conjugated Phalloidin (Solarbio, China) during 1 h at room temperature. Sequential images of the micro-spheroids were captured using a confocal laser scanning microscopy (CLSM, Leica TCS SP8) using respective filters. The Leica AF image processing software was used to construct 3D images.
RT-qPCR analysis
Total RNA was extracted from DPSC and micro-spheroids using an RNA isolation kit (Accurate Biotechnology, China). The total RNA (500 ng) from each sample was reverse-transcribed to cDNA (Accurate Biotechnology, Hunan, China). RT-qPCR was performed using an SYBR Green RT-qPCR kit (Accurate Biotechnology, Hunan, China). The gene expressions of osteogenic markers alkaline phosphatase (ALP), bone morphogenetic protein 2 (BMP2), runt-related transcription factor 2 (RUNX2), osteopontin (OPN), osteocalcin (OCN), and WNT6 were analyzed by RT-qPCR. The data were normalized to the internal control GAPDH. The primer sequences used for RT-qPCR are listed in Table 1.
Western blotting
Pretreated DPSC and micro-spheroids were lysed with RIPA buffer (Beyotime, China) containing protease inhibitors (PMSF; Beyotime, China) on ice for 30 min. The cell lysate was centrifuged at 12,000 rpm for 15 min to collect the supernatant. Total protein concentration was measured using the BCA protein assay kit (Thermo Scientific, USA). Total protein (20 μg) was loaded in 10% SDS-PAGE gel (Epizyme, China) for protein separation and transferred into polyvinylidene fluoride (PVDF) membranes (Millipore, Billerica, MA, USA) using wet blotting techniques. The membrane was blocked with 5% nonfat milk powder for 1 h and washed thrice with TBST. The membrane was then incubated at 4℃ overnight with primary antibodies, which included rabbit anti-human Collagen I (COL-I, Abcam, England), rabbit anti-human RUNX2 (Abcam, England), rabbit anti-human WNT6 (Bioss, China), and rabbit anti-human β-catenin (Bioss, China). After incubation, the primary antibodies were removed. Horseradish peroxidase (HRP)-conjugated secondary antibodies (Abcam, England) was incubated at room temperature for 1 h. The protein bands were detected using an enhanced chemical luminescence kit (Beyotime, China). The ImageJ software was used for the semi-quantification of band intensity.
Mice calvarial bone defect repair
Animal experiments were conducted following the protocols approved by the Laboratory Animal Ethics Committee of Guangdong Huawei Testing Co., LTD in this study (No.20220103). Forty male athymic nude mice (Zhuhai BesTest Bio-Tech Co., Ltd., China), 6-8 weeks old and weighing 19-26g were used. Animals were housed at a specific pathogen-free animal facility in stable conditions (22 ± 2°C) with a 12 h dark/light cycle and ad libitum access to food and water. Animals were allowed to acclimatize for 1 week before experiments and were regularly monitored for signs of pain/infection, food intake, and activity during the entire experimental period.
Before implantation, both DPSC and micro-spheroids were cultured in an osteogenic medium with or without CBD for 2 weeks. Then, DPSC were digested with 0.25% trypsin-EDTA and the micro-spheroids were washed in PBS. After centrifugation at 1,000 rpm for 5 min, the supernatant was carefully removed and the cells were resuspended in 10% (w/v) GelMA hydrogels (EFL-GM-60; Engineering For Life, China) which were sterilized by filtering through a 0.22 µm syringe filter. Constructs (≈7.5×105 DPSC /3×103 micro-spheroids per construct, n=8) with a diameter of 3 mm and a depth of 1 mm were exposed to LAP 405 nm blue light (Engineering For Life, China) for 5 s as seen in Fig. 5A. Controls transplanted with 10% (w/v) GelMA hydrogel. The 40 cranial defects were randomly separated into 5 groups: (a) control, (b) DPSC, (c) CBD (2.5 µM)-treated DPSC, (d) micro-spheroids, and (e) CBD (2.5 µM)-treated micro-spheroids. DPSC and micro-spheroids entrapped within the microgel units were cultured at 37 °C with 5% CO2 before implantation. To assess cell viability, the Live/Dead staining assay was performed on the GelMA hydrogel constructs cultured in vitro for 1, 3, and 7 days.
Preoperatively, all animals were anesthetized by intramuscular injections of ketamine hydrochloride (35 mg/kg) and xylazine (5 mg/kg). Following anesthesia, the calvarial bone was exposed, and a 3 mm circular bone defect was created with a trephine drill under normal saline irrigation. Constructs were placed into the defect, and the skin was sutured to close the wound. The animals received antibiotic therapy for 3 days. After 8 weeks, the mice were sacrificed by isoflurane inhalation through the approved Ethics Committees' methods and calvarial bone was collected and fixed with 4% paraformaldehyde for subsequent micro-computed tomography (micro-CT) and histological analysis.
Micro-CT analysis
Micro-CT images were taken using micro-CT equipment (Skyscan-1172; Bruker, Kontich, Belgium). The X-ray source is set at 60 kV and 100 μA the three-dimensional image is obtained by isotropy with the voxel size of 10 μm. The serial sections were reconstructed into a 3D image using reconstructions and osteogenic parameters conducted with NRecon v.1.6.9 software. CTAn (Bruker micro-CT, BE) was used for all image processing and quantification of mineralized tissue. The percentage of mineralized tissue was calculated with respect to the total explant volume. Bone volume was measured by using a fixed threshold setting for all images. CTvox (Bruker micro-CT, BE) was used to create 3D visualization. The osteogenic parameters such as bone volume/total volume (BV/TV), bone surface/total volume (BS/TV), bone mineral density (BMD), and trabecular thickness (Tb. Th) were measured.
Histology and immunostaining analysis
For histological analysis, all samples were fixed, demineralized, dehydrated, and embedded in paraffin. The sample blocks were sectioned to a thickness of 4 μm and stained with hematoxylin and eosin (H&E) as well as Masson’s trichrome. The newly formed mineralized area was measured using ImageJ software.
Micro-spheroids were fixed with 4% paraformaldehyde at room temperature for 2 h and washed with PBS. After centrifugation at 500 rpm for 5 min, micro-spheroids were snap-frozen, embedded in optimal cutting temperature compound (OCT) compound, and sectioned on a cryostat at 5-μm thickness. After blocking with 1% bovine serum albumin (BSA; Sigma‐Aldrich) for 1 h at room temperature, the frozen sections were incubated with the primary antibodies OCN (1:500; Proteintech, China) overnight. After a washing step with PBS, the secondary antibody was applied for 1 h (CoraLite488-conjugated Goat Anti-Rabbit IgG, 1:200; Proteintech, China). After nuclear staining with DAPI, sample imaging was performed with confocal laser scanning microscopy (CLSM, Leica TCS SP8). Immunofluorescence staining was quantified using ImageJ software from three different samples.
mRNA sequencing and bioinformatics analysis
The total RNA of DPSC and microspheres treated for 21 days with or without 2.5 µM CBD was extracted using Trizol® (Invitrogen, Carlsbad, CA). RNA samples were detected based on the A260/A280 absorbance ratio with a Nanodrop ND-2000 system (Thermo Scientific, USA), and the RIN of RNA was determined by an Agilent Bioanalyzer 4150 system (Agilent Technologies, CA, USA). Only qualified samples will be used for library construction. Paired-end libraries were prepared using an ABclonal mRNA-seq lib prep kit (ABclonal, China) following the manufacturer’s instructions. PCR products were purified (AMPure XP system) and library quality was assessed on an Agilent Bioanalyzer 4150 system. Finally, the library preparations were sequenced on an MGISEQ-T7 and 150 bp paired-end reads were generated (Shanghai Applied Protein Technology, China). The data generated from the BGI platform were used for bioinformatics analysis. RNA-seq sequencing data quality was verified by Fastqc. Then clean reads were separately aligned to the reference genome with orientation mode using HISAT2 software to obtain mapped reads. FeatureCounts was used to count the reads numbers mapped to each gene. And then Fragments Per Kilobase per Million (FPKM) of each gene were calculated based on the length of the gene and the reads count mapped to this gene. Differential expression analysis for mRNA was performed using R package edgeR. Differentially expressed RNAs with |log2(FC)| value > 1 and q value < 0.05, considered as significantly modulated, were retained for further analysis.
Statistical analysis
All data were obtained from at least three independent experiments with each experiment in triplicate under identical conditions and shown as the mean ± standard deviation (SD). To test the significance of observed differences between the study groups, one-way analysis of variance (ANOVA) or Student’s t-test of the GraphPad Prism Software (version 9.0, USA) was used. A value of p < 0.05 was considered a significant difference.