Abstract Background Radiation-induced jaw injury is one of the most severe complications after radiotherapy for head and neck cancer, which can disrupt patients’ health and quality of life. Although the direct target of inflammation and suppressed bone regeneration activity by ionizing radiation (IR) has been phenomenally observed, the underlying mechanisms and potential therapeutic targets remain blurred. Osteoimmunology emphasizes that dendritic cells (DCs) may contribute to bone diseases. Methods In this study, we assessed phenotypic and functional alterations of DCs in a radiation-induced jaw injury rat model through immunohistopathological staining. The effects of IR on bone marrow-derived dendritic cells (BMDCs) in vitro were further validated by flow cytometry, ELISA, mixed lymphocyte reaction (MLR) assay, and transwell. The cellular responses and differentiation of bone marrow mesenchymal stem cells (BMSCs) under BMDC-derived conditioned medium stimulation were evaluated through various cell staining, Quantitative real-time polymerase chain reaction (qRT-PCR), and western blotting. Flow cytometry, qRT-PCR, WB were employed to verify tolerogenic characteristics of Vitamin D3 (VitD3)-induced tolerogenic DCs (tolDCs). TolDCs were encapsulated in GelMA to develop an effective in vivo therapeutic approach for irradiated jaw defects. Results We revealed that IR activated the mature-inflammatory phenotype and corresponding biological functions of DCs through the nuclear factor kappa-B (NF-κB) signaling pathway. Exposure to conditioned medium from irradiated BMDCs induced oxidative stress and inflammatory responses in BMSCs, inhibiting their proliferation, migration, and osteogenic potential while potentiating adipogenic capacity. Furthermore, tolDCs were proven to be resistant to radiation-induced activation. Local administration of tolDCs was effective in improving bone regeneration of irradiated jawbone defects. Conclusions Hyperactivation of DCs served as a potential pathogenic factor in radiation-induced jaw injury, exacerbating local inflammation and abrogating the biological functions of BMSCs. The local transplantation of tolDCs was a promising therapeutic strategy for osteogenesis in radiation-induced jaw injury. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-025-04508-x. Keywords: Osteonecrosis of the jaw, Lonizing radiation, Dendritic cells, Osteoimmunology, Bone marrow mesenchymal stem cells Introduction Radiotherapy (RT) plays a pivotal role in managing head and neck cancer, with approximately 80% of the patients undergoing RT at least once during the course of their disease [[34]1]. Radiation-induced jaw injury is one of the potentially severe, delayed, and irreversible complications of RT, occurring in up to 30% of the patients, and the incidence of osteoradionecrosis of the jaw (ORNJ) is about 8–15% [[35]2–[36]4]. The characteristic clinical features of radiation-induced jaw injury include persistent pain, infection, exposure of necrotic bone, and ultimately functional injuries [[37]5]. At present, the gold standard therapeutic approaches for patients with radiation-induced jaw injury have not yet been defined. Possible interventions start from non-invasive measures like oral hygiene and antibiotics, escalate through sequestrectomy to segmental mandibulectomy [[38]6]. The symptoms and surgical treatments have negative impacts on practically all patient-related quality of life metrics and remain a medical challenge to surmount [[39]7]. Understanding the pathophysiology of radiation-induced jaw injury is crucial for developing effective and targeted treatment strategies, while there remain many gray areas to be explored at the cytological and molecular levels [[40]8–[41]10]. With the development of osteoimmunology, growing evidences highlight the extensive crosstalk between the skeletal and immune systems [[42]11]. Our previous studies [[43]12, [44]13] have shown that ionizing radiation (IR) alters the composition, activity, and immunological status of immune cells in the jawbone cavity, thus breaking the homeostasis of the immune microenvironment at the single cell level, and suggested that there existed a correlation between dendritic cells (DCs) and early stage radiation-induced jaw injury. DCs are central components of innate immunity and regulators of adaptive immunity. DCs are not only actively involved in all phases of tissue healing, from initial inflammation to tissue remodeling [[45]14], but also play a regulatory role in skeletal homeostasis and bone regeneration [[46]15]. Depending on the external signals, immature DCs (imDCs) can be selectively differentiated into either a mature or tolerogenic state [[47]16]. A biased differentiation of DCs may result in exacerbated inflammation or loss of tolerance. For example, an increase in highly activated DCs was observed in the progression of osteonecrosis of the femoral head [[48]17, [49]18]. However, to date, there is a lack of in-depth investigation into the correlation between DC phenotypic/functional alterations and radiation-induced jaw injury. Bone marrow mesenchymal stem cells (BMSCs), capable of differentiating into osteoblastic lineages and promoting osseointegration, have emerged as a central focus and target cell in the field of regenerative medicine [[50]19, [51]20]. Despite several studies have demonstrated the regulatory role of immune cells on functions of BMSCs, there is no agreement on which DC phenotype is beneficial or detrimental for BMSCs [[52]15, [53]21]. A better understanding of the crosstalk between DCs and BMSCs is proposed to have a positive impact on tissue regeneration in radiation-induced jaw injury. Tolerogenic DCs (tolDCs) refer to a maturation-resistant DC subset induced by antigenic or pharmacological stimuli, characterized by their low expression of co-stimulatory molecules, high expression of tolerance-associated markers, and anti-inflammatory cytokine production [[54]22]. Multiple studies have reported beneficial effects of tolDC therapy in the treatment of non-self-limiting inflammatory diseases and chronic wounds [[55]23, [56]24]. We hypothesize that tolDCs may also have therapeutic effects on radiation-induced jaw injury. Here, we observed the recruitment of DCs with enhanced maturation-related phenotypic and functional characteristics in a radiation-induced jaw injury rodent model. In vitro experiments further demonstrated that IR promoted the transition of DCs toward a mature and inflammatory phenotype via the nuclear factor kappa-B (NF-κB) signaling pathway. Conditioned medium (CM) from these irradiated DCs induced oxidative stress, inhibited osteogenic, and promoted lipogenic differentiation of BMSCs. Vitamin D3 (VitD3)-treated tolDCs exhibited radiation-resistant features, which effectively mitigated mature DC (mDC)-mediated undesired effects on the differentiation of BMSCs, thus promoting the healing of irradiated mandibular bone defects. Taking together, these findings indicate that DCs may serve as a potential therapeutic target for radiation-induced jaw injury. Materials and methods The work has been reported in line with the ARRIVE guidelines 2.0. Animal Wild-type Sprague Dawley (SD) and F344 rats were purchased from Jihui Company (Shanghai, China). Male rats aged 4 weeks were used for the in vitro experiments. Male rats aged 10 weeks were used for the animal model and animals were randomly assigned to all experimental groups. All the rats were housed under specific pathogen-free (SPF) conditions with a circadian rhythm of 12 h. All the experiments received ethical approval from the Animal Care and Experiment Committee of Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SH9H-2022-A799-SB). Rat model A medical linear accelerator (Elekta Synergy) was applied for 6MVp X-ray radiation delivery. The operation was taken 100 cm away from the source with an incident beam on the mandibles of rats. For the radiation-induced jaw injury model, the rats were randomly assigned to two groups: control group (n = 10) and irradiated group (n = 10). For the irradiated group, SD rats were generally anesthetized with tribromoethanol (400 mg/kg, i.p.). A single dose of 20 Gy (6 Gy/min) IR was delivered to the left mandibles. One week after RT, the first molar of the left mandible was extracted. The control group received sham IR and also underwent the dental procedure. The rats were sacrificed 3 and 5 weeks after jaw RT for mandibles and cervical lymph nodes (LNs). To construct the irradiated mandibular defect rat model, a single dose of 10 Gy IR was delivered to the right mandibles of F344 rats. The control rats (Ctrl group) were also anesthetized and received sham IR. One week after RT, a 1 cm skin incision was made at the lower edge of the mandibles of all rats, and the masseter muscles were meticulously dissected, followed by an incision of the periosteum. A full-thickness penetrating critical-sized defect with 4 mm diameter was made by a trephine drill. Subsequent to the cleansing of the surgical site with saline, the mandible defects were loaded with 20 µL gelatin methacryloyl (Gelma, EFL-GM, China) containing 1 × 10^6 mDCs (IR + mDC group), tolDCs (IR + tolDC group) from F344 rats or without cells (IR group) (n = 5 per group). The surgical site was then sealed utilizing a suture. Unirradiated mandibles were loaded with blank Gelma at the same time. Nine weeks after RT, the mandibles were collected and fixed with 4% formaldehyde for subsequent analyses. The major organs, including the livers, kidneys, lungs, hearts, and spleens, were also dissected for biosafety validation. The rats were sacrificed by inhaling CO[2]. We placed the rats in a pre-filled CO₂ chamber (20% displacement rate) and maintained exposure for 5 min after respiratory arrest. Then we confirmed death via bilateral thoracotomy. Micro-computed tomography (micro-CT) Rat mandible samples were placed into a sample holder and scanned using a micro-CT instrument (PINGSENG Healthcare, China) at a resolution of 18.26 μm at 40 kV and 250 µA. The morphology of bones was measured using Avatar 3 software. Bone parameters, including bone volume fraction (BV/TV), bone mineral density (BMD), trabecular thickness (Tb.Th), and trabecular number (Tb.N) were quantified. Histology The bone tissues were decalcified in 10% EDTA at room temperature for 2 weeks. All specimens were paraffin-embedded and then sectioned at a thickness of 5 μm for further hematoxylin-eosin (H&E), masson, immunohistochemistry (IHC), and immunofluorescence (IF) staining. H&E staining was performed to visualize the morphology and regeneration status of the tissues. Masson staining was performed to indicate collagen deposition and osteoid formation. For IHC and IF staining, sections were performed heat-mediated antigen retrieval in sodium citrate buffer for 20 min. The samples were then blocked in 5% bovine serum albumin solution for 1 h and incubated overnight at 4 °C with primary antibodies including CD11c (MBS25560, Mybiosource, USA), CD86 (sc-28347, Santa Cruz, USA), runt-related transcription factor 2 (RUNX2; [57]GB115631, Servicebio, China), CD4 (ab237722, Abcam, USA), OCN ([58]GB115684, Servicebio, China). Subsequently, IHC sections were incubated with a secondary antibody conjugated to horseradish peroxidase (HRP) for 1 h and visualized by 3,3’-diaminobenzidine (DAB) substrates. IF samples were incubated with corresponding secondary antibodies, including Alexa Fluor 488- and Cy3-labeled secondary IgG (Servicebio, China) for 1 h at room temperature. DAPI was used to counterstain the nuclei. The sections were imaged via a Zeiss Scope. A1 microscope (Carl Zeiss, Germany). Quantitative real-time polymerase chain reaction (qRT-PCR) Total mRNA was extracted from the cells and mandible tissues using TRIzol reagent (Takara Biotechnology, Japan) and then reverse-transcribed to cDNA from 1000 ng of total RNA using Hieff Unicon^® V Universal Multiplex One Step qRT‒PCR Probe Kit (Yeasen Biotechnology, China). The qPCR system was performed on a Light Cycler-^® 480 Instrument II (Roche, Switzerland) using SYBR Green Master Mix (Yeasen Biotechnology, China). Primers used in this study are listed in Table [59]S1. Glyceraldehyde-3-phosphate dehydrogenase (Gapdh) was used as the housekeeping gene. Isolation and IR of bone marrow-derived dendritic cells (BMDCs) BMDCs were isolated from the bone marrow of 4-week-old male SD and F344 rats euthanized by inhaling CO[2]. Bone marrow single-cell suspensions were generated from tibias and femurs. After lysing erythrocytes, cells were resuspended in RPMI-1640 containing 10% fetal bovine serum (FBS) (Cyagen, USA), 100 U/ml penicillin/streptomycin, 20 ng/mL granulocyte macrophage-colony stimulating factor (GM-CSF), and 10 ng/mL interleukin-4 (IL-4) (PeproTech, USA) at a density of 5 × 10^5 cells/ mL at 37 °C under 5% humidified CO[2] for 7 days. The medium was changed on day 3 and 6 with fresh growth factors. On day 7, floating and loosely adherent cells were harvested as immature DCs. BMDCs were also exposed to various IR doses (0, 2, 4, 8, 12, 16 Gy at 6.0 Gy/min) from 6MVp X-ray and cultured further for another 24 h. To obtain mDCs, 100 ng/mL lipopolysaccharides (LPS) (Sigma-Aldrich, USA) were added to the medium concomitantly with IR. To induce tolDCs, 1,25(OH)[2]D[3] (10 nM; Sigma-Aldrich, USA) dissolved in anhydrous ethanol was added to the culture medium throughout the whole period of DC induction and then stimulated by a single 8 Gy dose of IR and LPS. The control cells were induced with an equal volume of anhydrous ethanol. Isolation of BMSCs 4-week-old SD rats were used for BMSCs isolation. The femur and tibia were removed aseptically after the rats were euthanized by inhaling CO[2]. Bone marrow was flushed out and cultured in Dulbecco’s modified Eagle’s medium (DMEM)/low glucose with 10% FBS and 100 U/mL penicillin/streptomycin. The attached cells were passaged when cell density reached 90%. Passage 2–3 BMSCs were used for cell experiments. Preparation and collection of conditioned medium (CM) of BMDCs After IR and LPS stimulation for 24 h, mDCs were thoroughly washed with phosphate buffer saline (PBS) and cultured for another 24 h with fresh RPMI-1640 containing 10% FBS. The culture medium was centrifuged at 300 xg for 10 min, followed by supernatant collection. Supernatants were mixed with DMEM at a ratio of 1:1 as BMDC-conditioned medium (CM) to culture BMSCs in vitro. BMSCs cultured in DMEM complete medium were used as controls. Cell proliferation After exposure to IR for 24 h, imDC and mDC were incubated with 10 µL of cell counting kit-8 (CCK-8; Dojindo Molecular Technologies, Japan) and 90 µL of RPMI-1640 culture medium at 37 ℃ for 1 h before measuring the absorbance at 450 nm to quantify the proliferation activity of BMDCs. In another experiment, a CCK-8 assay was performed to check the viability of BMSCs co-cultured with DC-CM on day 1, day 3, and day 5. Flow cytometry analysis (FCA) The expression of surface markers in BMDCs was detected 24 h after IR by flow cytometry using antibodies as follows: Zombie Violet (BioLegend, USA), FITC anti-rat CD11c (Santa Cruz, USA), APC anti-rat major histocompatibility complex class II (MHCII; eBioscience, USA), APC anti-rat CD40 (eBioscience, USA), BV421 anti-rat CD80 (BD Pharmingen, USA) and PE anti-rat CD86 (BioLegend, USA). The cells were incubated with their respective antibodies for 40 min at 4 ℃ and subsequently analyzed on a BD FACSAria III flow cytometer (BD Biosciences, USA). Data analyses were performed by FlowJo software. Apoptosis analysis Apoptosis was quantified using the Annexin-V FITC apoptosis detection kit (BD Pharmingen, USA) according to the manufacturer’s instructions. Briefly, BMDCs were collected and resuspended in binding buffer 24 h after IR. 5 µL of FITC-Annexin V and 5 µL of propidium iodide (PI) were added to the buffer for 15 min of staining. The cells were analyzed by flow cytometry within 1 h. ELISA The concentration of IL-1β, IL-6 IL-10, tumor necrosis factor (TNF)-α, transforming growth factor (TGF)-β, IL-12, and interferon (IFN)-γ in supernatants of BMDCs were measured 24 h after exposure to IR by enzyme-linked immunosorbent assay (ELISA) kits (Multi Sciences, China) following the manufacturer’s instructions. Western blotting (WB) One hour after IR and LPS treatment, BMDCs were collected and lysed in radio immunoprecipitation assay lysis buffer with phenylmethanesulfonylfluoride and phosphatase inhibitors. For electrophoresis, 10% sodium dodecyl sulfate‒polyacrylamide gel electrophoresis gels were added with an equal amount of protein and electrically transferred to polyvinylidene fluoride membranes (Merck, Germany). The membranes were blocked, washed, and then incubated overnight with primary antibodies. The primary antibodies used in the current study were purchased from Cell Signaling Technology (USA) and listed as follows: anti-GAPDH (2118), α-Tubulin (2144), anti-phospho-P65 (pP65) (Ser536) (3033), anti-P65 (8242), anti-phospho-inhibitor of kappa B kinase α/β (pIKKα/β) (Ser176/180) (2697), anti-IKKα (2682), anti-phospho-inhibitory subunit of NF kappa B α (pIKBα) (Ser32) (2859), anti-IKBα (4812), anti-phospho-Janus kinase 2 (pJAK2) (3776), anti-JAK2 (3230), anti-phospho-signal transducer and activator of transcription 3 (pSTAT3) (9145), anti-STAT3 (12640). Then, the membranes were incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies for 1 h at room temperature. The signals were detected by chemiluminescence and semi-quantified through ImageJ software. The NF-κB pathway inhibitor JSH-23 (Abmole, USA) was added to the culture medium of BMDCs two hours before IR at the concentration of 20 µM to inhibit P65 nuclear translocation. The expression of osteogenic and lipogenetic proteins in BMSCs co-cultured with DC-CM were analyzed by WB. Protein bands were exposed to anti-osteocalcin (OCN; 20277-1-AP), anti-RUNX2 (20700-1-AP), anti-peroxisome proliferative activated receptor gamma (PPARG; 16643-1-AP; Proteintech, China), and anti-alkaline phosphatase (ALP; abs158623; Absin, China) antibodies. The following steps had been previously outlined. Migration assay After IR and LPS stimulation for 24 h, BMDCs were seeded into the upper chambers of the transwell inserts with a 8 μm pore membrane at the density of 2 × 10^5 /well. The lower chambers were RPMI-1640 with 100 ng/mL chemokine-CC motif-ligand 19 (CCL19; PeproTech, USA). After one day, the upper chamber membranes were stained with crystal violet staining solution, and the number of migrating cells was observed under an optical microscope (Nikon, Japan). Mixed leukocyte reaction (MLR) The BMDCs of each group were set as stimulator cells and incubated with mitomycin C (25 µg/mL) for 30 min at 37 ℃ to remove the proliferative ability. Responder cells were isolated from the spleens of F344 rats by lymphocyte separation medium (TBD science, China). Stimulator cells and responder cells were mixed at the ratio of 1: 10 and cultured for 72 h in round-bottomed 96 plates. The CCK-8 reagent was added to each well to detect the proliferation of lymphocytes. The absorbance was measured at 450 nm. Live/dead cell staining A calcein/PI viability/cytotoxicity assay kit (Beyotime, China) was applied to assess the toxicity of CM to BMSCs as well as GelMA to DCs. On the third day of co-culture, 1 µL of calcein and 1 µL of PI were added to each well for 15 min of staining. The cells were observed and imaged by a fluorescence microscope. RNA-sequencing (RNA-seq) and bioinformatics analysis The BMSCs were cultured with CM for 3 days. Total RNA was extracted and sent to DIATRE biological technology company (China) for library preparation, RNA-seq, and data analysis. DESeq2 package was carried out to perform differential expression analysis, and differentially expressed genes (DEGs) were determined by|log[2]FoldChange|≥1.5, and a P value < 0.05. Gene ontology (GO) analysis and Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analysis were utilized to detect the molecular and biological processes as well as signaling pathways. Detection of reactive oxygen species (ROS) One day after CM treatment, BMSCs were washed twice with PBS and incubated with 2,7-dichlorodi-hydrofluorescein diacetate (DCFH-DA; Beyotime, China) for 30 min at 37 ℃, and then imaged by a fluorescence microscope. The mean fluorescence intensity (MFI), which indicated the level of ROS, was measured by ImageJ software. Scratch test Seeding BMSCs in a 6-well culture plate, allowing them to grow to full confluence. A sterile pipette tip was used to create 3 straight scratches across the cells. The cells were then washed with PBS to remove debris. The culture medium was replaced by low-serum (2%) CM. BMSCs were cultured in a 37 °C incubator and photographed by microscope at 0 and 24 h at the same position. The wound areas and migration rates in all groups were analyzed using ImageJ software. ALP and Alizarin red S (ARS) staining BMSCs were co-cultured with mDC-CM and OriCell SD rat bone marrow mesenchymal stem cell osteogenic differentiation basal medium (Cyagen, USA). For ARS staining, the duration of osteogenic induction was 14 days, and 7 days for ALP staining. The cells were washed twice using PBS, followed by fixation and staining with ARS kit (Solarbio, China) or BCIP/NBT alkaline phosphatase color development kit (Beyotime, China) for 30 min. Oil red O staining BMSCs were co-cultured with DC-CM and OriCell SD rat bone marrow mesenchymal stem cell adipogenic differentiation basal medium (Cyagen, USA) for 7 days. Oil red O staining solution (Cyagen, USA) was utilized for staining following the manufacture’s protocols, and images were captured by an optical microscope. Cell seeding in hydrogels First, mDCs and tolDCs were mixed with the GelMA solution at the concentration of 1 × 10^6 cells/mL, then light cured using ultraviolet light (405 nm) for 10 s and cultured by RPMI-1640 complete medium containing 10% FBS for 5 days. Characterization of DCs loaded GelMA The GelMA was lyophilized using FreeZone (Labconco, USA), and the internal morphology was observed by scanning electron microscopy (GeminiSEM 300, ZEISS, Germany). The rheological analysis of GelMA or mDCs/tolDCs encapsulated GelMA was performed by a DHR-2 rheometer (TA, USA). The modulus change was recorded at a fixed angular frequency of 1 rad/s and shear strain of 1%, respectively. Statistical analysis All the data were obtained from at least three independent experiments. The values were presented as mean ± standard deviation (SD). The Student’s t-test and one-way ANOVA were used to perform statistical analyses using GraphPad Prism 7.0 (GraphPad Software, USA). P < 0.05 was considered statistically significant. Results The radiation-induced jaw injury rat model shows impaired osteogenesis and DCs activation To investigate the subacute and chronic jawbone complications caused by IR, we constructed a rat radiation-induced jaw injury model by a single dose of 20 Gy IR combined with mandibular tooth extraction (Fig. [60]1A). Rats were closely monitored over 5 weeks. The irradiated rats suffered weight loss, shedding, and impaired gingival healing (Supplementary Fig. [61]S1A-C). As seen in the sagittal and coronal plane of micro-CT, the bone density of tooth extraction sockets in the control groups increased over time. While there was almost no new bone formation at week 3 and a small amount of osteogenesis at week 5 in irradiated rats. Quantitative analysis of micro-CT results corroborates these observations. The values of BV/TV, BMD, and Tb.Th in the control group gradually increased over time. A statistically significant difference was observed between the CT values of radiation and unirradiated groups, and there was no difference between the irradiated groups at two time points (Fig. [62]1B-C). H&E staining indicated the emergence of nascent bones across control groups. The tooth socket was replaced by fibrous tissue in irradiated mandibles. Moreover, we observed sequestra with empty osteocyte lacunae and adipocytes in bone marrow at week 5, which was known as bone marrow steatosis (Fig. [63]1D). The changing trends of osteogenesis-related and lipogenesis-related genes, including Alp, osteopontin (Opn), Ocn, Runx2, Osterix, collagen I (Col1) and Pparg, were consistent with micro-CT and histological changes (Supplementary Fig. [64]S1D). RUNX2 was a key transcription factor regulating early orthogenic differentiation. Its expression in the irradiated mandibles was significantly lower than that in the control group and gradually decreased over time (Supplementary Fig. [65]S1E-F). All these results suggest that bone regeneration was suppressed, and bone marrow structure was altered in the radiation-induced jaw injury model. Fig. 1. [66]Fig. 1 [67]Open in a new tab Phenotypic transformation of DCs in radiation-induced jaw injury rat model. (A) Flowchart of construction of radiation-induced jaw injury rat model. (B, C) Micro-CT and analysis of ROI (tooth extraction socket) in radiation-induced jaw injury rat model at week 3 and week 5. (D) Representative H&E staining images of tooth extraction socket and bone marrow area in the sagittal plane of the rat mandibles. (Newly-formed bone: NB; blue arrows: necrotic bone; yellow arrows: adipocytes) (E, F) Representative IF images showing the presence of CD11c^+CD86^+ DCs (white arrows) in the mandibles from rats with radiation-induced jaw injury, and the quantitative analysis of DC numbers. (G, H) Representative IHC images and quantification showing the presence of CD11c^+ DCs in the cervical lymph nodes from rats with radiation-induced jaw injury. (I, J) Representative IHC images and quantification showing the presence of CD4^+ T cells in the mandibles from rats with radiation-induced jaw injury. Data were expressed as means ± SD. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Graphs were created with BioRender.com DCs can up-regulate MHC-II and co-stimulatory molecules after activation and maturation. To demonstrate the effect of IR on DC activity, we utilized multiplex immunofluorescence staining to localize CD11c^+CD86^+ cells as activated DCs. The staining and quantitative results showed that the number of double-positive cells in the bone marrow of irradiated mandible was significantly higher than that of the corresponding controls at both points (Fig. [68]1E-F). After recognizing and capturing antigens in peripheral tissues, DCs can migrate to the draining LNs, where they interact with naïve T cells, effectively induce adaptive immune responses, and further exacerbate inflammation. We performed IHC staining of the cervical LNs, the CD11c-positive area fraction of irradiated rats reached 8.2% and 15.5% at week 3 and 5 respectively, which was considerably higher compared with rats that received sham IR. The positive cells in the control group were mostly located in the lymphoid follicles. While in the irradiated rats, positive cells were located in both the lymphoid follicles and paracortex zones (T-cell area) (Fig. [69]1G-H). Additionally, IHC staining was performed on jawbones to label CD4^+ T cells. At week 3, there was no difference between the proportion of CD4^+ cells in the bone marrow between the experimental and control groups, which might be attributed to post-extraction inflammatory responses. By week 5, the control group exhibited a decline in the proportion of positive areas, whereas the irradiated group maintained high expression levels (Fig. [70]1I-J). To sum up, IR caused macroscopic and microenvironmental remodeling of the jawbone. Dendritic cells were activated, which might contribute to the amplification of inflammation and the progression of lesions. DCs transform into a mature phenotype after IR in vitro To further validate the effects of IR on DCs in vitro, we first imposed a gradient dose of IR on BMDCs (Fig. [71]2A). One day after IR, a dose above 4 Gy reduced the proliferation rate of BMDCs dose-dependently and increased the proportion of apoptosis in imDCs and mDCs. However, there was no significant difference in apoptosis between 4 Gy and 8 Gy (Supplementary Fig. [72]S2A-C). Since patients with head and neck tumors often undergo up to 70 Gy IR, we would adopt the highest dose as possible. To ensure 50% cell viability, a single dose of 8 Gy was used for subsequent experiments. Fig. 2. [73]Fig. 2 [74]Open in a new tab Radiation-induced BMDC activation and maturation in vitro. (A) Flowchart of BMDC culture and irradiation process. BMDCs were stimulated with 100 ng/mL LPS and exposed to gradient irradiation. (B, C) Flow cytometry plots showing gating strategy for MHCII^+, CD40^+, CD80^+ and CD86^+ populations in CD11c^+ DCs, and their corresponding bar charts. (D) Bar charts illustrating the concentration of TNF-α, IL-6, IL-10, TGF-β, IL-1β, IL-12p70 and IFN-γ in supernatant of irradiated and LPS treated BMDCs by ELISA. (E) Representative western blotting images revealed the expression of NF-κB pathway-associated factors in irradiated and LPS treated BMDCs. (F) Relative protein expression of p-P65/P65, p-IKKα/β/IKKα and p-IKBα/IKBα. (G) Representative western blotting images of irradiated BMDCs treated with NF-κB pathway inhibitor JSH-23 from whole cell lysate. (H) Relative protein expression of p-P65/P65. (I) Flow cytometry analysis of CD11c^+MHCII^+ and CD11c^+CD86^+ cells in irradiated BMDCs treated with NF-κB pathway inhibitor JSH-23. Data were expressed as means ± SD. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Graphs were created with BioRender.com. Full-length blots of E, G are presented in Supplementary Fig. [75]S7 One day after RT, DCs were detected for co-stimulatory molecules by flow cytometry. As indicated in the gating strategy, IR up-regulated the expression of CD40, CD80, CD86, and MHCII in both imDCs and mDCs (Fig. [76]2B-C). Pro- and anti-inflammatory cytokines in the supernatants of DCs were measured by ELISA. The imDCs secreted only a small amount of cytokines, and there was no difference with or without IR, except for TGF-β. For mDCs, more cytokines were secreted. IR significantly increased the concentration of TNF-α, and reduced the concentration of IL-6, IL-10, and TGF-β. However, IR didn’t alter the secretion level of IL-1β, IL-12p70, and IFN-γ. The changes in cytokine mRNA levels were consistent with ELISA results (Fig. [77]2D, S2D). In addition, IR also enhanced the migration of imDCs and mDCs in the presence of CCL19 (100 ng/mL) (Supplementary Fig. [78]S2E-F), as well as the ability of mDCs to stimulate lymphocyte proliferation (Supplementary Fig. [79]S2G), which were also the characteristics of DC activation. Several studies have revealed activation of the NF-κB and JAK-STAT signaling pathways in DCs across multiple infectious, allergic, and autoimmune conditions [[80]25–[81]27]. However, whether these two pathways involved in the radiation-induced DC maturation have not been fully elucidated. WB along with inhibitory experiments revealed the mechanism underlying the maturation of BMDCs induced by IR at the protein level. The phosphorylation levels of P65, IKKα/β, and inhibitory subunit of IkBa in imDCs and mDCs were significantly increased after RT, with imDCs lower than mDCs (Fig. [82]2E-F). The NF-κB pathway inhibitor JSH-23 restrained P65 nuclear translocation of irradiated imDC (Fig. [83]2G-H). Flow cytometry also showed that JSH-23 decreased the MFI of CD86 and MHC-II (Fig. [84]2I). However, the JAK-STAT pathway failed to be activated under IR conditions. There was no significant difference of p-JAK2 and p-STAT3 expression compared to their total proteins observed in imDCs or mDCs (Supplementary Fig. [85]S3A-D). In brief, when IR was applied to imDCs solely, the cells were in a semi-mature state characterized by low levels of co-stimulatory molecules, migration, MLR capability, and cytokine production. BMDCs achieved full maturation when induced by both LPS and IR to aggravate local inflammation. To better elucidate the role of DC inflammatory response in radiation-induced jaw injury, mDCs were used for follow-up experiments. Irradiated DC-CM induces dysfunction of BMSCs through inflammatory-reparative imbalance and oxidative stress BMSCs are pluripotent stem cells with properties including multipotency, self-renewal, and immunoregulation, serving as the basis for bone regeneration. IR-activated DCs may reprogram the landscape of jawbone microenvironment, thus indirectly affecting BMSCs. To investigate the role of molecular signals from mDC on BMSCs, we cultured BMSCs with CM from irradiated/unirradiated mDCs. BMSCs cultured in CM-free medium were the controls (Fig. [86]3A). Fig. 3. [87]Fig. 3 [88]Open in a new tab Irradiated DC-CM impairs BMSC proliferation and migration through oxidative stress and inflammatory response. (A) Graphical representation of the experimental strategy for BMDCs culture medium (CM) and BMSCs co-culture system. BMDCs were stimulated with 100 ng/mL LPS and 8 Gy irradiation. The CM was collected the next day for BMSC culture. (B) Representative fluorescence images for live/dead staining of BMSCs cultured in CM from unirradiated/irradiated mDCs for 72 h. The green and red channels represent the Calcein-AM and PI staining of live and dead cells respectively. (C, D) GO and KEGG enrichment analysis of the differentially expressed mRNAs (|log[2]FoldChange|≥1.5, P value < 0.05, and P value adjusted using a false discovery rate) between BMSCs from mDC-8 Gy and mDC-0 Gy group on the third day of co-culture. (E) Effects of irradiated mDC-CM on proliferation of BMSCs detected by CCK8 on day 1, 3, 5. (F) qRT-PCR results of inflammation-related gene expression in BMSCs cultured in CM from unirradiated/irradiated mDCs for 72 h. (G, H) Reactive oxygen species detection and quantification of BMSCs cultured in unirradiated/irradiated mDC-CM. (I, J) Representative light microscope images showing the migration of BMSCs after scratches were applied to the cells cultured in unirradiated/irradiated mDC-CM for 24 h. The percentage of closed wound area was used to calculate the migration ability of each group. Data were expressed as means ± SD. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Graphs were created with BioRender.com Live/dead staining showed that BMSCs in both the control group and CM co-culture group remained alive and intact on day 3. Only a small amount of cell debris and dead cells were observed (Fig. [89]3B). RNA-seq was performed on BMSCs cultured in CM for 3 days. GO analysis revealed that the top 10 enriched terms for upregulated genes in BMSCs from mDC-8 Gy group compared mDC-0 Gy group included regulation of inflammatory response, positive regulation of response to external stimulus and wound healing, while GO terms enriched in downregulated genes included extracellular matrix and ossification (Fig. [90]3C). KEGG enriched in downregulated genes consisted of Wnt signaling pathway and ECM-receptor interaction (Fig. [91]3D). GO and KEGG analyses between the mDC-0 Gy and control groups were shown in Supplementary Fig. [92]S4. These results indicated a dysregulation between inflammatory and reparative responses in BMSCs. ROS production was significantly triggered by DC-CM treatment in an IR-dependent manner (Fig. [93]3G-H), accompanied by disordered paracrine signaling. BMSCs manifested increased mRNA expression of pro-inflammatory cytokines and chemokines, including Ccl6, Ccl20, Cxcl2, Cxcl3, Il1a, Il1b, and Il6 (Fig. [94]3F). CCK-8 assay was performed on day 1, 3, and 5 after co-culture. DC-CM significantly decreased the proliferation of BMSCs. However, no difference was observed between the irradiated and unirradiated DC-CM treatment groups (Fig. [95]3E). Scratch assay suggested that CM co-culture could inhibit the migratory ability of BMSCs, irradiated DC-CM further reduces wound healing area (Fig. [96]3I-J). Abnormal differentiation of BMSCs result from irradiated DC-CM Previous studies and our rat model have suggested that radiation-induced mandibular injury is characterized by suppressed osteogenesis and pathological adipocytic infiltration within the bone marrow [[97]28, [98]29]. Given the potential influence of immunological factors on cellular differentiation, we evaluated how mDC-CM regulates osteogenic and adipogenic differentiation balance of BMSCs. qPCR revealed that CM from irradiated mDCs suppressed the osteogenesis of BMSCs, reducing mRNA expression of Runx2, Alp, Opn, integrin binding sialoprotein (Ibsp) and Ocn of BMSCs (Fig. [99]4A). We found that CM decreased the ALP activity and mineralization of BMSCs compared to the control group by ALP and ARS staining, while irradiated DC-CM could aggravate the inhibition (Fig. [100]4B). WB supported the result of qPCR that irradiated DC-CM down-regulated osteogenesis-related proteins including RUNX2, ALP and OCN (Fig. [101]4C-D). Oil red O staining was adopted to assess the adipogenic differentiation of BMSCs (Fig. [102]4F-G). The application of post-radiation DC-CM increased adipogenic differentiation of BMSCs, the expression of the adipogenic marker genes like enhancer binding protein alpha (Cebpa), enhancer binding protein beta (Cebpb), perilipin (Plin1), and Pparg (Fig. [103]4E), as well as the lipogenesis-related protein PPARG (Fig. [104]4H-I). Taken together, irradiated DCs disturbed the adipogenic and osteogenic balance of BMSCs. Fig. 4. [105]Fig. 4 [106]Open in a new tab Irradiated DC-CM disrupts the osteogenic-adipogenic differentiation balance in BMSCs. (A) qRT-PCR results of osteogenesis-related gene expression in BMSCs treated with unirradiated/irradiated mDC-CM. (B) ARS staining (up) and ALP staining (down) of unirradiated/irradiated mDC-CM treated BMSCs. (C) Representative western blotting images of osteogenesis-related proteins in BMSCs cultured with unirradiated/irradiated mDC-CM. (D) Relative protein expression of ALPL, RUNX2, and OCN. (E) qRT-PCR results of lipogenesis-related gene expression in BMSCs treated with unirradiated/irradiated mDC-CM. (F, G) Oil O staining and quantification of unirradiated/irradiated mDC-CM treated BMSCs. (H) Representative western blotting images of lipogenesis-related protein in BMSCs cultured with unirradiated/irradiated mDC-CM. (I) Relative protein expression of PPARG. Data were expressed as means ± SD. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Full-length blots of C, H are presented in Supplementary Fig. [107]S8 TolDCs exhibit hyporesponsiveness to IR and reverse adverse effects of mDCs on BMSCs We generated tolDCs through VitD3 induction, which is a well-validated protocol (Fig. [108]5A). TolDCs exhibited maturation resistance. qRT-PCR showed that irradiated tolDCs (tolDC-8 Gy) down-regulated gene levels of inflammatory cytokines Il1b and Tnf-α, and up-regulated anti-inflammatory factor Il-10 than unirradiated tolDCs (tolDC-0 Gy) (Fig. [109]5B). After IR, the percentage of MHC-II^+ DCs did not change in tolDCs as detected by flow cytometry. Although IR increased the expression of CD40, CD80, and CD86 in tolDCs, the magnitude of this activation was significantly lower than that of the irradiated mDCs (Fig. [110]5C). The phosphorylation levels of P65, IKKα/β, and IkBa in irradiated tolDCs were lower than mDCs. There was no statistical difference between unirradiated cells (Fig. [111]5D-E). Fig. 5. [112]Fig. 5 [113]Open in a new tab TolDCs exhibit radioresistance and have capacity to counteract the effect of mDCs on BMSCs. (A) Flowchart of inducing tolDCs with VitD3. VitD3 was added to the culture medium throughout the whole period of DC induction. tolDCs were then stimulated with 100 ng/mL LPS and exposed to 8 Gy irradiation. (B) qRT-PCR results of inflammation-related gene expression in unirradiated/irradiated tolDCs. (C) Flow cytometry analysis of MHCII^+, CD40^+, CD80^+ and CD86^+ populations in unirradiated/irradiated mDCs and tolDCs. (D, E) Representative western blotting images of unirradiated/irradiated mDCs and tolDCs. Relative protein expression of p-P65/P65, p-IKKα/β/IKKα and p-IKBα/IKBα. (F) ARS staining (up) and ALP staining (down) of BMSCs cultured with unirradiated/irradiated mDC-CM and tolDC-CM. (G) Representative western blotting images of osteogenesis-related proteins in BMSCs cultured with unirradiated/irradiated mDC-CM and tolDC-CM. (H) Relative protein expression of ALPL, RUNX2 and OCN. (I, J) Oil O staining and quantification of BMSCs cultured with unirradiated/irradiated mDC-CM and tolDC-CM. (K) Representative western blotting images of lipogenesis-related protein in BMSCs cultured with unirradiated/irradiated mDC-CM and tolDC-CM. (L) Relative protein expression of PPARG. Data were expressed as means ± SD. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Full-length blots of E, G, K are presented in Supplementary Fig. [114]S9 We also collected tolDC-derived CM (tolDC-CM) to induce the differentiation of BMSCs. TolDCs restored the osteogenic differentiation ability of BMSCs. ALP and ARS staining results showed that irradiated tolDCs induced more ALP activity and mineralization than mDCs (Fig. [115]5F). WB revealed upregulated expression of osteogenic marker proteins (ALPL, RUNX2, and OCN) in the tolDC-8 Gy group compared to the mDC-8 Gy group. However, RUNX2 and OCN levels in the tolDC-8 Gy group were still lower than those in the mDC-0 Gy group (Fig. [116]5G-H). We also observed less lipid droplet accumulation in tolDC-CM treated BMSCs (Fig. [117]5I-J). The protein level of PPARG was down-regulated as well (Fig. [118]5K-L). TolDCs accelerate bone healing in irradiated mandibular defect rat model To validate that excessive inflammation of DCs aggravate radiation-induced jaw injury, and that the inhibition of DC maturation may repair or even reverse the lesion in vivo, an irradiated mandibular defect rat model was constructed. GelMA loaded with tolDCs/mDCs was placed into the defect area (Fig. [119]6A). As demonstrated in Supplementary Fig. [120]S5A, the GelMA exhibited a porous network structure. The rheological analysis confirmed that the storage modulus (Gʹ) of GelMA was significantly higher than loss modulus (Gʹʹ). The modulus of GelMA, as a function of time, was not significantly impacted by the DC encapsulation (Supplementary Fig. [121]S5B). Both tolDCs and mDCs encapsulated within GelMA maintained viability (Supplementary Fig. [122]S5C). Fig. 6. [123]Fig. 6 [124]Open in a new tab Local transplantation of tolDCs promotes bone healing of irradiated mandibular defects in rats. (A) Flowchart of irradiated mandibular defect rat model construction and mDC/tolDC therapy. (B, C) Micro-CT coronal images (top) and 3D reconstruction images (bottom) of irradiated rat mandibular defect repair after local transplantation of mDCs and tolDCs. (C) Corresponding measurements of BMD and BV/TV show the healing efficiency of mandible defects in rats. (D) Representative H&E staining of mandible defects loaded with mDCs and tolDCs, showing the histological morphology of newly-formed bone (NB; black), bone marrow (MB; red), vessel (V; green), sequestrum (S; orange) and GelMA (blue). (E) Representative Masson staining of the mandible defects loaded with mDCs and tolDCs, showing the histological morphology of newly-formed bone (NB) and mature bone (MB). (F, G) Representative IHC images and quantification of showing the presence of OCN^+ osteoblasts in the mandible defects loaded with mDCs and tolDCs. Data were expressed as means ± SD. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Graphs were created with BioRender.com According to the radiographs, 9 weeks after RT, the IR group displayed less bone deposition as compared to the control group. The IR + tolDC group possessed stronger periosteal reaction and bone regeneration ability than IR group, while there was nearly no new bone formation in IR + mDC group. The quantitative analysis of BV/TV revealed the highest values in the control group, which were 1.69 and 3,73 times higher than that of the IR and IR + mDC groups respectively. There was no statistical difference in BV/TV values between the control group and IR + tolDC group. Additionally, tolDC-encapsulated hydrogel significantly increased the BMD of the newly-formed mandible than the IR and IR + mDC group, achieving levels comparable to the control group (Fig. [125]6B-C). HE staining of major organs showed that the irradiated rats suffered from renal cortical and glomerular atrophy. There were no obvious morphological changes of organs among irradiated rats, indicating the satisfactory safety profile of the cell therapy (Supplementary Fig. [126]S6). HE staining also showed substantial newly-regenerated bone extending from the defect margin toward the central region in both the control and IR + tolDC groups. The new bones had numerous bone marrow and vascular-like structures, suggesting robust osteogenic activity. In contrast, the IR group displayed limited bone regeneration accompanied by fibroblast-like components at the margin of the defect. Unabsorbed GelMA was encapsulated within fibrous tissue in this group. As for the IR + mDC group, there was an absence of new bone formation, and there were no osteocytes observed in bone lacuna in some bone fragments, namely the sequestrum (Fig. [127]6D). Masson staining indicated the emergence of nascent bone in the IR and IR + tolDC groups, while control group exhibited more depositions of mature bone within the defect area (Fig. [128]6E). OCN was an osteogenic marker expressed in mature bone cells and localized in the newly formed bone tissue. A significant reduction in OCN-positive areas was observed in both the IR and IR + tolDC group compared to the control cohort. Although tolDC administration partially rescued OCN expression compared to IR counterparts, the restored levels remained suboptimal relative to the control ones (Fig. [129]6F-G). Discussion Growing evidences have confirmed the importance of the immune microenvironment in bone repair [[130]30–[131]32]. High doses of IR promotes the recruitment and activation of different immune cells, thus breaking the homeostasis of the osteoimmune microenvironment, resulting in radiation-induced jaw injury and eventually osteoradionecrosis [[132]12, [133]33]. When exposed to diverse pathogenic stimuli, DCs exhibit either activated or suppressed states, modulating both inflammatory responses and adaptive immune responses, which may either exacerbate or relieve inflammatory jawbone diseases [[134]34, [135]35]. In this study, we intended to elucidate the role of dendritic cells in disease progression through the utilization of a well-established rat ORNJ model and cell experiments. A notable elevation of mature DC percentage within the irradiated jawbone marrow was observed compared to controls. Simultaneously, the irradiated rats exhibited more DCs in cervical lymph nodes concurrent with CD4⁺ T cell infiltration in the jawbone, implying DC involvement and functional activation, which might explain the disruption of immune homeostasis in the jawbone. BMDCs cultured with GM-CSF and IL-4 were used to investigate the effect of IR on DCs in vitro. BMDCs exhibit cellular heterogeneity [[136]36]. Some studies suggested that BMDCs resembled monocyte-derived inflammatory dendritic cells [[137]37]. Helft et al. [[138]38] performed unsupervised hierarchical clustering of the transcriptome of GM-CSF-induced DCs (GM-DC), revealing that GM-DC clustered most closely with migratory DCs, characterized by specific expression of maturation-related, migratory, and immunomodulatory transcripts [[139]39, [140]40]. Therefore, BMDCs only represent part of the DC population in vivo. IR-induced BMDCs acquired a unique semi-mature state, characterized by increased surface expression of the MHC-II and co-stimulatory molecules, coupled with enhanced migratory capacity through the NF-κB axis, while maintaining baseline levels of cytokine secretion and limited potential in MLR assay. Bacteria-derived LPS provided critical secondary signals for the complete maturation of DCs, accompanied by increased secretion of pro-inflammatory cytokines. Meyer proposed the theory of ‘radiation, trauma, and infection’, suggesting that bacterial infection is a primary factor of ORNJ [[141]9]. Post-RT dental extraction provides a favorable environment for bacterial colonization by oral pathogens such as Actinomyces spp., Streptococcus intermedius, etc [[142]41–[143]43], which greatly increases the incidence of osteomyelitis and osteonecrosis [[144]44]. DCs, as the sentinels of the immune system, can sense and alert the body to pathogens or danger signals [[145]45]. In radiation-induced jaw injury complicated by secondary infection, hyperactivation of DCs is likely to exacerbate regional inflammation, establishing a self-perpetuating circle of inflammation-necrosis-infection [[146]46]. Radiation-induced immune dysregulation seriously disrupts bone homeostasis, particularly affecting the function of BMSCs. For example, the expansion of B cells and CD8⁺ T cell subsets after RT impaired BMSC osteogenesis, leading to systemic bone loss [[147]47]. DCs serve as indispensable regulators within the osteoimmune microenvironment, establishing bidirectional modulatory control over bone remodeling through intercellular crosstalk with cells of the skeletal system [[148]15, [149]21, [150]48]. DC depletion directly hindered the heterotopic ossification of biomaterial [[151]49]. In our study, we found that irradiated mature DCs and DC-derived secretive cell factors triggered inflammatory responses in BMSCs, thereby suppressing proliferation, migratory ability, and osteogenic differentiation while enhancing adipogenic differentiation. These alterations were in accordance with the compositional remodeling observed in the bone marrow of radiation-induced jaw injury rat models. Although IR can impose direct cytotoxic effects on stromal cells, DCs further amplify the adverse effects on BMSCs. However, in the current study, we only discussed the synergistic effects of multiple cytokines of mDCs. The mechanistic contributions of a specific cytokine remain to be fully elucidated. We hypothesized that by suppressing inflammatory activation of DCs, radiation-induced injury could be ameliorated. Adoptive cell transfer using patients’ own tolDCs has been successfully translated to the clinic to treat autoimmune or chronic inflammatory diseases [[152]50, [153]51]. It has been reported that tolDCs can be generated using additional agents or lentiviral vector-based approaches [[154]52–[155]55]. Lentiviral transduction remains technically challenging. Previous studies have demonstrated modest efficiency (30–40%), or required higher vector doses—a strategy that might trigger undesired DC maturation or cell toxicity [[156]56, [157]57]. VitD3 is a potent natural modulator of both innate and adaptive immunity, selectively suppressing the NF-κB transcription in cDCs with adequate safety and efficacy to develop clinical-grade tolerogenic tolDCs [[158]58, [159]59]. GelMA has been widely used in tissue engineering to provide a physiologic microenvironment for transplanted cells because of its compositional similarity to natural extracellular matrices [[160]60, [161]61]. We verified that the tolDCs encapsulated GelMA effectively promoted bone regeneration in irradiated jaw defects. Regarding the therapeutic mechanisms of tolDCs according to recent studies, they promote hyporesponsiveness of T cells [[162]62, [163]63]. Besides, tolDCs impart a robust anti-inflammatory stimulus that mitigates inflammation through a by-stander effect [[164]64]. Nevertheless, the underlying molecular mechanisms of tolDCs therapy remain to be fully elucidated. Future studies should also test different cell doses on hydrogel to optimize therapeutic effect. Conclusion To summarize, our study highlights that the hyperactivation and maturation of DCs served as a potential pathogenic factor in radiation-induced jaw injury. DCs exacerbated the local inflammatory responses, induced oxidative stress, and abrogated the biological functions of BMSCs. The administration of tolDCs greatly promoted the progression of jawbone regeneration post-IR, suggesting that tolDCs might be a promising therapeutic strategy against radiation-induced jaw injury (Fig. [165]7). Fig. 7. [166]Fig. 7 [167]Open in a new tab Schematic conceptualization of this study. IR activated the NF-κB signaling pathway to promote activation and maturation of DCs, amplifying inflammatory signals through the release of pro-inflammatory cytokines. The mDCs triggered oxidative stress of BMSCs, showing increased intracellular ROS levels, inhibited osteogenic differentiation, and enhanced adipogenic differentiation, which accelerated the progression of radiation-induced jaw injury. VitD3-induced tolDCs exhibited IR resistance and anti-inflammatory properties, possessing the therapeutic potential to accelerate bone regeneration in irradiated jaw defects through local administration. (IR: ionizing radiation; mDC: mature dendritic cell; tolDC: tolerogenic dendritic cell; BMSC: bone marrow mesenchymal stem cell; ROS: reactive oxygen species; vitamin D3: vitD3) Electronic supplementary material Below is the link to the electronic supplementary material. [168]Supplementary Material 1^ (4.1MB, docx) Acknowledgements