Abstract Background Synovitis is a key factor in temporomandibular joint osteoarthritis (TMJOA) and could be an early sign of the disease. Notably, synovitis and its macrophage component represent a target of interest for developing treatments. Dental pulp stem cells (DPSCs) are derived from the neural crest, coincidentally with maxillofacial tissues, thus attracting significant interest in situ maxillofacial regenerative medicine. However, there is a relative scarcity of studies investigating the role of DPSCs in the temporomandibular joint (TMJ) synovial macrophage. Objective This study aimed to evaluate the regulatory and reparative capabilities of DPSCs on synovitis in TMJOA rat models and to elucidate their specific mechanisms of action on synovial macrophages. Method In vivo, three groups were established: the control group, which had neither induction nor treatment; the OA group, consisting of rats with OA but without any treatment; and the DPSCs treatment group, where rats with OA received DPSCs therapy. Progressive TMJOA was induced in rats via intra-articular injection of complete Freund’s adjuvant and sodium iodoacetate. After 2 weeks, DPSCs were injected into the joint cavity as a therapeutic intervention. Meanwhile, normal saline was injected into the joints of rats in both the control and OA groups. Four weeks after treatment, histological analysis was performed to evaluate the repair of synovitis. In addition, an in vitro co-culture system, consisting of macrophages and DPSCs. Each group was assessed using techniques, including mRFP-GFP-LC3 lentivirus transfection, Mito-Tracker Red and Lyso-Tracker Green staining, transmission electron microscopy, and Western blot analysis. Furthermore, apoptotic primary neutrophils were co-cultured with polarized macrophages to observe the phagocytic ability of macrophages towards apoptotic primary neutrophils under different treatments in vitro. Results In the rat model, compared with the OA group, the DPSCs group exhibited reduced cell infiltration and collagen deposition, along with elevated levels of anti-inflammatory CD206 and decreased levels of inflammatory CD86, and enhanced the ability of macrophages to phagocytize apoptotic cells in vivo and in vitro. Notably, DPSCs exhibited enhanced efficacy in upregulating autophagosome expression, promoting co-localization with mitochondria and lysosomes, and modulating the expression of mitochondrial mitophagy proteins. Furthermore, inhibition of mitophagy in M1 macrophages partially attenuated the M2 polarization effect and phagocytosis induced by DPSCs. Conclusion DPSCs significantly mitigate synovitis in TMJOA rats by enhancing M2 polarization and efferocytic functions of macrophages. This study underscores the potential of DPSCs as a therapeutic strategy for TMJOA by modulating synovial macrophage functions through the regulation of mitophagy. Graphical Abstract [41]graphic file with name 13287_2025_4468_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s13287-025-04468-2. Introduction Synovitis could potentially precede temporomandibular joint osteoarthritis (TMJOA) and is crucial in its progression [[42]1–[43]4]. The healthy synovial membrane comprises two layers: the lining surrounding the joint cavity and the sublining layer within the tissue. These layers predominantly comprise fibroblasts and tissue-resident macrophages, which are rich in blood vessels and facilitate the recruitment of cells during inflammatory responses [[44]5]. The balance within the joint depends on the joint’s microenvironment. In the temporomandibular joint (TMJ), synovial macrophages are the predominant cell type in the synovial membrane [[45]6]. In inflammatory conditions, there is a notable increase in the number of macrophages. The dysregulation in apoptotic processes of inflammatory macrophages and synovial fibroblasts underlies the pathogenesis of osteoarthritis. Professional phagocytes, including macrophages, are crucial in clearing apoptotic cells. The presence of dying cells in tissues is rarely noticeable, and apoptotic materials typically do not cause inflammation due to the remarkably efficient clearance process known as “efferocytosis“ [[46]7]. Efferocytosis eliminates cellular debris with inflammatory potential and initiates intracellular reprogramming, promoting pro-resolution signaling pathways [[47]8]. Depending on the immune context, macrophages can polarize into classically activated pro-inflammatory M1 macrophages or activated anti-inflammatory M2 macrophages. Studies have shown a significant rise in M1 macrophages within the inflamed synovial membrane [[48]9–[49]11]. Mesenchymal stem cells (MSCs) regenerative therapy holds promising potential for treating osteoarthritis. These cells interact with different innate immune cells, including macrophages and neutrophils [[50]12, [51]13]. Through direct contact or paracrine effects, MSCs can exert regulatory influence on macrophage maturation, migration, and proliferation [[52]4]. They possess the ability to induce the production of transforming growth factor-beta (TGF-ß) and arginase-1 (Arg-1) in macrophages, thereby promoting their polarization from an M1 phenotype towards an M2-like phenotype through secretion of cyclooxygenase-1 (COX-1) and prostaglandin E2 (PGE2) [[53]14]. Dental pulp stem cells (DPSCs) play a role in systemic immune regulation and suppression of inflammation. Their homology to craniofacial tissue makes them particularly promising for repairing craniofacial tissue. DPSCs have demonstrated safety and efficacy in treating stroke, COVID-19, and diabetes [[54]15–[55]17]. Their low immunogenicity enables their utilization by individuals and family members, while their storage value and therapeutic potential garner increasing attention. Consequently, DPSCs are poised to offer a safer and more regenerative alternative to traditional medicines [[56]18]. In a mouse model, DPSCs transfusion has been shown to manage local rheumatoid arthritis synovitis and autoimmune status effectively [[57]19]. However, there is a relative scarcity of studies investigating the role of DPSCs in TMJOA synovitis. Autophagy is a cellular degradation and recycling mechanism that plays a crucial role in maintaining the stability of the intracellular environment and resisting external stress. It regulates macrophage phagocytosis, antigen presentation function, and polarization direction to modulate their involvement in inflammation development and resolution [[58]20]. Artificial mitochondrial transplantation (AMT) can reverse the aging of mesenchymal stromal cells and improve their immunomodulatory properties in lipopolysaccharide (LPS)-induced synovial cell inflammation by upregulating autophagy [[59]21]. Autophagy plays an essential role in the occurrence and development of synovitis, and the regulation of autophagy may become a potential target for future treatment of synovitis [[60]22]. Mitophagy is a selective autophagy that removes damaged mitochondria [[61]23]. Previous studies have demonstrated that TNF-α induces mitophagy in synovial fibroblasts of rheumatoid arthritis, and inhibition of this process can ameliorate arthritic synovitis [[62]24]. However, a dearth of research investigating mitophagy in macrophages associated with TMJ synovitis exists. Therefore, further investigation is imperative to comprehensively elucidate the autophagy mechanisms underlying synovitis in TMJOA. Given the crucial role of synovitis in the progression of TMJOA, and the current insufficient understanding of the potential of DPSCs in the treatment of TMJOA synovitis, this study aims to explore the hypothesis that DPSCs alleviate TMJOA synovitis by regulating macrophage polarization and efferocytosis. Specifically, we hypothesize that DPSCs can alleviate synovial inflammation by regulating autophagy, promoting the polarization of synovial macrophages from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, and enhancing their efferocytosis. This study aims to clarify the effects of DPSCs on macrophage polarization and efferocytosis in synovitis of TMJOA, and to explore the potential therapeutic mechanism by which DPSCs regulate the inflammatory microenvironment of TMJ. Methods Animal experiments in vivo Animal model The Sprague-Dawley (SD) rats utilized in this study were procured from the Laboratory Animal Research Center at Zhejiang Chinese Medical University. This study received approval from the Ethics Committee of Zhejiang Chinese Medical University on Feb 6, 2023 (Project name: Investigation into the Mechanism of Cartilage Repair by Dental Pulp Mesenchymal Stem Cells in Temporomandibular Arthritis. Ethics Approval Number: IACUC-20221031-14). A total of 18 SD rats were randomly assigned to three groups: the Control group (n = 6), the OA group (n = 6), and the DPSCs group (n = 6). To induce TMJOA, a mixture of 50 µL of monosodium iodoacetate (MIA) at a concentration of 0.5 mg (Sigma-Aldrich, USA), 25 µL of normal saline (NS), and 25 µL of complete Freund’s adjuvant (CFA) (Sigma-Aldrich, USA) was administered intra-articularly once into both TMJs and induced for 2 weeks (Fig.[63]S1). The control group did not receive any induction during this phase. Following the 2-week MIA/CFA induction period, the OA and DPSCs groups underwent weekly intra-articular injections into bilateral TMJs for 4 consecutive weeks. Specifically, the OA group received 50 µL of normal saline, while the DPSCs group received 50 µL of DPSCs suspension. Meanwhile, in the control group, 50 µL of normal saline was injected into the bilateral temporomandibular joints once a week for 4 consecutive weeks. These injections were administered using the 30G insulin needle (Becton, Dickinson and Company, USA). The rats exhibited no obvious signs of suffering throughout the experimental period. At the end of the 4-week treatment period, the rats were euthanized via intraperitoneal injection of Shutai 50 solution (BIOFIVEN, China) at a dose of 20 mg/kg body weight, with anesthesia being achieved within 3–5 min. This study adhered to the ARRIVE guidelines 2.0 for the reporting of in vivo experiments (Fig. [64]1). Fig. 1. Fig. 1 [65]Open in a new tab Injection time point diagram Histology and immunohistochemical/immunofluorescence staining For the histological examination, samples from the temporomandibular joints were initially fixed in 4% paraformaldehyde for 3 days. Subsequently, they were decalcified in a 14% Ethylene Diamine Tetraacetic Acid (EDTA) solution at room temperature for 90 days. The samples were then systematically dehydrated using an automatic dehydrator and embedded in paraffin. Sagittal sections, each 3 μm in thickness, were cut from the paraffin blocks. After deparaffinization and rehydration, the sections were stained with Hematoxylin-Eosin (HE) and Masson staining for morphological observation. The assessment of the synovial membranes was performed on H&E-stained slides, focusing on three key features: the synovial lining cell layer, stromal cell density, and inflammatory infiltrate. The above indicators were semiquantitatively evaluated (from 0, absent to 3, strong), and each feature was graded separately. The scores for these parameters were then combined and interpreted as follows: a total score of 0–1 indicated no synovitis, 2–4 indicated low-grade synovitis, and 5–9 indicated high-grade synovitis [[66]25]. Histology sections were utilized for immunohistochemical (IHC) and immunofluorescent (IF) staining. The slices were initially subjected to dewaxing, followed by rehydration and subsequent washing in phosphate buffer (PBS) for three cycles of 5 min each. Subsequently, the slices were soaked overnight in a citric acid solution in a water bath at 60℃ to facilitate antigenic repair. After repair, the slices were washed three times with PBS and then treated with endogenous peroxidase blocker at room temperature for 10 min before being rewashed with PBS three times. The sections were then sealed with 5% goat serum at room temperature for 1 h (IHC staining only). Next, the sections were incubated overnight at 4℃ with the primary antibody. Then, the secondary antibody was applied using IHC or IF staining techniques and incubated at room temperature for 1 h. For IHC-stained sections, diaminobenzidine and hematoxylin stains were used before dehydration and sealing took place. For IF-stained slices, DAPI solution was applied followed by sealing using glass slides. Antibodies used for IHC/IF staining were as follows: P62 (HUABio, R1309-8, China), Beclin1 (ABclone, A7353, China), LC3B (Novusbio, NB100-2220, USA), CD206 (Proteintech, 18704-1-AP, China), CD86 (Proteintech, 13395-1-AP, China), Col2 (Abcam, ab39012, UK), MMP13 (Abcam, ab39012, UK). TUNEL staining The TUNEL assay was conducted following the protocol provided by the manufacturer (TUNEL BrightGreen Apoptosis Detection Kit, Vazyme Biotech, China) to identify cell death within the synovial membrane. This assay leveraged the green channel at a wavelength of 488 nm for detection. DAPI was employed as a counterstain to visualize the nuclei detected in the blue channel at 460 nm. The imaging process was carried out using a fluorescent microscope (Olympus BX43, Japan). Access to gene expression omnibus (GEO) datasets The dataset of relevant studies was retrieved from the GEO database, and the [67]GSE49604 dataset was downloaded. The gene expression profile, [68]GSE49604 on the [69]GPL8432 platform, was found for further analysis. The experimental group comprised primary human synovial macrophages from patients with arthritis, while the control group consisted of primary human peripheral blood mononuclear cells from healthy individuals. Utilize GEO2R for differentially expressed genes (DEGs) between macrophages of human synovitis and healthy macrophages, followed by subsequent visualization and statistical analysis of the DEGs. The threshold was set to obtain DEGs as absolute log fold change|log[2]FC|>1 and P < 0.05. The heatmap was plotted by [70]https://www.bioinformatics.com.cn, an online platform for data analysis and visualization. Cell culture in vitro Cell culture The RAW264.7 macrophages were sourced from Punosai Life & Technology Co., LTD. These cells were cultured in Dulbecco’s modified Eagle medium supplemented with 10% Fetal Bovine Serum (FBS), 100 U/ml penicillin, and 100 µg/ml streptomycin and incubated in a humidified incubator with 5% CO[2] at 37 °C. After three passages, the cells were harvested for subsequent experiments. Additionally, 100 ng/mL LPS (Sigma-Aldrich, USA) and 10 ng/mL IFN-γ (PeproTech, USA) were used to differentiate RAW264.7 cells into M1-type macrophages. The DPSCs were generously provided by the Institute of Medicine, Chinese Academy of Sciences & Tongce Biological Dental Stem Cell Bank and Research and Development Center and the donors had signed informed consent [[71]26]. The DPSCs were cultured with alpha modification of Eagle’s medium (containing 10% fetal bovine serum) at 37 °C with 5% CO[2]. DPSCs were identified using a previously described method, and cells with three passages were used [[72]26] (Fig.[73]S2). RAW264.7 cells and DPSCs co-culture system The study was divided into four groups: the control group, the LPS group, the LPS + DPSCs group, and the LPS + DPSCs + Mdivi-1 group. In the control group, RAW264.7 macrophages received no treatment. For the LPS group, RAW264.7 macrophages were differentiated into M1-type macrophages using 100 ng/mL LPS (Sigma-Aldrich, USA) and 10 ng/mL IFN-γ (PeproTech, USA). A co-culture model was established with DPSCs (2 × 10⁶/well) in the upper layer and RAW264.7 macrophages (2 × 10⁵/well) in the lower layer. In the LPS + DPSCs group, the cells were stimulated with LPS and IFN-γ for 24 h before being co-cultured with DPSCs for 24 h. In the LPS + DPSCs + Mdivi-1 group, RAW264.7 macrophages were pre-treated with the mitophagy inhibitor Mdivi-1 (25 µmol/L, Abcam, UK) for 3 h before LPS/IFN-γ treatment and subsequent co-culture with DPSCs [[74]23]. After a 24-h co-culture period between DPSCs and RAW264.7 cells, RAW264.7 cells were systematically harvested for further experimental analysis (Fig. [75]2). Fig. 2. Fig. 2 [76]Open in a new tab In vitro model diagram of cell co-culture. DPSCs were placed in the upper chamber, and RAW264.7 cells were cultured in the lower chamber Western blot The RAW264.7 cells of the above groups were lysed in RIPA buffer (Beyotime, Shanghai, China) for 20 min, followed by centrifugation at 12,000 g for 4 min. The primary antibodies of Beclin1 (Proteintech, 11306-1-AP, China), P62 (Abcam, ab211324), LC3B (Novusbio, NB100-2220, USA), Parkin (Proteintech, 14060-1-AP, China), PINK1 (Proteintech, 23274-1-AP, China), Arginase-1 (Proteintech, 16001-1-AP, China) and GAPDH (Abcam, ab181602, UK) were utilized. The membranes underwent incubation with primary antibodies, followed by the application of horseradish peroxidase-conjugated secondary antibodies to facilitate the detection process. The relative gray level of the protein bands was determined using an enhanced chemiluminescence system (ASPEN, AS1059, China). The AlphaEaseFC software processing system ensured accurate protein expression level quantification. Autophagy detection through transmission electron microscopy The RAW264.7 cells of the above groups were first gently washed twice with phosphate-buffered saline (PBS) to remove any residual substances, followed by fixation in 2.5% glutaraldehyde (Sinopharm, China) for 24 h. Subsequently, the samples were post-fixed with 1% osmic acid (Sinopharm, China) for 2 h, dehydrated using graded ethanol (Sinopharm, China), and then infiltrated and embedded with epon 812 (Sinopharm, China). Ultrathin slices of the samples were sectioned using an ultramicrotome (Leica, Germany) and examined under Transmission Electron Microscopy (TEM) (Thermo Fisher Scientific, USA). Autophagy detection through mRFP-GFP-LC3 adenovirus transfection The RAW264.7 cells in each group were transfected with the Ad-mRFP-GFP-LC3 adenoviral vector to detect the autophagic flux levels. Before transfection, the Ad-mRFP-GFP-LC3 adenoviral vector (Hanbio Biotechnology, China) was retrieved from the − 80℃ freezer and thawed slowly on ice. Based on the number of cells, the adenovirus was diluted to the working concentration to achieve a multiplicity of infection (MOI) of 50. The diluted adenoviral solution was added to the cell culture wells to ensure adequate contact with the cells. The culture plate was then incubated at 37℃ in a 5% CO₂ incubator for 4 h. After incubation, the adenovirus culture medium was aspirated and replaced with fresh complete culture medium (DMEM containing 10% FBS). After the cells were further cultured for 72 h, the fluorescence signals of mRFP-GFP-LC3 in the cells were observed and imaged using confocal laser scanning microscopy (Zeiss, Germany), and the autophagy-related changes were analyzed. Autophagy detection through lysosome and mitochondria colocalization The distribution relationship between mitochondria and lysosomes in the above groups of RAW264.7 macrophages was explored through fluorescence staining and colocalization analysis. The mitochondria of live cells were stained with a working solution of Mito-Tracker Red (Beyotime, China) at a concentration of 100 nM and incubated at 37℃ for 20 min. Subsequently, the cells were further stained with a working solution of Lyso-Tracker Green (Beyotime, China) at a concentration of 50 nM and incubated at 37℃ for 15 min. Cell nuclei were counterstained with Hoechst 33,342. The co-localization analysis of lysosomes and mitochondria was performed using ImageJ software. Efferocytosis assay Primary neutrophils were isolated from C57BL/6 mice and then cultured in RPMI-1640 medium (Hyclone, USA) for 48 h without serum to induce apoptosis. The primary neutrophils were subsequently labeled with PKH67 dye (Sigma-Aldrich, USA) following the manufacturer’s instructions. Then, PKH67-labeled apoptotic primary neutrophils were added in the Control, LPS, LPS + DPSCs, and LPS + DPSCs + Midiv1 groups at a ratio of 10:1, and incubated for another 48 h. Subsequently, the cells were thoroughly washed three times with PBS to eliminate unengulfed primary neutrophils. The cells were subsequently labeled with Dil (Beyotime, China) solution according to the manufacturer’s instructions. The engulfment ability of macrophages towards apoptotic primary neutrophils was visualized using immunofluorescence microscopy. The efferocytotic index was calculated as follows: (number of macrophages containing apoptotic bodies)/(total macrophages) × 100%, then normalized to the control group set as 100%. Statistical analysis All experiments were conducted with a minimum of three repetitions to ensure the reliability of the results. The data obtained were presented as the means ± standard deviation. Data was analyzed using GraphPad Prism 8.0 software (GraphPad Software, USA). To discern differences between the two groups, Student’s t-test was employed. When comparing three or more groups, a one-way analysis of variance (ANOVA) was utilized to assess the variability among the groups. Subsequently, Tukey’s test was applied to determine the specific between-group differences. For this study, statistical significance was defined as P < 0.05. Result DPSCs reduce synovial inflammation and fibrosis Firstly, we sought to assess the protective impact of DPSCs on synovitis in vivo. The OA rat model was induced by injecting complete Freund’s adjuvant (CFA) and sodium iodoacetate (MIA). The animals were divided into Control, OA, and DPSCs groups, which received weekly injections of normal saline (NS) and DPSCs suspension for 4 weeks. After 4 weeks of DPSCs injections into the joint cavity, TMJ joint specimens of rats were collected for Masson and HE staining. The morphology of the TMJ synovium in the control group was normal. However, following the induction of arthritis via MIA/CFA, OA rats exhibited pronounced pathological alterations in TMJ synovium, characterized by synovial collagen fibrohyperplasia accompanied by vascular opacity (as evidenced by Masson staining) and infiltration of inflammatory cells (demonstrated through HE staining). After four weeks of treatment with DPSCs, the DPSCs group showed a marked reduction in the number of synovial lining cell layers and blood vessels, alongside a decrease in both the density and quantity of infiltrating inflammatory cells and collagen fibers within the synovial tissue when compared to the OA group (Fig. [77]3a-b). The synovitis scores of the OA groups were significantly higher than those of the control and DPSCs groups (Fig. [78]3c). The findings suggest that applying DPSCs significantly attenuates synovial inflammation and fibrosis in OA synovitis. Fig. 3. [79]Fig. 3 [80]Open in a new tab DPSCs ameliorate synovium inflammation and synovial fibrosis in TMJOA synovitis. (a-b) The synovium thickness, inflammatory cell infiltration, and collagen fiber deposition in the Control, OA and DPSCs groups were assessed by Masson’s trichrome and HE staining, respectively. Scale bar: 20 μm. Amplification region: 10 μm. (c) Statistical analysis of synovitis scores in HE-stained sections. Data are presented as mean ± SD. Statistical significance was determined using one-way ANOVA with Tukey’s post hoc test. ***p < 0.001, ****P < 0.0001 DPSCs promote synovial macrophage M2 polarization and efferocytosis Subsequently, we employed immunofluorescence to assess the shift in polarization of TMJ synovial macrophages. Notably, after the administration of MIA and CFA, there was a significant increase in both the M1 macrophage-associated markers (CD86) and the M2 macrophage-associated markers (CD206) within the synovial membrane of the OA group. In comparison, the group treated with DPSCs displayed a significant upregulation of the M2 macrophage marker CD206 and a concurrent downregulation of the pro-inflammatory M1 macrophage marker CD86 when contrasted with the OA group (Fig. [81]4a-d). Efferocytosis is an indispensable process through which dead and dying cells are removed by phagocytic cells. In this study, immunofluorescent TUNEL staining showed that the number of apoptotic cells in synovial tissue increased with the progression of inflammation, and the number of apoptotic cells in the DPSCs group was down-regulated. Immunofluorescence staining of CD206 and TUNEL revealed that the injection of DPSCs augmented the co-localization between CD206^+ macrophages and apoptotic cells labeled with TUNEL in vivo, thereby further substantiating the capacity of DPSCs to induce polarization of synovial macrophages towards an M2 phenotype and enhance their phagocytic activity towards apoptotic cells in OA synovium (Fig. [82]4e-f). To investigate the effects of DPSCs on macrophage phagocytosis and function in vitro, we stimulated RAW264.7 cells with LPS/γ-IFN to simulate an inflammatory environment and co-cultured these LPS-stimulated cells with DPSCs for 24 h. Subsequently, the macrophages were stained with Dil and co-cultured with apoptotic neutrophils labeled with PKH67 for 48 h to assess the phagocytosis level. As anticipated, it was also confirmed in vitro that the efferocytosis function of macrophages was impaired in the LPS microenvironment, resulting in a reduced ability of macrophages (Dil-labeled) to effectively clear fluorescent apoptotic neutrophils (PKH67-labeled) compared with the control group. In addition, co-culture with DPSCs enhanced the phagocytosis activity of macrophages (Fig. [83]4g-h). Based on these findings, we propose that DPSCs can potentially promote the polarization of M1 synovial macrophages toward the M2 phenotype and enhance their efferocytosis. Fig. 4. [84]Fig. 4 [85]Open in a new tab DPSCs promote the polarization of synovial macrophages towards the M2 phenotype and enhance their phagocytosis in TMJOA synovitis. (a-b) Immunohistochemistry staining was performed to visualize the expression and localization of CD86 and CD206. Scale bar: 50 μm. (c-d) Quantitative analysis of CD86 and CD206 protein expression. (e) The expression and co-localization of CD206 and TUNEL were observed in synovial tissue by immunofluorescent staining. Scale bar: 50 μm. (f) Quantification of CD206^+ macrophages engulfing apoptotic cells as a proportion of total CD206^+ cells. (g) Fluorescent staining for Dil (red) in RAW264.7 cells and PKH67 (green) in apoptotic neutrophils after co-culture for 48 h. Scale bar: 20 μm. (h) Quantification of Dil-PKH67-positive macrophages (yellow) as a proportion of total macrophages. Data are presented as mean ± SD. Statistical significance was determined using one-way ANOVA with Tukey’s post hoc test. *p < 0.05, **p < 0.01, ***p < 0.001, ****P < 0.0001 Autophagy is identified as the key signal in macrophages of synovitis To elucidate the mechanisms underlying the therapeutic effects of DPSCs on macrophages, we searched the National Center for Biotechnology Information (NCBI) GEO database. The differentially expressed genes (DEGs) in the gene expression profiles ([86]GSE49604) were identified using GEO2R, an online data analysis tool. Subsequently, the resulting dataset was analyzed using volcano plots to visualize the DEGs. We identified 507 DEGs between macrophages of human synovitis and healthy macrophages (Fig. [87]5a). Gene Ontology (GO)/Kyoto Encyclopedia of Genes and Genomes (KEGG) heatmap was plotted by [88]https://www.bioinformatics.com.cn, an online data analysis and visualization platform. KEGG pathway enrichment analysis showed that these genes were involved in many signaling pathways, mainly related to lysosomal, autophagy, and apoptosis activity (Fig. [89]5b, d). As shown in Fig. [90]5c, the top two enriched biological process (BP) terms were the neutrophil activation involved in immune response and neutrophil degranulation. Taken together, autophagy activity and the immune response involved in neutrophils may play an essential role in macrophages of synovitis. Fig. 5. [91]Fig. 5 [92]Open in a new tab Functional enrichment based on DEGs. (a) Volcano plot depicting the distribution of DEGs between human arthritis macrophages and healthy controls from the [93]GSE49604 dataset. Red, blue, and gray dots represent gene expression levels corresponding to upregulated, downregulated, and insignificant expression. DEGs, differentially expressed genes. (b) KEGG pathway enrichment analysis of DEGs in human arthritis macrophages and healthy controls. KEGG, Kyoto Encyclopedia of Genes and Genomes. (c) GO-BP analysis for DEGs shows significant terms through (c) a bubble plot. GO, Gene Ontology; BP, biological process; (d) Gene expression in the adherens junction pathway was plotted by PATHVIEW DPSCs enhance macrophage M2 polarization and efferocytosis by enhancing mitophagy LC3B, P62, and Beclin1 are the most commonly used markers to indicate autophagy activity. Consistent with our expectations, we observed a significant upregulation of Beclin1 and LC3B expression and a significant downregulation of P62 expression in the synovium of the OA group. These findings suggest that MIA/CFA-induced mitophagy can be activated, and DPSCs further enhance mitophagy to suppress MIA/CFA-induced inflammation in vivo (Fig. [94]6a-d). To elucidate the mechanism by which DPSCs regulate macrophages, we established an in vitro co-culture system of DPSCs and macrophages by stimulating RAW264.7 cells with LPS and γ-IFN. To confirm the regulatory effect of DPSCs on macrophage polarization in vitro, macrophages were pre-stimulated with LPS and then co-cultured with or without DPSCs. Immunofluorescence staining of macrophages showed that DPSCs reduced the expression of CD86 ^+ cells and promoted polarization toward the M2 phenotype (CD206 ^+ cells) (Fig. [95]6e-f). These results demonstrate that DPSCs inhibit LPS-stimulated macrophage M1 polarization and reverse LPS-suppressed macrophage M2 polarization. These findings indicate that DPSCs may promote the M2 polarization of macrophages by autophagy, thereby mitigating synovitis. Fig. 6. [96]Fig. 6 [97]Open in a new tab DPSCs promote autophagy and M2 polarization in macrophages. (a) Immunohistochemical analysis of autophagy-associated proteins P62, Beclin1 and LC3 in synovial macrophages of various treatment groups. Scale bar: 10 μm. (b-d) Quantifying the histomorphometric analysis of the expression of proteins P62, Beclin1 and LC3. (e-f) Immunofluorescence staining of CD86 and CD206 in RAW264.7 macrophages. Scale bar: 10 μm. Data are presented as the mean ± SD. Statistical significance was determined using one-way ANOVA with Tukey’s post hoc test. *p < 0.05, **p < 0.01, ***p < 0.001, ****P < 0.0001 Furthermore, transmission electron microscopy illustrated that LPS and γ-IFN stimulation escalated the number of autophagosomes within macrophages. TEM analysis revealed an increase in the number of autophagosomes following treatment with DPSCs in M1 macrophages (Fig. [98]7a). We utilized an RFP-GFP-LC3 reporter assay to investigate autophagic flux in cells treated with LPS and DPSCs. This assay reveals autophagosome formation through the visibility of both RFP and GFP signals, presenting as yellow puncta. Upon the fusion of autophagosomes with lysosomes, the acidic environment quenches GFP signals, leaving only RFP signals and, thus red autolysosomes visible. Our findings indicated that DPSCs prompted an increase in autophagosome formation (yellow) (Fig. [99]7b, f). Next, we evaluated the expression of mitophagy-associated proteins in M1-polarized macrophages. We found that P62 expression was decreased, while Becin1 and LC3 II/I ratio protein expression levels were significantly increased in LPS-induced RAW264.7 cells. The expression of P62 protein in RAW264.7 cells was further downregulated. In contrast, Becin1 and LC3 II/I ratio protein expression were further upregulated upon LPS-induced co-culture of RAW264.7 cells with DPSCs (Fig. [100]7c, d). Full-length blots/gels are presented in Supplementary Figure [101]S3. The mitophagy pathway encompasses various mechanisms, with the most classical and comprehensively understood being the Pink1/Parkin pathway. To elucidate the impact of DPSCs on mitophagy, we evaluated the activation level of the PINK1/Parkin pathway. Western blot analysis results demonstrated that LPS stimulation upregulated the expression of PINK1 and Parkin in M1 macrophages, and treatment with DPSCs further augmented PINK1 and Parkin expression (Fig. [102]7e, g). Full-length blots/gels are presented in Supplementary Figure [103]S4. Fig. 7. [104]Fig. 7 [105]Open in a new tab DPSCs enhance macrophage mitochondrial autophagy in vitro. (a) The visualization of the ultrastructure of macrophages via transmission electron microscope (TEM). In the images, membrane-like vesicles in macrophages were observed. The distribution of autophagosomes (green circle) in macrophages was visualized using the TEM. Scale bar: 1 μm. Amplification region: 500 nm. (b, f) Autophagocytic flux in macrophages was analyzed by mRFP-GFP-LC3 (yellow puncta). Scale bar: 10 μm. (c-d) Western blot showing P62, Beclin1 and LC3-II/I expression in macrophages after various treatments. (e, g) Western blot showing the expression of PINK1 and Parkin in macrophages after various treatments. Data are presented as the mean ± SD. Statistical significance was determined using one-way ANOVA with Tukey’s post hoc test. *p < 0.05, **p < 0.01, ***p < 0.001, ****P < 0.0001 To investigate the interplay between macrophage mitophagy and M2 polarization, we implemented a simultaneous intervention involving LPS, DPSCs, and Mdivi-1 to impede mitochondrial mitophagy. Furthermore, fluorescence microscopy revealed that LPS induction significantly increased the co-localization of mitochondria and lysosomes in macrophages. The overlapping signals of MitoTracker and LysoTracker were significantly improved in M1 macrophages following co-culture with DPSCs, indicating that DPSCs could regulate mitochondrial autophagy in M1 macrophages. The application of the mitochondrial autophagy inhibitor, Midiv-1, markedly reduced the colocalization signals between MitoTracker and LysoTracker (Fig. [106]8a-b). The Western blot analysis revealed an upregulation of Arg-1 (M2 macrophage-associated markers) expression in M1 macrophages upon co-culture with DPSCs. Furthermore, treatment with Mdivi-1 significantly attenuated the expression of Arg-1 compared to the LPS + DPSCs group (Fig. [107]8c-d). Full-length blots/gels are presented in Supplementary Figure [108]S5. The above assessments encompassed alterations in M2 polarization among macrophages across varied groups, revealing that mitophagy suppression partially annulled the M2 polarization enhancement of DPSCs. Moreover, the ratio of phagocytic apoptotic neutrophils in the Midiv-1 treatment group decreased significantly (Fig. [109]8e, f). In conclusion, the data indicated that mitophagy may promote the phagocytosis and polarization of macrophages. Fig. 8. [110]Fig. 8 [111]Open in a new tab DPSCs facilitate M2 polarization and effercytosis of macrophages by inducing mitophagy. (a-b) The autolysosomes, mitochondria, and nucleus were labeled by Lyso-Tracker Green and Mito Tracker Red, respectively. Scale bar: 20 μm. (c-d) Representative Western blot image of Arg1 in macrophages after treatment of LPS, DPSCs, and Midivi-1. (e-f) Fluorescent staining for Dil (red) in RAW264.7 cells and PKH67 (green) in apoptotic neutrophils after co-culture for 48 h. Scale bar: 20 μm. Phagocytosis was observed under the fluorescence microscope. Data are presented as the mean ± SD. Statistical significance was determined using one-way ANOVA with Tukey’s post hoc test. *p < 0.05, **p < 0.01, ***p < 0.001, ****P < 0.0001 Discussion Synovial inflammation, also known as synovitis, is a crucial factor in the progression of osteoarthritis from its earliest clinical stages and is a key driver of pain initiation [[112]27, [113]28]. Current research indicates that DPSCs have some potential in controlling symptoms of synovitis, but most studies have focused on their effects on fibroblast-like synoviocytes (FLSs). These studies have revealed that DPSCs can mitigate the pathological course of synovitis by influencing FLSs and the secretion of inflammatory factors [[114]19, [115]29]. However, the pathogenesis of synovitis is complex, involving not only FLSs but also the interaction of various immune cells, especially the polarization state of macrophages, which plays a key role in inflammatory responses. In this study, we aim to investigate the impact of DPSCs on macrophages in TMJOA. Based on our previous findings and a comprehensive literature review, we propose that DPSCs play a crucial role in synovial inflammation and macrophage polarization. The homing ability of stem cells facilitates their targeting of inflamed and damaged areas, potentially expediting the repair process [[116]30]. Moreover, stem cell homing has the potential to modulate macrophage polarization and thereby regulate the inflammatory response [[117]4, [118]31]. DPSCs have garnered attention for their potential to combat inflammation and prevent fibrotic tissue formation, positioning them as viable options for addressing various conditions related to the immune system, inflammation, and fibrosis [[119]32]. The preliminary histological evaluation demonstrated that DPSCs effectively mitigated synovitis, as evidenced by reduced in collagen fibroplasia and inflammatory cell infiltration. These findings support our initial hypothesis and highlight the potential therapeutic role of DPSCs in modulating synovial inflammation. Synovial macrophages play a role in regulating joint inflammation and OA severity [[120]33, [121]34]. M1 macrophages release proinflammatory cytokines, while M2 macrophages release anti-inflammatory cytokines that contribute to the resolution of inflammation [[122]35]. The new study found that electrical signals improve DPSC-mediated paracrine patterns by up-regulating macrophage M2 polarization [[123]17, [124]36]. In addition, the conditioned medium (CM) of stem cells from human exfoliated deciduous teeth (SHEDs) can exhibit significant anti-inflammatory effects on synovitis by inducing the polarization of M2 macrophages. However, it remains unclear which specific secretory factors or signaling pathways play a dominant role in inducing the polarization of M2 macrophages [[125]37]. Consistent with these findings, we discovered that DPSCs induced M2 polarization of TMJ synovial macrophages in vivo and in vitro, but the specific mechanism remained unclear. Through further bioinformatics analysis, we speculate that macrophage autophagy and immune activities may play a crucial role in the pathogenesis of synovitis. However, the impact of DPSCs on macrophage autophagy remains to be elucidated. Additionally, the intricate relationship between autophagy and macrophage polarization is still not fully understood. In inflammation, efferocytosis by macrophages is a key process for clearing apoptotic cells, crucial for maintaining inflammatory balance and tissue repair. When large numbers of neutrophils are recruited to the site of injury in the early stages of inflammation and complete their phagocytic and bactericidal tasks, they rapidly enter apoptosis. If these apoptotic neutrophils are not cleared promptly, they may release pro-inflammatory contents, further exacerbating the inflammatory response [[126]38]. A previous study indicated that individuals with knee osteoarthritis exhibit a higher prevalence of apoptotic synovial cells than healthy controls. The fraction of synovial tissue macrophages participating in efferocytosis and the amount of material engulfed by individual macrophages significantly decreased in patients with osteoarthritis [[127]39]. Consistent with these findings, a notable decrease in the proportion of synovial tissue macrophages engaged in efferocytosis was observed in TMJOA rats. In light of these observations, we further investigated the effects of DPSCs on macrophage function. Our results demonstrate that the administration of DPSCs significantly enhances the phagocytic activity of macrophages, both in vivo and in vitro. This finding is novel and has not been previously reported, suggesting that DPSCs may play a unique role in modulating macrophage function and promoting the clearance of apoptotic cells in inflammatory conditions. The reprogramming of macrophage polarization is closely associated with autophagy [[128]40, [129]41], a process that elucidates MSC-mediated immunomodulation [[130]42]. Research suggests that the absence of essential autophagy genes or specific autophagy-related components can lead to increased production of TNF-αand IL-6, key proinflammatory cytokines. This deficiency also promotes the development of proinflammatory cell phenotypes typical of M1 macrophages. These effects have been observed in both in vivo animal models and in vitro cell culture systems, highlighting the critical role of autophagy in regulating inflammatory responses and the balance between M1 and M2 macrophage polarization [[131]43]. Low-intensity pulsed ultrasound attenuated mature IL-1β production primarily by augmenting autophagy-mediated degradation of pyruvate kinase, muscle, and sequestosome1 in macrophages, thereby ameliorating synovitis [[132]44]. Some researchers synthesized ROS reactive polymer, which self-assembled with astaxanthin and autophagy activator rapamycin to form nanoparticles. These nanoparticles could restore mitochondrial membrane potential, induce intracellular autophagy, and effectively repolarize M1 macrophages into M2 phenotype, thus significantly slowing down the progression of synovitis [[133]45]. These findings indicate a potential modulation of synovial macrophage polarization by autophagy, yet definitive evidence is currently lacking. Mitophagy has recently garnered increasing attention in the field. It is a crucial intracellular clearance mechanism for maintaining mitochondrial function and quality, effectively modulating cellular metabolism, survival, and programmed cell death pathways [[134]46]. The novel functionalized nanoparticles have modulated synovial macrophage pyroptosis and mitophagy interactions to mitigate osteoarthritis progression [[135]34]. However, research on TMJOA synovitis mitophagy remains scarce. In this research, we investigated the impact of LPS and DPSCs on mitophagy levels in macrophages. The findings suggest that co-culture with DPSCs increases mitophagy in macrophages, which is associated with M2 polarization and reduced inflammation. Additionally, DPSCs induce mitophagy in a PINK1/Parkin pathway-dependent manner. The PINK1/Parkin pathway is a well-researched mechanism that triggers mitophagy, a process where damaged mitochondria are targeted for degradation. When mitochondria are damaged, their membrane potential decreases, preventing PINK1 from moving to the inner membrane. Instead, PINK1 accumulates on the outer mitochondrial membrane. Here, it interacts with the translocase of the outer mitochondrial membrane, leading to its phosphorylation at Ser65. This phosphorylation activates Parkin, a ubiquitin ligase, which then ubiquitinates mitochondrial proteins, marking the damaged mitochondria for degradation [[136]47, [137]48]. The ubiquitin chains are recognized by p62, an autophagy adaptor protein. p62 connects LC3 to polyubiquitinated proteins during autophagosome formation, targeting them for degradation. Finally, p62 is degraded in the autolysosome, completing mitophagy [[138]48]. Consistent with the classical theory of mitophagy, our study showed increased mitophagy in the OA model, further enhanced by DPSCs treatments. This enhancement is evidenced by the upregulation of mitophagy-associated proteins, including LC3, PINK1, Parkin, and Beclin1, coupled with the downregulation of the mitophagy-inhibitory protein P62. It has been reported that IFN-γ induced autophagy of cervical cancer cells, further promoting macrophage phagocytosis [[139]49]. To demonstrate the correlation between mitophagy and macrophage polarization and phagocytosis, we employed Mdivi-1 to inhibit mitophagy and observed a decrease in the M2 polarization index and phagocytosis capacity of macrophages. These findings suggest that DPSCs promote mitophagy in macrophages, thereby modulating M2 polarization and efferocytosis, consequently alleviating synovial inflammation. This study is groundbreaking as it unveils the role and mechanism of DPSCs in TMJ synovitis. Furthermore, it demonstrates the correlation between mitochondrial autophagy and synovial macrophage polarization and function. Nevertheless, the study has certain limitations. Currently, investigating the interplay between stem cells and macrophages has emerged as a prominent research area. The novel study found that “superactivated” macrophages release the lipid metabolite 11,12-EET through Gasdermin D, mediating the metabolic communication between macrophages and stem cells and thus promoting the repair of tissue damage [[140]50]. However, in our study, a detailed exploration of the impact of DPSC on macrophage mitophagy mechanisms was lacking. Although our study evidenced the participation of PINK1/Parkin in mitophagy initiation, the precise processes through which DPSCs trigger the PINK1/Parkin pathway and how mitophagy induces M2 polarization and macrophage efferocytosis remain ambiguous, demanding further investigation. Conclusion In conclusion, our comprehensive investigation has elucidated the pivotal role of DPSCs in modulating macrophage polarization both in vitro and in vivo. Through intra-articular injection, DPSCs have demonstrated a remarkable capacity to mitigate the infiltration of M1-polarized macrophages while concurrently augmenting the presence of M2-polarized macrophages within rat synovial tissue. Additionally, the enhancement of macrophage efferocytosis function further underscores the therapeutic potential of DPSCs. Notably, our study has unraveled the intricate involvement of mitophagy in these processes, revealing that DPSCs facilitate macrophage M2 polarization and efferocytosis by promoting mitophagy. These findings collectively underscore the immense significance of DPSCs in the treatment of TMJOA. Future research should explore its potential mechanism and optimize its application in clinical practice. Electronic supplementary material Below is the link to the electronic supplementary material. [141]Supplementary Material 1^ (2.2MB, docx) Acknowledgements