Abstract Non-healing skin wounds pose significant clinical challenges, with biologic products like exosomes showing promise for wound healing. Saliva and saliva-derived exosomes, known to accelerate wound repair, yet their extraction is difficult due to the complex environment of oral cavity. In this study, as a viable alternative, we established human minor salivary gland organoids (hMSG-ORG) to produce exosomes (MsOrg-Exo). In vitro, MsOrg-Exo significantly enhanced cell proliferation, migration, and angiogenesis. When incorporated into a GelMA-based controlled-release system, MsOrg-Exo demonstrated controlled release, effectively improving wound closure, collagen synthesis, angiogenesis, and cellular proliferation in a murine skin wound model. Further molecular analyses revealed that MsOrg-Exo promotes proliferation, angiogenesis and the secretion of growth factors in wound sites. Proteomic profiling showed that MsOrg-Exo’s protein composition is similar to human saliva and enriched in proteins essential for wound repair, immune modulation, and coagulation. Additionally, MsOrg-Exo was found to modulate macrophage polarization, inducing a shift towards M1 and M2 phenotypes in vitro within 48 h and predominantly towards the M2 phenotype in vivo after 15 days. In conclusion, our study successfully extracted MsOrg-Exo from hMSG-ORGs, confirmed the effectiveness of the controlled-release system combining MsOrg-Exo with GelMA in promoting skin wound healing, and explored the potential role of macrophages in this action. Supplementary Information The online version contains supplementary material available at 10.1186/s12951-024-02811-y. Keywords: Wound Healing, Exosome, Organoid, Human minor salivary gland, Macrophage Background Chronic non-healing skin wound remains a challenging condition for clinical management. The use of biologic products has been shown to promote wound healing and regeneration [[40]1–[41]5]. Previous studies have investigated the therapeutic potential of exosomes in wound repair, with the majority of studies focusing on stem cell-derived exosomes [[42]6–[43]11]. Saliva contains a variety of bioactive components, including growth factors, antimicrobial peptides and immunoglobulins, and has been demonstrated to accelerate oral and skin wound healing [[44]12–[45]15]. In addition, it has been revealed that saliva-derived exosome enhanced cutaneous wound healing by promoting angiogenesis [[46]16]. Therefore, saliva represents a promising source of exosomes for therapeutic use in skin wound treatment. 3D cultured organoids are composed of a large number of mature, functional cells and can produce abundant exosomes, overcoming the limitation of low yield in traditional 2D cell culture [[47]17]. The therapeutic use of organoid-derived exosome has been studied in various diseases [[48]18–[49]21], while its application in the context of skin wound healing remains largely unexplored. Due to the complex nature of oral environment [[50]22, [51]23], the extraction of pure, sterile saliva-derived exosome is extremely challenging in clinical settings. As an alternative, establishing salivary gland organoids in vitro enables the production of saliva-derived exosomes that can be used for bioengineering techniques. Human minor salivary glands (hMSG) are universally present throughout the oral mucosa and can be easily accessed [[52]24], offering advantages over major salivary glands for tissue harvesting. Harvesting major salivary glands is more technically demanding and associated with complications, including fistula formation and scarring at the incision site. Thus, minor salivary glands are the ideal tissue source for organoid construction. Previous studies have successfully developed a preliminary model of hMSG organoid (hMSG-ORG) [[53]25, [54]26]. In this study, we aimed to (1) incorporate human minor salivary gland organoid-derived exosome (MsOrg-Exo) with a Gelatin Methacryloyl (GelMA) hydrogel to establish a sustained-release system, and to (2) investigate the therapeutic effect of MsOrg-Exo loaded GelMA in skin wound healing. Methods hMSG-ORG culture The minor salivary glands samples used in this study were obtained from patients undergoing lip reduction surgery. Participants ranged in age from 17 to 54 years (N = 13, mean = 35.69). Patients with a history of lip inflammation, lip trauma, or autoimmune diseases were excluded. To isolate cells from the tissues, the samples were dissected into small fragments. The enzymatic digestion process used 0.25% (w/v) collagenase type II (Worthington, US) for 1 h and TrypLE Express (Thermo Fisher, US) for 10 min. The enzymatic solution was then filtered through a 70 μm filter. Isolated cells were embedded in Growth Factor-Reduced Matrigel (Corning, US) in 24-well plates, with the Matrigel solidifying for 20 min at 37 °C. A hMSG-ORG growth medium (HGM) containing the essential growth factors and small molecules was added to the wells (Supplementary Table [55]1), and the plates were incubated at 37 °C and 5% carbon dioxide (CO[2]). Media were changed every 3 days. Organoids were subcultured every 7 days at a ratio of 1:2 to 1:4, and resuspended in cell freezing media (Corning, US) for storage at -80 °C. The brightfield images of organoids was visualized by Incucyte S3 (Sartorius, USA) at fixed intervals. Cell culture To isolate hMSG mesenchymal stem cells (hMSG-MSCs), the hMSG tissues were minced into pieces under 1 mm3 and placed at the bottom of 25 cm2 culture flasks. The culture flask was placed vertically. Then, 3.5 ml of DMEM-High medium (HyClone, US) containing 10% FBS was added to the flask. After 6 h of incubation at 37 °C and 5% CO2, the culture flask was placed horizontally. Media were changed every 3 days. After observing the hMSG-MSCs migrating from the tissue under microscope, cells were passaged using 0.25% trypsin (HyClone, US) at 80% confluence in a 1:3 split ratio. Excessive tissue fragments were filtered out during the first passage. HUVEC, HaCaT, HSF and RAW 264.7 cell lines were all cultured with the aforementioned procedures in DMEM-High medium. Tissue and organoid histology The organoids and tissue were fixed with 4% paraformaldehyde (Servicebio, China), dehydrated overnight at 4 °C in 30% sucrose, and embedded in paraffin. The embedded samples were then sliced into 5-µm-thick sections. Hematoxylin and eosin (H&E), periodic acid-Schiff (PAS), Masson’s trichrome, and Alcian Blue staining were performed under standard protocols. For immunofluorescence staining, primary antibodies included E-Cad (1:200; Proteintech, US), AQP5 (1:500; Santa Cruz, US), CK5 (1:200; ABclonal, China), CK7 (1;200; Proteintech, US), AMY1 (1:800; CST, US). The fluorescent secondary antibodies included Alexa Fluor 594/488-conjugated IgG (1:1000; CST, US). Nuclei were then stained with DAPI (CST, US) for 10 min. Images were acquired using a confocal microscope (Zeiss, Germany). Immunohistochemistry staining used anti-CD31 (1:300; Bioss, China) and anti-KI67 (1:200; Abcam, US) antibody. After deparaffinization, rehydration and antigen retrieval, the sections were incubated overnight at 4 °C with the primary antibodies. The sections were then incubated for 30 min at room temperature with an HRP-conjugated secondary antibody (ZSGB-bio, China). Diaminobenzidine (DAB) reagent (ZSGB-bio, China) was applied for 1 min. Nuclei were counterstained with hematoxylin for 30 s. ImageJ software was used to quantify the integrated optical density. Functional swelling assay Compounds for the stimulant treatment were prepared in following concentration: carbachol 100 µM (Sigma, US), isoproterenol 1 µM (Sigma, US), vasoactive intestinal peptide (VIP) 200 nM (Tocris, UK). An equivalent volume of DMSO was added for control. Brightfield images of the organoids were captured using Incucyte at 5-minute interval over a 2-hour period. Isolation and identification of MsOrg-Exo The supernatant from the hMSG-ORGs culture was collected each time the medium was changed. After collecting a sufficient volume, ultracentrifugation was performed for exosome isolation. Specifically, the supernatant was centrifuged at 1000 g for 10 min, followed by 6000 g for 5 min, then 12,000 g for 30 min, and finally, ultracentrifuged at 100,000 g for 90 min. The MsOrg-Exo was resuspended in PBS and stored at -80 °C. The structure of MsOrg-Exo was observed using transmission electron microscopy (TEM; JEOL, Japan). The protein amount of MsOrg-Exo was further quantified using a BCA protein assay kit (Applygen, China). Nanoparticle tracking analysis (NTA) was performed using NanoSight (Malvern Panalytical, UK) for particle size measurement. MsOrg-Exo and hMSG-ORG of equal protein amount were lysed for western blot analysis following standard protocol. Exosome uptake experiment Before experiment, MsOrg-Exo were stained according to the instructions of the exosome membrane staining kit. Specifically, DiR dye (50:1) was added to an appropriate amount of exosome suspension, and the solution was mixed and incubated at 37 °C for 30 min. The solution was then transferred to a filter tube and centrifuged at 3000 g until the upper layer is free of liquid. The stained exosomes were collected from the filter membrane using PBS. For the exosome uptake experiment, HUVEC cells were treated with the stained exosomes. After 24 h of treatment, the cells were fixed, and immunofluorescence staining was performed following the steps described above. Cell migration assay Experiments were conducted using fully confluent HUVEC, HaCaT and HSF cells grown in a 6-well plate. A straight scratch was made on the cell surface using a 200 µL pipette tip, and the wells were washed three times with PBS. Each well was then incubated with either PBS or 500 ng/µL MsOrg-Exo at 37 °C. Images were captured at 12 and 24 h after treatment, and the area of wound was measured using ImageJ software. Cell proliferation assay HUVEC, HaCaT and HSF cells were seeded in 96-well plates at a density of 3000 cells/well in DMEM-High basic medium, and then treated with 500 ng/µL MsOrg-Exo or an equal volume of PBS. The cell area of each well was sequentially measured every hour over the following 36 h using the Incucyte system. Tube formation assay HUVECs were seeded at a density of 3 × 10⁴ cells per well on Matrigel-coated 96-well plates, and then treated with 0, 100, 300, or 500 µg/mL MsOrg-Exo in DMEM-High medium for 8 h. Brightfield images were captured at 4 and 8 h. The number of branches were counted using ImageJ software. Preparation and characterization of GelMA Firstly, 0.25% (w/v) solution of lithium phenyl-2,4,6-trimethylbenzoylphosphinate (LAP; EFL, China) was prepared by dissolving 25 mg of LAP in 10 mL PBS. The LAP solution was mixed with dried GelMA (EFL, China) at various concentrations and heated to 70 °C for complete dissolution. The GelMA solutions were then immediately filtered through a preheated 0.22 μm sterile injection filter. Next, GelMA hydrogel was formed by crosslinking with 405 nm ultraviolet light for 10 s. MsOrg-Exo was added to the GelMA solution before crosslinking. Scanning electron microscopy (SEM; JEOL, Japan) was used to examine the surface morphology of GelMA. After being frozen and lyophilized using a freeze dryer, the GelMA samples were taped to a stub and sputter-coated with platinum before being introduced into the SEM chamber for imaging. To detect the methacrylamide signals, crosslinked GelMA was cut into small pieces and dissolved in deuterated reagent. Proton nuclear magnetic resonance (H-NMR) was performed using spectrometer (Bruker, Germany) to test for the H spectrum. The storage modules and loss modules of GelMA before and after crosslinking were measured using rheometer (TA, US). GelMA solution pre-mixed with LAP was placed into the instrument. 60 s into the testing procedure, the samples were irradiated with 405 nm UV light for 20 s, after which the test went on for an additional 60 s. The compression test use electronic universal testing machine (MTS, China). The appropriate type of compression fixture was selected according to the sample size requirements. The initial force was set to 0.01 N, and the compression rate was set to 0.2 mm/min. The test was started, and the equipment recorded the variation of force with displacement during the compression test. For the degradation experiment, cross-linked GelMA samples were immersed in PBS at 37 °C. Every 2 days, three samples were collected and dried under vacuum. The weight of the dried samples was measured, and the degradation ratios were calculated to construct the degradation curve. Mouse skin wound model Mice (C57BL/6J, male, 7 weeks) were randomly divided into 4 groups (N = 5): (1) Blank; (2) 80 µL 10% GelMA; (3) 80 µL 10% GelMA with 500 ng/µL MsOrg-Exo; (4) 80 µL PBS with 500 ng/µL MsOrg-Exo. After anesthesia, two full-thickness skin wounds with a diameter of 8 mm were created on the back. The prepared silicone ring was sutured to the skin around the wound so that the wound edges aligned with the inner circle of the silicone ring, according to Wang et al [[56]27]. For groups 2) and 3), the GelMA droplets were crosslinked by irradiating with 405 nm UV light for 20 s. For group 4), the MsOrg-Exo solution was injected into the dermis from the inner side of the wound. The wounds were covered with Tegaderm dressing and wrapped with an elastic bandage. Macroscopic assessment of the wounds was performed on days 0, 5, 10, and 15. The wound edges were manually segmented using ImageJ. On day 15, all mice were anesthetized to harvest wound tissue for further analysis, and then euthanized using cervical dislocation maneuver. Exosome controlled-release test In vitro, 25 µL GelMA hydrogel containing 500 ng/µL MsOrg-Exo were immersed in PBS in a 96-well plate, denoted as GelMA + MsOrg-Exo system. The supernatant was collected every day for 15 days and the protein content was measured using BCA Protein Assay Kit. In vivo, 80 µL of GelMA containing DiR-labeled MsOrg-Exo was applied to the wound on the left side of the mouse and immediately crosslinked using 405 nm UV light for 20 s. For right-sided wounds, 80 µL of MsOrg-Exo solution was injected into the dermis from four directions around the wound. Mice were anesthetized and subjected to IVIS imaging on day 0 and day 7. RNA extraction and real-time qPCR Total RNA was extracted from tissue using TRIzol reagent (Invitrogen, US). The RNA was then reverse transcribed into cDNA using the FastKing RT Kit (TIANGEN, China) following the manufacturer’s instructions. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was carried out using QuantiNova SYBR Green PCR Master Mix (QIAGEN, Germany) on a Bio-Rad CFX system. Primer sequences are listed in Supplementary Table [57]2. Proteomics analysis Saliva from healthy individuals (N = 3) and MsOrg-Exo samples (N = 3) were collected, and protein quantification was performed using the BCA method, followed by standard processing [[58]28]. Briefly, samples were mixed with urea buffer in Vivacon tubes and centrifuged. TCEP, IAA, and trypsin were then added sequentially, with incubation at the corresponding temperatures. After another centrifugation, the peptides were collected and reconstituted with ammonium solution. The samples were then subjected to elution, chromatographic separation, and mass spectrometry analysis using the proteomics analysis system (Thermo Scientific, US). Proteomic data were analyzed using MaxQuant software and compared with the UniProt database. The top 150 proteins with the highest abundance in MsOrg-Exo were selected for GO and KEGG pathway enrichment analysis. Flow cytometry analysis The treated RAW 246.7 cells were collected from a 6-well plate using a cell scraper. After centrifugation, the cells were resuspended in TruStain FcX buffer (BioLegend, US) and incubated for 10 min. Following another centrifugation, the cell pellet was resuspended in a mixture of cell surface protein fluorescent antibodies diluted 1:100 and incubated at room temperature in the dark for 20 min. The cells were then washed with cold PBS, resuspended in fixation buffer (Invitrogen, US), and incubated at room temperature in the dark for 20 min. The pellet was centrifuged and washed three times with permeabilization buffer (Invitrogen, US). Subsequently, the intracellular protein fluorescent antibodies were diluted 1:50, and the cell pellet was resuspended in this mixture and incubated at room temperature in the dark for 60 min. Finally, the cells were washed three times with Permeabilization buffer, resuspended in an appropriate amount of cold PBS, and analyzed by flow cytometry. The primary antibodies used in this study were as follows: anti-F4/80-AF488, anti-CD80-BV421, anti-CD86-PE, anti-CD206-APC (BioLegend, US). Opal multiplexed immunofluorescence PerkinElmer Opal 7-Color Manual IHC kit (Perkin-Elmer, US) was used to multiplexed immunofluorescence staining. According to the protocol, after deparaffinization, rehydration and antigen retrieval, the sections underwent the following five steps for each marker consecutively: (1) blockage with antibody diluent; (2) incubation with primary antibody for 1 h; (3) detection using Opal Polymer HRP Ms + Rb secondary antibody; (4) visualization using Opal TSA plus agent; (5) microwave treatment in citrate buffer. The primary antibodies used for each protein were as follows: anti-CD86 (1:500; CST, US), anti-CD206 (1:500; CST, US) and anti-F4/80 (1:500; CST, US). Statistical analysis The results are presented as mean ± standard error. The data were analyzed using GraphPad Prism (Version 9.0.0; US). Unpaired two-tailed student’s t-test was used for analysis between two groups. One-way ANOVA was used for analysis among multiple groups. P-values < 0.05 were considered significant (*P < 0.05, **P < 0.01, ***P < 0.001). Results Establishment of hMSG-ORG and exosome isolation Single-cell growth was tracked continuously. Formation of small, spherical, acinar-like organoids with a diameter of approximately 100 μm was observed on the third day. Subsequently, the acini gradually increased in size, reaching a diameter of 1–2 mm after two weeks of culture (Fig. [59]1a). Fig. 1. [60]Fig. 1 [61]Open in a new tab Establishment of hMSG-ORG and exosome isolation. A. Bright-field images of primary culture of hMSG-ORG from day 0 to day 13 (scale bar = 500 μm) B. H&E / PAS / Alcian Blue staining of hMSG and hMSG-ORG showing salivary mucins in hMSG-ORG (scale bar = 100 μm) C. Immunofluorescence images of salivary gland markers in hMSG and hMSG-ORG (nuclei stained with DAPI; scale bar = 100 μm) D. Bright-field images showing acinar swelling in hMSG-ORG after stimulation with Carbachol / Isoprenaline / VIP (scale bar = 1 mm) E. TEM images of MsOrg-Exo (scale bar = 100 nm) F. Mean particle diameter and peak value of MsOrg-Exo measured by NTA G. Western blot analysis of exosomal markers CD81, TSG101, and the endoplasmic reticulum marker Calnexin in MsOrg-Exo and hMSG-ORG H. Exosomal protein content obtained from equal amounts of hMSG-MSC supernatant or hMSG-ORG supernatant measured by BCA assay (N = 3) Histological staining of hMSG-ORGs showed structural similarity with normal hMSG tissue. H&E staining of hMSG-ORGs revealed abundant, variably sized luminal structures. Salivary mucins were visible within larger lumens, as stained purple-red on PAS staining and blue on Alcian Blue staining (Fig. [62]1b). For immunofluorescence staining, the acinar markers AMY1 and AQP5 were both expressed in hMSG-ORGs, with AMY1 displaying a characteristic ring-like structure around the basal surface of the acini, similar to hMSG tissue. The epithelial cell marker E-Cadherin was expressed around the periphery of acini and lumens in both hMSG and hMSG-ORGs. Additionally, the basal cell marker CK5 and luminal cell marker CK7 were expressed. Moreover, the luminal cells were surrounded by basal cells in hMSG-ORGs, which is a hallmark histologic finding in hMSG tissue (Fig. [63]1c). These findings indicated that the hMSG-ORGs cultured in vitro largely mimicked salivary gland characteristics. To verify whether hMSG-ORGs can mimic salivary gland function in vitro, we performed functional swelling assays using a muscarinic receptor agonist carbachol, a β-receptor agonist isoproterenol, and a non-adrenergic non-cholinergic peptide neurotransmitter VIP. Real-time observation showed significant swelling of acinar-like structures in hMSG-ORGs within 2 h of drug administration, whereas no significant changes were observed in hMSG-ORGs treated with DMSO, indicating that hMSG-ORGs can respond to different neurotransmitter stimuli in vitro (Fig. [64]1d). After the establishment of hMSG-ORGs, the culture supernatant was collected for exosome extraction (MsOrg-Exo). TEM scanning of MsOrg-Exo exhibited a cup-shaped morphology with a diameter of approximately 100 nm (Fig. [65]1e). NTA showed that the average particle size of MsOrg-Exo was 133.9 ± 67.0 nm (Fig. [66]1f). Exosomal surface proteins CD81 and TSG101 were shown to be expressed at higher levels in MsOrg-Exo (Fig. [67]1 g). Furthermore, compared to exosomes derived from hMSG-MSCs, a significantly increased number of exosomes were extracted from an equal volume of supernatant collected from the hMSG-ORG culture system (Fig. [68]1 h). These results indicated that MsOrg-Exo exhibited the fundamental characteristics of exosomes, and had advantages over exosomes extracted from the 2D culture system. In Vitro effects of MsOrg-Exo on cell migration, proliferation, and angiogenesis We treated HUVEC cells with DiR-labeled MsOrg-Exo, resulting in abundant red fluorescent particle accumulation around the cell nuclei, which indicated that MsOrg-Exo could be internalized (Fig. [69]2a). Subsequently, we treated HUVEC cells, HaCaT cells, and HSF cells with MsOrg-Exo. Scratch assay results showed that 12- and 24-hours post-treatment, the remaining scratch area in HUVEC, HaCaT, and HSF cells was significantly smaller compared to the control group (Fig. [70]2b-c). The area of cell growth in HUVEC, HaCaT, and HSF cells treated with MsOrg-Exo was larger compared to the control group, at any given time point within 36 h post-treatment (Fig. [71]2d). Based on these results, we concluded that MsOrg-Exo promoted the migration and proliferation of skin cells in vitro. Fig. 2. [72]Fig. 2 [73]Open in a new tab In vitro effects of MsOrg-Exo on cell migration, proliferation, and angiogenesis. A. Immunofluorescence images showing the internalization of MsOrg-Exo by HUVEC cells (exosomes labeled with DiR, nuclei stained with DAPI) B. Scratch assay conducted on HUVEC, HaCaT, and HSF cells treated with MsOrg-Exo, with bright-field images captured at 0 h and 24 h post-treatment (scale bar = 100 μm) C. Statistical analysis of the remaining scratch area at 0 h, 12 h, and 24 h (N = 5) D. Proliferation assay of HUVEC, HaCaT, and HSF cells treated with MsOrg-Exo, with cell growth area continuously tracked by Incucyte over 36 h (N = 3) E. Bright-field images of HUVEC cells in the tube formation assay at 4 h and 8 h with MsOrg-Exo concentration of 500 µg/ml (scale bar = 100 μm) F. Statistical analysis of the number of branches formed at MsOrg-Exo concentrations of 0, 100, 300, and 500 µg/ml (N = 5) In vitro tube formation assays demonstrated that HUVEC cells began to form branching structures approximately 4 h post MsOrg-Exo treatment, and more complete tubular structures after 8 h. In contrast, HUVEC cells treated with PBS during the same period exhibited almost no significant branching or tubular structures (Fig. [74]2e). Statistical analysis of tube formation under different concentrations of MsOrg-Exo, using the number of branch structures formed after 8 h as the observation index, showed significant differences between the control group (0 µg/ml) and the 100, 300, and 500 µg/ml groups (Fig. [75]2f). These findings suggested that MsOrg-Exo promoted angiogenesis in vitro. Development of an exosome controlled-release system using GelMA Before constructing the controlled release system, the mechanical properties of GelMA were first characterized. Upon exposure to 405 nm ultraviolet light for 20 s, GelMA transitioned from a highly flowable liquid to a solid gel (Fig. [76]3a). SEM images revealed that as the concentration of GelMA increased, its micropores slightly decreased in size (Fig. [77]3b). H-NMR analysis of the cured GelMA showed two characteristic peaks around 5.5 ppm, indicating the presence of two methacrylate vinyl groups (Fig. [78]3c). Rheological results demonstrated that cross-linked GelMA is an elastic solid with a storage modulus significantly higher than the loss modulus, consistent with the general properties of hydrogels (Fig. [79]3d). Compression test showed that GelMA is easily deformable and resistant to fracturing under stress (Fig. [80]3e). Additional degradation experiment proved that after 14 days of immersion, GelMA degraded by less than 30% (Fig. [81]3f). The mechanical properties of GelMA made it a suitable choice as a long-term wound dressing material in animal experiments. Fig. 3. [82]Fig. 3 [83]Open in a new tab Development of an exosome controlled-release system using GelMA. A. Appearance of 10% GelMA before and after crosslinking with 405 nm UV light for 20 s (left, before crosslinking; right, after crosslinking) B. SEM images showing pore sizes of 5% / 10% / 15% GelMA C. NMR spectrum of GelMA, with arrows indicating the characteristic peaks of methacrylate vinyl groups D. Changes in storage and loss module of 10% GelMA before and after photo-crosslinking E. Stress-strain curve of 10% GelMA F. Degradation ratio of 10% GelMA in 14 days G. In vitro release profiles of MsOrg-Exo loaded in GelMA at different concentrations (N = 3) H. 7-day release experiment in mice. The left wound was treated with 10% GelMA loaded with MsOrg-Exo, and the right wound was treated with an injection of MsOrg-Exo. MsOrg-Exo was pre-labeled with DiR, and images were captured using IVIS For in vitro exosome controlled-release experiments, MsOrg-Exo in the 5% GelMA system was fully released in 12 days, while in the 15% systems, cumulative release rates were around 70% by day 14. Higher concentrations of GelMA prolonged the release duration of exosomes (Fig. [84]3e). We subsequently applied the GelMA + MsOrg-Exo system to mice for in vivo controlled-release experiments. We tracked the sustained release of exosomes using DiR-labeled MsOrg-Exo. After 7 days, strong fluorescence was still present at the wound site with GelMA sustained release, whereas MsOrg-Exo injected subcutaneously on the other side had been completely absorbed (Fig. [85]3f). The GelMA + MsOrg-Exo system represented a controlled-release system for exosomes both in vitro and in vivo. In vivo promotion of wound healing by MsOrg-Exo The normal wound healing in mice primarily relies on skin contraction, while human wound healing is mainly achieved through cell proliferation and active migration. To better simulate human wound conditions, we used silicone rings to fix the wound, thereby minimizing the skin contraction in mice. On day 5 of administration, there were no noticeable differences in healing among the four groups upon visual inspection. By day 10, the GelMA + MsOrg-Exo controlled-release system group (hereafter referred to as the “controlled-release group”) showed significant granulation tissue and fibrous tissue proliferation, with the wound edges migrating towards the center. By day 15, the blank group’s wound edges showed no substantial proliferation or migration, only slight skin contraction; the GelMA control group (hereafter referred to as the “GelMA group”) showed a small amount of white fibrous tissue at the wound edges; the controlled-release group wounds were nearly completely healed with almost no subcutaneous tissue visible and robust hair growth around the wound; the healing condition of the group with injected MsOrg-Exo (PBS + MsOrg-Exo group, hereafter referred to as the “injection group”) was intermediate between the above two groups (Fig. [86]4a). Quantitative analysis of wound area showed that on day 10 and day 15, the healing rate of the sustained release group was significantly higher than that of the other three groups (Fig. [87]4b-c). Fig. 4. [88]Fig. 4 [89]Open in a new tab In vivo promotion of wound healing by MsOrg-Exo. A. Photographs and simulated images of mouse wound healing at days 0, 5, 10, and 15 (silicone ring inner diameter = 9 mm) B-C. Statistical analysis of mouse wound healing rates at days 0, 5, 10, and 15 (N = 5) On day 15, skin around the mouse wounds was sampled for analysis. H&E staining showed a larger area of granulation tissue with new blood vessels in the controlled-release and injection groups, whereas the blank group’s wound tissue was thinnest (Fig. [90]5a). Masson trichrome staining revealed richer blue-stained collagen fibers in the wound areas of the controlled-release and injection groups, with the most pronounced effect in the controlled-release group (Fig. [91]5b). The expression level of related genes in wound skin tissue was quantified using qRT-PCR. Compared with the blank group and the GelMA group, the controlled-release group showed significantly higher expression levels of VEGFα, HGF, bFGF, TGFβ, CK10, and CK19. Compared with the injection group, the controlled-release group also showed higher expression levels of these genes, though without statistical significance (Fig. [92]5c). Immunohistochemical staining showed significantly more and denser CD31-positive cells in the controlled-release group, indicating a notable promotion of angiogenesis (Fig. [93]5d). Additionally, the controlled-release group had more KI67-positive nuclei, suggesting a higher number of actively proliferating cells; the injection group also showed increased cell proliferation, though without statistical significance (Fig. [94]5e). Fig. 5. [95]Fig. 5 [96]Open in a new tab Proteomic analysis of MsOrg-Exo. A. H&E staining of mouse wound at postoperative day 15, with blue lines indicating wound width and arrows pointing to areas of epidermal discontinuity (scale bar = 500 μm) B. Masson staining of mouse wound at postoperative day 15, with blue-stained areas indicating collagen fibers (scale bar = 500 μm) C. qPCR analysis of expression levels of growth factors VEGFα, HGF, bFGF, TGFβ, epidermal differentiation marker CK10, and hair follicle stem cell marker CK19 in wound tissue at postoperative day 15 (N = 5) D-E. Immunohistochemical staining and semi-quantitative analysis of protein expression levels of CD31 and KI67 in wound tissue at postoperative day 15 (scale bar = 100 μm) These results suggested that the GelMA + MsOrg-Exo controlled-release system promoted wound healing in mice by regulating growth factor expression, angiogenesis, and cell proliferation. Proteomic analysis of MsOrg-Exo Next, we performed proteomic analysis of MsOrg-Exo and compared it with the human saliva proteome. The results showed a high overlap between the protein species in MsOrg-Exo and human saliva proteins (Fig. [97]6a), suggesting that MsOrg-Exo may exert functions similar to human saliva to a considerable extent. Fig. 6. [98]Fig. 6 [99]Open in a new tab Proteomic analysis of MsOrg-Exo. A. Venn diagram of proteins in MsOrg-Exo and human saliva; 790 proteins identified in MsOrg-Exo, 954 proteins identified in human saliva samples, with 740 proteins common to both B. Differential clustering heatmap of proteins in MsOrg-Exo and human saliva (N = 3) C-D. GO and KEGG pathway enrichment analysis of the top 150 proteins expressed in MsOrg-Exo In differential protein clustering analysis, we identified several upregulated proteins in MsOrg-Exo that are related to wound healing, including VCL, which mediates the epidermal growth factor pathway; COL1A and COL2A, important components of skin fibers; ITGB1, which regulates keratinocyte migration; SPARC and THBS1, which promote fibroblast proliferation and migration; MAP2K2, related to inflammation induction; LAMB2, which promotes endothelial cell adhesion, migration, and angiogenesis; and RHOA, which promotes epidermal stem cell proliferation and migration (Fig. [100]6b). These upregulated proteins potentially mediated the healing process in skin wounds treated with MsOrg-Exo. In the Biological Process (BP) section of GO analysis, the pathway with the most enriched proteins was “wound healing” (16.55%). The subsequent categories were “negative regulation of proteolysis” (12.95%), “epidermis development” (12.95%), and “humoral immune response” (12.23%), all of which are involved in different aspects of wound healing regulation (Fig. [101]6c). In KEGG analysis, 16 pathways were statistically significant, with the most enriched pathway being “focal adhesion” (8.67%), which could assist in the adhesion connection of organoids with Matrigel during growth. The next enriched pathways were “complement and coagulation cascades” (8%) and the “PI3K-Akt pathway” (8%); the former could participate in the inflammatory immune response and coagulation during wound healing, while the latter could regulate skin cell proliferation and apoptosis, promoting angiogenesis by participating in key processes such as protein synthesis, metabolism, and the cell cycle (Fig. [102]6d). Macrophages mediate the regulation of wound healing by MsOrg-Exo In the proteomic analysis, we observed that the proteins in MsOrg-Exo are highly related to immune responses. Therefore, we attempted to treat RAW 264.7 cells with MsOrg-Exo and conducted subsequent observations and analyses. Bright field images showed that after 48 h of culture, approximately 5% of RAW cells in the control group underwent natural differentiation. In contrast, about 60% of RAW cells treated with MsOrg-Exo exhibited varying degrees of polarization, becoming polygonal or developing pseudopodia (Fig. [103]7a). Further flow cytometry analysis revealed that after 24 and 48 h of MsOrg-Exo treatment, the expression of M1 and M2 markers in RAW cells was significantly increased. CD80 (M1 marker) showed the highest proportion, while CD206 (M2 marker) exhibited the greatest fold increase, with the degree of increase being positively correlated with the treatment duration (Fig. [104]7b). These results demonstrate that MsOrg-Exo can promote macrophage polarization in vitro in a short period. Fig. 7. [105]Fig. 7 [106]Open in a new tab Macrophages mediate the regulation of wound healing by MsOrg-Exo. A. Bright-field images of RAW 264.7 cells treated with MsOrg-Exo at 24 h and 48 h, with arrows indicating polygonal polarized RAW cells (scale bar = 100 μm) B. Flow cytometry analysis of macrophage polarization markers CD80, CD86, and CD206 in RAW cells treated with MsOrg-Exo (n = 3) C-D. qPCR analysis of M1 markers CD80, TNFα, IL1β, and M2 markers IGF1, IL10, IL13 expression levels in wound tissue of mice at postoperative day 15 (N = 5) E-F. Opal staining (nuclei stained with DAPI) and semi-quantitative analysis (N = 5) of macrophage-related proteins CD86, CD206, and F4/80 in wound tissue of mice at postoperative day 15 Next, we performed qRT-PCR analysis on the wound tissue of mice on the 15th day to verify macrophage polarization in vivo. Among M1-related genes, the level of CD80 was significantly downregulated in the controlled-release group, while the expression level of the pro-inflammatory factor TNFα showed no significant difference between the groups. The expression level of IL1b, which promotes M1 polarization, was relatively lower in the controlled-release group but without statistical significance (Fig. [107]7c). Among M2-related genes, the expression level of the M2 marker IGF1 and M2 mediator IL10 were significantly upregulated in the controlled-release group. The expression level of IL13, which stimulates M2 polarization, was upregulated in both the controlled-release and injection groups but without statistical significance (Fig. [108]7d). We then performed Opal staining to detect the expression of CD86 and CD206 in the same tissue section. The results showed that on the 15th day, there was no significant difference in the expression level of CD86 between the groups (Fig. [109]7e), while the expression level of CD206 in the controlled-release group was significantly higher than in the other three groups (Fig. [110]7f). These results indicated that 15 days post-administration, the recruitment of M2 macrophages in the controlled-release group could be detected within the tissue. Therefore, we believe that MsOrg-Exo may assist macrophages in transitioning to both M1 and M2 types in the early stages of wound healing (within 48 h). In the later stages of wound healing, it assists macrophages in transitioning more towards the M2 type, which is in accordance with the commonly accepted role of macrophages throughout the wound healing process. Discussion In this study, we developed hMSG-ORGs, which can be cultured and passaged long-term in vitro. We verified the presence of mucin and the localized expression of salivary gland marker proteins such as AMY1, AQP5, CK5, and CK7. The MsOrg-Exo isolated from the culture supernatant through ultracentrifugation are the first exosomes successfully extracted from human salivary gland organoids. Their morphology, size, and surface proteins were verified by various methods, meeting the general standards of exosomes. In vitro experiments showed that MsOrg-Exo could be taken up by cells and promoted the migration and proliferation of HUVEC, HaCaT, and HSF cells while enhancing the tube formation capability of HUVEC cells. In vivo, MsOrg-Exo incorporated into GelMA facilitated wound healing on the backs of mice within 15 days. Proteomic analysis provided deeper insights into the protein composition of MsOrg-Exo, which overlaps significantly with that of human saliva, suggesting MsOrg-Exo as a potential alternative to saliva. Furthermore, the proteins enriched in MsOrg-Exo positively influenced wound healing through various pathways, including immune-related ones. Further analysis indicated that MsOrg-Exo might promote wound healing by inducing macrophage polarization during different stages of the healing process. The construction of salivary gland organoids began in 2008 when Clevers team used salivary gland stem cells for 3D culture in vitro and successfully performed orthotopic transplantation in mice [[111]29]. In 2015, research group pioneered the isolation and culture of adult minor salivary gland epithelial stem/progenitor cells (hMSG-EpiPCs) and hMSG-MSCs, and then mixed them for 3D culture to construct functional hMSG-ORG [[112]26]. This approach is advantageous as it provides a clear cell composition for further research. However, when replicating this method, we found that hMSG-EpiPCs quickly aged during planar passage in vitro, with most cells failing to persist beyond five passages. Thus, we need to explore alternative methods. In 2016, the Coppes team reported separating salivary gland cells through enzymatic digestion and directly seeding the digested cells into hydrogels for organoid culture [[113]30]. Our study used a similar approach. We found that cells from digested human minor salivary glands could develop into acinar-like structures in Matrigel within 3 days, and they could be passaged after about 1 week of culture. In 2022, Yeo-Jun Yoon successfully established organoids from the three major human salivary glands that could be stably cultured and passaged for four months [[114]31]. In our research, the longest cultured hMSG-ORGs retained their acinar morphology and could be passaged for about two months, potentially due to the structural and functional differences between major and minor salivary glands. However, the digestion method allows all cell types to be incorporated into the hMSG-ORGs. Our study confirmed the differential localization of acinar and duct markers in hMSG-ORGs, but further analysis is needed to identify specific subtypes or other potential cell types. As mentioned earlier, salivary exosomes have been found to promote wound healing [[115]16], and they may also play a role in treating autoimmune diseases and controlling the progression of oral cancer [[116]32]. Furthermore, existing research on salivary gland-derived exosomes all involves cultivating salivary gland stem cells, including mesenchymal stem cells from the labial glands [[117]33] or parotid glands [[118]34], to extract exosomes for the treatment of salivary gland injuries or functional disorders. Previous studies have also attempted to culture salivary gland-like organoids using dental pulp stem cells to extract exosomes [[119]35, [120]36], but no further downstream research was conducted. Our study is the first to extract exosomes from genuine salivary gland organoids and demonstrate their significant role in wound healing. Wound healing is another theme of this study. Building appropriate animal models is crucial in wound healing research. Rats and mice, particularly mice, are widely used due to their easy maintenance, straightforward surgical procedures, and abundant research reagents [[121]37]. However, the natural wound healing pattern in rodents differs from humans. Mouse subcutaneous tissue is primarily composed of the panniculus carnosus muscle, which causes rapid wound contraction after injury [[122]38]. In contrast, human wound healing relies on re-epithelialization and granulation tissue formation without muscle involvement. Thus, specialized surgical procedures may be needed to reduce skin contraction when constructing skin wound models in mice. In some previous studies, researchers ignored this effect and directly used the difference between the experimental and control groups to demonstrate drug efficacy without additional treatment on mouse dorsal skin [[123]39, [124]40]. However, this method has drawbacks: the natural healing time for mouse dorsal skin is short, with significant wound reduction observed three days post-operation and complete healing within seven days, making it challenging to observe changes promptly. In this study, we primarily referred to the mouse wound model protocol proposed by Wang et al. in 2013 [[125]27], using silicone rings to restrict skin contraction. This method is widely used in studies investigating wound healing in mice [[126]41, [127]42]. Different phenotypes of macrophages are recruited at different stages of wound healing. After tissue injury, once monocytes enter the wound, bacterial LPS and pro-inflammatory cytokines from tissue cells trigger their differentiation into M1-like macrophages in the early stage of wound healing, clearing pathogens and cellular debris [[128]43, [129]44]. In the later stage of healing, IL-4 and IL-13 from tissue cells stimulate macrophages to switch to the M2 type, releasing factors like VEGF and TGF-β, which promote cell growth, collagen production, and angiogenesis [[130]45, [131]46]. In chronic wounds, pro-inflammatory M1 macrophages persist and fail to transition to the anti-inflammatory M2 phenotype, leading to delayed healing [[132]47]. In our study, MsOrg-Exo promoted the short-term polarization of RAW cells to M1 in vitro and helped accelerate the polarization of macrophages to M2 in vivo for faster wound healing in mice. However, due to limited mice availability, we conducted only a single round of sampling. At the time of sampling, all four wound groups had entered the proliferation phase. Thus, we could not verify the effect of MsOrg-Exo in vivo during the early stages of wound healing (inflammation stage), particularly on macrophages. Future studies should include more groups for comparison of histological and molecular biology changes at different healing stages. The human body contains a large number of minor salivary glands located superficially [[133]24], making them more suitable for research and clinical studies than the three major salivary glands. Our research utilized hMSG to construct engineered hMSG-ORGs and demonstrated the pro-wound healing effects of MsOrg-Exo both in vitro and in vivo. This breakthrough in the application of salivary gland organoids provides a strong theoretical basis for future clinical research on MsOrg-Exo in regenerative medicine. Further research is needed to explore the potential clinical use of autologous or allogeneic MsOrg-Exo for treatment or to combine MsOrg-Exo with new biocompatible materials for dressings, or to use MsOrg-Exo as an alternative to stem cell therapy for salivary gland injury. Conclusions Our study established hMSG-ORG to produce MsOrg-Exo, which significantly enhanced cell proliferation, migration, and angiogenesis in vitro. When combined with GelMA, control-released MsOrg-Exo improved wound healing in vivo by promoting collagen synthesis, angiogenesis and growth factor secretion. Proteomic analysis revealed that MsOrg-Exo is enriched in proteins essential for wound repair. Additionally, MsOrg-Exo modulated macrophage polarization, shifting towards M2 phenotype in long-term treatment. Electronic supplementary material Below is the link to the electronic supplementary material. [134]Supplementary Material 1^ (94.2KB, pdf) Acknowledgements