Abstract Osteochondral defects pose significant challenges in joint health, leading to pain, decreased mobility, and substantial healthcare costs due to surgical interventions. This study aimed to develop and evaluate Si-A/PUE@HA composite hydrogels as scaffolding materials for cartilage tissue engineering, addressing the limitations of current treatment options. Utilizing a multi-step synthesis method that incorporates both chemical and physical crosslinking techniques, we assessed the hydrogels’ cytocompatibility, chondrogenic differentiation potential, immunomodulatory properties, and in vivo cartilage regeneration capabilities. In vitro results demonstrated high cell viability and proliferation rates of bone marrow mesenchymal stem cells (BMSCs) cultured in Si-A/PUE@HA hydrogels, with significant expression of chondrogenic markers such as Acan, Sox9, and Col2a1. Notably, the hydrogels exhibited a favorable immunomodulatory effect, promoting an anti-inflammatory M2 macrophage phenotype, which is crucial for tissue regeneration. In vivo studies confirmed substantial new tissue formation and integration with surrounding cartilage, as evidenced by micro-CT analysis, alongside excellent biocompatibility with no significant adverse effects observed in major organs over a 12-week period. Single-cell RNA sequencing analysis revealed a favorable immune microenvironment and enhanced chondrogenesis linked to hydrogel treatment. In conclusion, Si-A/PUE@HA hydrogels represent a promising biomaterial with potential applications in cartilage repair and regenerative medicine, warranting further investigation in larger clinical trials to validate their efficacy and safety for future therapeutic use. Graphical abstract [56]graphic file with name 12951_2025_3648_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12951-025-03648-9. Keywords: Cartilage regeneration, Tissue engineering, Chondrogenic differentiation, Stem cell therapy, Macrophage polarization, Single-Cell RNA sequencing Introduction Osteochondral defects represent a significant clinical challenge due to their complex nature, involving damage to both articular cartilage and underlying subchondral bone [[57]1–[58]3]. These defects can arise from various etiologies, including trauma, osteoarthritis, and degenerative conditions [[59]4–[60]6]. The limited regenerative capacity of cartilage, compounded by its avascular nature, often leads to chronic pain, decreased mobility, and an overall decline in quality of life for affected patients [[61]7–[62]9]. The economic burden associated with these conditions is substantial, as they frequently necessitate surgical interventions and extensive rehabilitation efforts [[63]10]. Thus, there is an urgent demand for innovative therapeutic strategies aimed at enhancing cartilage repair and regeneration. Current treatment modalities for osteochondral defects include microfracture, autologous chondrocyte implantation, and osteochondral grafting [[64]11–[65]13]. Despite their widespread use, these approaches often yield variable success rates and may result in the formation of fibrocartilage instead of the desired hyaline cartilage [[66]14–[67]16]. This limitation underscores the need for further research to explore alternative methods that can effectively promote cartilage regeneration and restore joint function. Advances in biomaterials and tissue engineering have opened new avenues for the development of scaffolds that can mimic the native extracellular matrix, providing a conducive environment for cellular proliferation and differentiation. Recent studies have highlighted the potential of hydrogels as effective scaffolding materials in cartilage tissue engineering [[68]17]. Their unique properties, such as high water content, biocompatibility, and the ability to facilitate cell migration, make hydrogels an attractive choice for cartilage repair applications [[69]18–[70]20]. For example, hydrogels can be synthesized to possess specific mechanical and biochemical cues that promote chondrogenic differentiation of mesenchymal stem cells (MSCs) [[71]21, [72]22]. Moreover, the incorporation of bioactive agents within hydrogel matrices can enhance their regenerative potential by providing sustained release of growth factors that support cartilage formation [[73]23, [74]24]. In this context, the development of composite hydrogels represents a promising strategy to optimize the mechanical properties and biological performance of scaffolds for cartilage regeneration. Therefore, we have developed a hydrogel named Si-A/PUE@HA. This composite hydrogel is designed through the combination of silane, arginine, puerarin, and hyaluronic acid (HA), utilizing both chemical and physical crosslinking methods to significantly enhance the material’s mechanical strength and bioactivity. Specifically, silane provides excellent structural stability and adjustable mechanical properties [[75]25]; arginine, a natural amino acid, promotes cell adhesion and proliferation while improving biocompatibility [[76]26]; puerarin, a natural isoflavone compound, offers strong antioxidant and anti-inflammatory effects, contributing to the regulation of the cellular microenvironment [[77]27, [78]28]; and hyaluronic acid, a natural polysaccharide, effectively promotes cell migration and cartilage tissue repair [[79]29]. The synergistic effect of these bioactive components allows the composite hydrogel to exhibit outstanding advantages in both mechanical performance and biological functionality. Such innovative materials are crucial as they have the potential to overcome the limitations of traditional scaffolds, which often lack sufficient support for cell adhesion, proliferation, and tissue repair [[80]30]. The primary objective of this research is to evaluate the efficacy of Si-A/PUE@HA hydrogels in promoting chondrogenic differentiation of stem cells and enhancing cartilage repair in vivo. This multi-faceted approach will involve assessing cellular responses in vitro, as well as evaluating the functional outcomes of cartilage regeneration in appropriate animal models. By integrating advanced hydrogel technology with stem cell therapy, this study aims to contribute valuable insights into the development of effective treatment strategies for osteochondral defects. In summary, the increasing prevalence of osteochondral defects necessitates the exploration of novel therapeutic strategies that can facilitate cartilage regeneration. The use of advanced composite hydrogels offers a promising avenue for improving treatment outcomes, and the findings of this research may pave the way for future clinical applications in regenerative medicine. Continued investigation in this field is essential to address the existing challenges and enhance the quality of life for patients suffering from cartilage-related injuries. Martials and methods Preparation and characterization of Si-A/PUE@HA composite hydrogels The Si-A/PUE@HA composite hydrogels were synthesized through a precisely controlled multi-step process that integrated both chemical and physical crosslinking to create a structurally robust and bioactive material. For comparison, a negative control hydrogel (NC@HA), composed solely of hyaluronic acid (HA) without silane, arginine, or puerarin, was prepared using the same physical processing steps but without the addition of bioactive components. Initially, 1.0 g of hyaluronic acid (HA, Sigma-Aldrich, USA) was fully dissolved in 100 mL of deionized water under continuous magnetic stirring at room temperature (~ 25 °C) to form a clear and homogeneous solution. Subsequently, 2.0 mL of polyethoxysilane (silanol, Aladdin, China) was gradually added dropwise into the HA solution under vigorous stirring for 2 hours, facilitating pre-crosslinking through silanol condensation reactions. To impart bioactivity, 0.5 g of L-arginine (Sigma-Aldrich, USA) and 0.3 g of puerarin (Yuanye Bio-Technology, China) were incorporated into the pre-crosslinked solution and stirred for an additional 1 hour to ensure uniform distribution throughout the hydrogel matrix. Chemical crosslinking was then induced by adding 0.2 mL of hexamethylene diisocyanate (HDI, Sigma-Aldrich, USA), followed by thorough stirring for 1 hour. The resulting mixture was cast into polytetrafluoroethylene (PTFE) molds and allowed to cure at room temperature for 24 hours. Upon curing, the hydrogels were demolded and extensively washed with phosphate-buffered saline (PBS, pH 7.4, Gibco, USA) to eliminate unreacted monomers. To further reinforce mechanical strength, the hydrogels were subjected to three freeze–thaw cycles, consisting of freezing at − 20 °C for 12 hours and thawing at room temperature for 6 hours. The microstructure and surface morphology of the Si-A/PUE@HA composite hydrogels were analyzed using scanning electron microscopy (SEM, Hitachi SU8010, Japan). Prior to imaging, the hydrogel samples were freeze-dried using a lyophilizer (Labconco FreeZone 2.5, USA), fractured in liquid nitrogen, and sputter-coated with a thin layer of gold using a Quorum Q150R Plus sputter coater (UK) to enhance conductivity. SEM imaging was conducted at an accelerating voltage of 3.0 kV. Elemental distribution within the hydrogels was examined using energy-dispersive X-ray spectroscopy (EDS) integrated with the SEM system (Hitachi SU8010, Japan). This analysis was used to confirm the presence and uniform dispersion of key elements in the hydrogel matrix. The surface chemical composition of the hydrogels was investigated using X-ray photoelectron spectroscopy (XPS, Thermo Scientific ESCALAB 250Xi, USA). Spectra were recorded to identify characteristic binding energy peaks corresponding to carbon (C1s), oxygen (O1s), nitrogen (N1s), and silicon (Si2p), verifying the successful incorporation of functional components. The mechanical and viscoelastic properties of the hydrogels were assessed using a rotational rheometer (TA Instruments Discovery HR-2, USA) equipped with a parallel plate geometry (diameter: 20 mm). Frequency sweep measurements were conducted at room temperature to evaluate the storage modulus (G’) and loss modulus (G”) across a strain range of 0.1–1000%. The release profile of puerarin from the hydrogel was measured by immersing 1 g of the hydrogel in 10 mL of phosphate-buffered saline (PBS, pH 7.4) at 37°C. At designated time intervals, aliquots were withdrawn and replaced with fresh PBS. The concentration of puerarin released was quantified using a UV-Vis spectrophotometer (Shimadzu UV-2600, Japan) at a detection wavelength of 250 nm. The adhesive properties of the hydrogels were evaluated by applying the hydrogel to different substrates, including human skin, polymer (latex gloves), glass, and metal. The adhesion performance was qualitatively assessed and recorded using a Canon EOS 90D digital camera (Japan). Preparation of hydrogel extract for injection The Si-A/PUE@HA hydrogel was sterilized under ultraviolet (UV) light. After sterilization, both Si-A/PUE@HA and NC@HA hydrogels were added to DMEM at a concentration of 0.1 g/mL and incubated at 37 °C for 24 h. The supernatant was then collected and filtered through a 0.22 μm membrane filter. Standard DMEM medium without any hydrogel extract was used for the Control group in all in vitro experiments. The filtered solution was stored at 4 °C–− 20 °C for long-term preservation. Cell isolation and culture RAW 264.7 macrophage cells and rat bone marrow mesenchymal stem cells (BMSCs) were used for in vitro experiments. RAW 264.7 cells were obtained from the Cell Bank of the Chinese Academy of Sciences and cultured in DMEM supplemented with 10% fetal bovine serum (FBS, Gibco, USA) and 1% (v/v) penicillin-streptomycin (P/S, Gibco, USA). Rat BMSCs were purchased from Shanghai Fuyu Biotechnology Co., Ltd. and cultured according to the manufacturer’s instructions. BMSCs were maintained in α-minimal essential medium (α-MEM, Gibco) supplemented with 10% FBS and 1% penicillin-streptomycin. Both cell types were incubated at 37 °C with medium changes every two days. Only early-passage cells (passages 3–6) were used in the experiments. Cell proliferation and viability assays The biocompatibility of the Si-A/PUE@HA hydrogel was evaluated using a live/dead cell staining kit (Invitrogen, USA). In brief, BMSCs were cultured in the Si-A/PUE@HA hydrogel for 1, 3, and 7 days. Cells were washed three times with PBS and stained with PBS containing 2 mM calcein-AM (green fluorescence) and 4 mM propidium iodide (PI, red fluorescence) for 15 min at 37 °C. After another gentle PBS wash, the cells were visualized under a fluorescence microscope (Leica, USA). Cell proliferation was measured using the Cell Counting Kit-8 (CCK-8, Beyotime, China) on days 1, 3, and 7. Cells treated under different conditions were incubated in medium containing 10% CCK-8 at 37 °C for 2 h, and absorbance was measured at 450 nm using a microplate reader (BioTek, USA). Normal growth medium served as the control group. For the BMSCs proliferation assay, a bromodeoxyuridine (BrdU) incorporation kit (Cell Signaling Technology, USA) was used following the manufacturer’s instructions. BMSCs were seeded at 1 × 10^6 cells per well in a 6-well plate and exposed to various treatments for 24 h. BrdU was added to each well for the last 12 h of treatment. Cells were then fixed, washed twice with PBS, and incubated with mouse anti-BrdU antibody for 1 h at room temperature. Nuclei were stained with 4′,6-diamidino-2-phenylindole (DAPI) for 5 min. Immunofluorescence images were captured using a fluorescence microscope (Leica, Germany), and proliferation was quantified by calculating the percentage of BrdU-positive cells relative to DAPI-stained cells, or by counting BrdU-positive cells per square millimeter. Reactive oxygen species (ROS) assay To evaluate the hydrogel’s effect on reactive oxygen species (ROS) scavenging, BMSCs were seeded at 1 × 10^6 cells per well in a 6-well plate and incubated for 24 h to ensure proper attachment. After replacing the medium with extracts from Gel and Si-A/PUE@HA hydrogels, cells were incubated for an additional 6 h. Intracellular ROS levels were visualized using 2′,7′-dichlorofluorescin diacetate (DCFH-DA, Beyotime, China). Cells were incubated with 10 µM DCFH-DA in the dark for 20 min, and fluorescence images were captured using a fluorescence microscope (Leica, Germany) from five randomly selected areas per group. Quantification of fluorescence intensity was performed using ImageJ software. Wound healing assay BMSCs were seeded into 6-well culture plates at an appropriate density and incubated under standard conditions (37 °C, 5% CO₂) until a confluent monolayer was formed. A uniform linear scratch was created across the cell monolayer using a sterile 200 µL pipette tip to simulate a wound. Detached cells were carefully removed by washing the wells twice with sterile phosphate-buffered saline (PBS, pH 7.4). Fresh culture medium specific to each experimental group was then added to the wells. Immediately after scratching, images of the wound area were captured at 0 h using an inverted phase-contrast microscope. The cells were subsequently incubated, and additional images of the scratch area were taken at 12 h and 24 h to monitor cell migration and wound closure. The extent of wound closure was quantitatively analyzed using ImageJ software (NIH, USA) by calculating the percentage of the wound area covered over time. Chondrogenic differentiation of BMSCs To investigate the effects of various extracts on chondrogenic differentiation, BMSCs were harvested and pelleted after trypsinization, following a previously described protocol. The pellets were cultured in 15 mL centrifuge tubes, centrifuged (200 g, 5 min), and after 24 h, the medium was replaced with chondrogenic differentiation medium (CM) prepared with Si-A/PUE@HA extracts. The CM contained 10 ng/mL TGFβ3 (Sigma, USA), 1 mM sodium pyruvate (Sigma, USA), 0.1 µM dexamethasone (Sigma, USA), 1% ITS Premix (Sigma, USA), and 1 µM ascorbate-2-phosphate (Sigma, USA). CM without extract served as the control group. After three weeks, the chondrogenic pellets were cryo-sectioned (7 μm thickness) and subjected to histological analysis, including hematoxylin and eosin (HE), alcian blue (AB), safranin O staining, and immunohistochemistry (IHC). The Bern score was used to assess in vitro chondrogenesis. Real-time quantitative polymerase chain reaction (RT-qPCR) was conducted to assess the expression of chondrogenesis-related genes, including collagen type I α1 (Col1a1), collagen type II α1 (Col2a1), SRY-related HMG-box gene 9 (Sox9), and aggrecan (Acan). Total RNA was extracted using the EZ-press RNA Purification Kit (EZBioscience), followed by cDNA synthesis using a 4× Reverse Transcription Master Mix (EZBioscience). RT-qPCR was performed using a 2× SYBR Green qPCR Master Mix (EZBioscience) on a Light Cycler system (Bio-Rad, USA). Relative mRNA expression levels were calculated using the 2-ΔΔCt method and normalized to the housekeeping gene GAPDH. Primer sequences are listed in Table S1. Macrophage polarization assay Macrophage polarization induced by Si-A/PUE@HA hydrogel extracts was assessed via immunofluorescence staining, specifically evaluating the expression of M1 markers (iNOS, CD86) and M2 markers (CD163). To simulate post-implantation inflammation, RAW 264.7 cells were seeded in 6-well plates and treated with 100 ng/mL lipopolysaccharide (LPS, Sigma, USA) for 24 h. Cells were then exposed to normal growth medium or Si-A/PUE@HA hydrogel extracts for 3 days. After treatment, cells were fixed with 4% paraformaldehyde, permeabilized with 0.1% Triton X-100, and blocked with 5% bovine serum albumin (BSA). Primary antibodies against iNOS (Abcam, UK), CD86 (Abcam, UK), F4/80 (Abcam, UK), and CD163 (Abcam, UK) were applied at a 1:500 dilution and incubated overnight at 4 °C. Secondary antibodies conjugated with Alexa Fluor 488 or Alexa Fluor 594 (Abcam, UK) were applied for 1 h, followed by DAPI staining for 5 min. Fluorescence images were captured using a microscope. Flow cytometry was used to further assess CD86 (PE-labeled, eBioscience, USA) and CD163 (FITC-labeled, eBioscience, USA) expression levels. Cells were scraped, centrifuged, blocked with 5% BSA, stained with the appropriate antibodies, and analyzed using a flow cytometer (BD, USA). CytExpert 2.4 software was used for data analysis. Additionally, the expression of polarization-related genes (TNF-α, IL-6, IL-1β, IL-4, IL-10, and IL-1ra) was evaluated by RT-qPCR. Surgical procedure for osteochondral defect modeling Forty 6-week-old male Sprague Dawley (SD) rats were randomly assigned into groups according to the experimental design. The procedure was as follows: after anesthetizing the rats, shaving the fur, and disinfecting the surgical area, a medial incision was made on the knee joint. The patellar ligament and patella were everted to expose the femoral condyle. A micro electric drill was then used to create a defect (2 mm in diameter, 1.5 mm in depth) at the center of the trochlear groove. In the model group, the defect site was rinsed with PBS. In the two experimental groups, the defects were filled with 20 µL of NC@HA or Si-A/PUE@HA hydrogel using a microinjector. In the control group, the knee joint was exposed but no drilling was performed. The patella was repositioned, the incision was sutured, and the area was disinfected again. Postoperatively, all rats were housed in a specific pathogen-free (SPF) animal facility and allowed to move freely. Rats were sacrificed at 4 and 8 weeks after surgery for further analysis. Micro-CT analysis At 6 and 12 weeks post-surgery, the Sprague Dawley (SD) rats were euthanized, and their femurs were harvested. Samples were fixed in 4% paraformaldehyde solution (Servicebio, China) at 4 °C overnight to preserve their structural integrity. Morphological evaluation was subsequently performed using a micro-computed tomography (Micro-CT) scanner (SkyScan 1276, Bruker, Germany). The scanning parameters were set as follows: a voxel size of 10 μm, voltage of 70 kV, current of 200 µA, a 1 mm aluminum filter, and an integration time of 350 ms to ensure high-resolution imaging. The raw Micro-CT data were reconstructed using NRecon software (version 1.7.4.2, Bruker, Germany). For three-dimensional visualization, CTvox software (version 3.3, Bruker, Germany) was utilized to convert the contoured two-dimensional images into detailed 3D models. Specific regions of interest (ROIs) were extracted from the 3D reconstructions using DataViewer software (version 1.5.1.9, Bruker, Germany). Quantitative analysis of bone regeneration was conducted using CTAn software (version 1.20.3, Bruker, Germany). Key parameters, including the bone volume fraction (BV/TV), were calculated to assess subchondral bone reconstruction. Additionally, the area of bilateral bone defects in the coronal sections was measured using ImageJ software (NIH, USA) for further analysis. In vivo cartilage regeneration evaluation At 6 and 12 weeks post-surgery, the repaired articular cartilage was harvested for morphological evaluation. The samples were fixed in 4% paraformaldehyde solution for 5 days to preserve tissue integrity. Following fixation, the samples underwent continuous trimming during a 2-month decalcification process. Upon completion of decalcification, the specimens were dehydrated through a graded ethanol series, embedded in paraffin, and sectioned into slices of 5 μm thickness using a microtome. For histological analysis, the sections were stained following standard protocols with hematoxylin and eosin (H&E) for general morphology, Safranin O/Fast Green for glycosaminoglycan content, and Toluidine Blue (TB) for cartilage matrix evaluation. Immunohistochemical staining for type II collagen (COL2) was performed to assess cartilage-specific matrix formation. Briefly, paraffin-embedded sections were dewaxed and rehydrated. Endogenous peroxidase activity was quenched using hydrogen peroxide (H₂O₂). Permeabilization was achieved with Triton X-100, followed by phosphate-buffered saline (PBS) washing. The sections were then blocked with a suitable blocking buffer and incubated overnight at 4 °C with a primary anti-collagen type II antibody (1:200, Abcam, USA). Subsequently, the sections were incubated with a secondary antibody, and signal detection was conducted using a chromogenic substrate. The stained sections were visualized and imaged under a light microscope. All histological images were independently assessed by a pathologist with extensive experience in cartilage histopathology, who was blinded to the experimental groups. Cartilage repair quality was quantitatively evaluated using the Modified O’Driscoll Scoring System and the International Cartilage Repair Society (ICRS) Visual Histological Assessment Scale (Table [81]S2−3). Single-Cell RNA sequencing with 10x genomics chromium Articular cartilage samples were collected from 10 experimental rats, ground, and prepared for single-cell RNA sequencing (scRNA-seq) to evaluate cellular responses. Viable cells were isolated and treated with red blood cell lysis buffer, followed by magnetic bead separation to remove erythrocytes and dead cells. Cell viability was assessed using trypan blue staining (Thermo Fisher Scientific, Waltham, MA, USA) and a hemocytometer (Thermo Fisher Scientific). After counting, cell suspensions were adjusted based on a target of 7000 cells per capture. For samples with concentrations below the recommended 700–1200 cells/µL, the cells were centrifuged and resuspended in a smaller volume before loading into the 10x Genomics Single Cell A Chip. Reverse transcription and library preparation were performed using the 10x Genomics Single Cell v2 kit, following the manufacturer’s protocol. The libraries were multiplexed and sequenced using an Illumina NextSeq-500 platform with a high-output (400 m) kit in a single lane. Quality control of the scRNA-seq data was performed using the Cell Ranger pipeline (10x Genomics), which aligned the reads to the mm10 reference genome and generated a digital gene expression matrix. This matrix was filtered and normalized using the Seurat R package (version 2.3.4). Cells with fewer than 500 unique genes, greater than 50,000 UMI counts, or over 10% mitochondrial reads were excluded from further analysis. Genes that were not detected in any cells were also removed from subsequent analysis. Single-cell RNA sequencing data processing The gene expression matrix was normalized and scaled using the NormalizeData and ScaleData functions in Seurat. Principal component analysis (PCA) was conducted using the RunPCA function for dimensionality reduction and clustering. DecontX was employed to predict contamination levels and remove abnormally expressed marker genes from mixed datasets. Batch effects at the sample level were corrected using Harmony. Cell subpopulations were visualized using UMAP, and characteristic gene expression was evaluated using Seurat functions such as Dotplot and FeaturePlot. Differential expression analysis was performed with Seurat’s FindAllMarkers function, assuming a negative binomial distribution for likelihood ratio testing. Only genes expressed in more than 25% of cells in a cluster with a log2 fold change greater than 0.25 were considered for differential expression. Cell type identification was carried out using the “Single R” software, along with insights from published studies and expert annotations. Pathway enrichment analysis Functional enrichment analysis, including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, was conducted to determine whether differentially expressed genes (DEGs) were significantly enriched in specific GO terms or metabolic pathways. GO is an internationally standardized classification system for gene function. DEGs were mapped to GO terms representing biological functions, and gene counts within each term were calculated. A hypergeometric test was then performed to identify GO terms that were significantly enriched in the gene list compared to a reference gene list. GO terms and KEGG pathways with a false discovery rate (FDR) of P < 0.05 were considered significantly enriched. Toxicity evaluation of Si-A/PUE@HA At 12 weeks post-surgery, the rats were euthanized, and major organs, including the heart, liver, spleen, lungs, and kidneys, were collected. These organs were subjected to hematoxylin and eosin (H&E) staining and Masson staining, as well as F4/80 and α-SMA immunofluorescence staining, to comprehensively evaluate the biosafety of the Si-A/PUE@HA composite hydrogel. Statistical analysis All data were expressed as the mean ± standard deviation (SD). Statistical analyses were performed using one-way analysis of variance (ANOVA) followed by Tukey’s post hoc test. SPSS 22.0 software was used for statistical analysis. A P-value of less than 0.05 was considered statistically significant Fig. [82]1. Fig. 1. [83]Fig. 1 [84]Open in a new tab Schematic illustration of the synthesis and therapeutic application of the Si-A/PUE@HA hydrogel. Hyaluronic acid and siliconol are combined with arginine through physical crosslinking and puerarin through self-assembly to form an injectable hydrogel. The hydrogel is injected into the osteochondral defect site, where it promotes macrophage polarization from M1 to M2 and facilitates the differentiation of bone marrow mesenchymal stem cells (BMSCs) into chondrocytes, thereby accelerating cartilage regeneration Result Structural and physicochemical characterization of the Si-A/PUE@HA hydrogel The Si-A/PUE@HA hydrogel was developed through a self-assembly and physical crosslinking strategy involving hyaluronic acid, silicool, arginine, and puerarin (Fig. [85]2A), resulting in a multifunctional material with enhanced mechanical strength, bioactivity, and adhesive properties. The hydrogel exhibited a distinct sol–gel transition, transitioning from a flowable sol to a stable gel (Fig. [86]2B), enabling in situ application and conformal coating on irregular surfaces. Its excellent injectability was further demonstrated by smooth extrusion and the formation of self-supporting structures (Fig. [87]2C), highlighting its potential for minimally invasive delivery in clinical settings. Scanning electron microscopy (SEM) revealed a highly porous and interconnected network structure (Fig. [88]2D, Supplementary Fig. [89]1), which is critical for facilitating nutrient diffusion, cell infiltration, and sustained drug release. Energy-dispersive X-ray spectroscopy (EDS) mapping confirmed the uniform distribution of carbon (C), oxygen (O), sodium (Na), and silicon (Si) throughout the hydrogel matrix (Fig. [90]2E), validating the homogeneous integration of the hydrogel components. X-ray photoelectron spectroscopy (XPS) further confirmed the successful incorporation of silicool and puerarin, as evidenced by characteristic peaks for C 1 s, O 1 s, N 1 s, and Si 2p (Fig. [91]2F). Rheological analysis revealed a sol–gel transition behavior, where the storage modulus (G′) surpassed the loss modulus (G″) around 37 °C (Fig. [92]2G), indicating the hydrogel’s temperature-responsive transformation from a sol to a gel state. This transition demonstrates that the hydrogel acquires solid-like mechanical properties under physiological temperature, which is critical for maintaining structural integrity in vivo. The hydrogel also exhibited a sustained and controlled puerarin release over 14 days (Fig. [93]2H), supporting its potential for long-term therapeutic applications. Notably, the hydrogel adhered strongly to various substrates, including skin, polymer, glass, and metal surfaces (Fig. [94]2I), showcasing its versatile adhesive capability for biomedical applications. Collectively, these results demonstrate that the Si-A/PUE@HA hydrogel integrates injectability, mechanical robustness, sustained drug release, and broad-spectrum adhesion, offering significant promise for applications in drug delivery, tissue engineering, and regenerative medicine. Fig. 2. [95]Fig. 2 [96]Open in a new tab Characterization and performance analysis of the Si-A/PUE@HA hydrogel. (A) Schematic illustration of the hydrogel formation through physical crosslinking and self-assembly of hyaluronic acid, siliconol, arginine, and puerarin. (B) Photographs showing the sol–gel transition behavior of the Si-A/PUE@HA hydrogel. (C) Demonstration of the injectability of the Si-A/PUE@HA hydrogel. (D) SEM images showing the porous microstructure of the hydrogel at different magnifications. (E) SEM and corresponding energy-dispersive X-ray spectroscopy (EDS) mapping images highlighting the distribution of elements: carbon (C, red), oxygen (O, green), sodium (Na, cyan), and silicon (Si, purple). (F) X-ray photoelectron spectroscopy (XPS) spectrum confirming the chemical composition of the hydrogel. (G) Rheological analysis illustrating the storage modulus (G’) and loss modulus (G’’) of the hydrogel over a range of strain percentages. (H) Cumulative release profile of puerarin (PUE) from the hydrogel over 14 days. (I) Adhesion performance of the hydrogel on various substrates: skin, polymer, glass, and metal In vitro cytocompatibility The in vitro cytocompatibility of the Si-A/PUE@HA hydrogel was systematically evaluated using bone marrow mesenchymal stem cells (BMSCs). Live/dead staining results (Fig. [97]3A, B) showed that the Si-A/PUE@HA hydrogel supported high cell viability, with only a few dead cells (red fluorescence) observed, comparable to the control and NC@HA groups, indicating negligible cytotoxicity. Cell proliferation was further assessed using the BrdU incorporation assay and the CCK-8 assay. BrdU staining (Fig. [98]3C, D) revealed a significant increase in BrdU-positive cells in the Si-A/PUE@HA group compared to the NC@HA group (p < 0.05), demonstrating enhanced DNA synthesis and cell proliferation. Similarly, CCK-8 analysis (Fig. [99]3E) showed no significant difference in cell viability between the Si-A/PUE@HA group and the control, confirming the hydrogel’s ability to support cell growth over time. The antioxidative capacity of the hydrogel was examined under oxidative stress induced by hydrogen peroxide (H₂O₂). As shown in Fig. [100]3F, BMSCs exposed to H₂O₂ exhibited intense green fluorescence, indicating elevated intracellular reactive oxygen species (ROS). In contrast, treatment with the Si-A/PUE@HA hydrogel markedly reduced ROS levels, as quantified by DCFH-DA fluorescence intensity (Fig. [101]3G), confirming its potent antioxidative effect. To investigate the hydrogel’s impact on cell migration and wound healing, a scratch assay was performed. Brightfield images (Fig. [102]3H) and quantitative analysis (Fig. [103]3I, J) demonstrated that the Si-A/PUE@HA hydrogel significantly promoted BMSC migration and wound closure at both 12 and 24 h compared to the NC@HA and control groups (p < 0.01), highlighting its strong pro-healing capability. Collectively, these results confirm that the Si-A/PUE@HA hydrogel exhibits excellent in vitro cytocompatibility by supporting cell viability and proliferation while providing significant antioxidative and pro-migratory effects. These properties position the Si-A/PUE@HA hydrogel as a promising candidate for applications in tissue engineering and regenerative medicine. Fig. 3. [104]Fig. 3 [105]Open in a new tab In vitro biocompatibility, antioxidative stress, and wound healing evaluation of Si-A/PUE@HA hydrogel. (A) Live/dead cell staining images of cells treated with Control, NC@HA, and Si-A/PUE@HA groups. Live cells are stained with Calcein-AM (green), and dead cells with PI (red). (B) Quantification of the percentage of live cells in each group. (C) Immunofluorescence images showing BrdU (green) and DAPI (blue) staining to assess cell proliferation in different groups. (D) Quantification of BrdU-positive cells relative to DAPI-stained nuclei. (E) CCK-8 assay evaluating cell viability across the Control, NC@HA, and Si-A/PUE@HA groups. (F) Fluorescence microscopy images of intracellular reactive oxygen species (ROS) detected by DCFH-DA staining under oxidative stress induced by H₂O₂, with and without hydrogel treatment. (G) Quantification of ROS levels based on relative fluorescence intensity. (H) Wound healing assay images at 0 h, 12 h, and 24 h showing cell migration in different treatment groups. (I) Quantification of wound healing percentage at 12 h. (J) Quantification of wound healing percentage at 24 h. *Statistical significance: ns (not significant), *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 Promotion of in vitro chondrogenesis in BMSCs To evaluate the chondrogenic differentiation potential of the Si-A/PUE@HA hydrogel, bone marrow mesenchymal stem cells (BMSCs) were cultured in chondrogenic medium for 21 days. All cell pellets developed a whitish, round morphology, indicating the formation of cartilage-like tissue. Histological analyses were conducted to assess cartilage matrix deposition. Hematoxylin and eosin (H&E) staining (Fig. [106]4A) revealed that all groups exhibited cartilage tissue formation, with the Si-A/PUE@HA group showing the most abundant extracellular matrix (ECM) deposition. Alcian Blue staining (Fig. [107]4B) and Safranin O staining (Fig. [108]4C) further confirmed the enhanced synthesis of glycosaminoglycans (GAGs) and proteoglycans in the Si-A/PUE@HA group compared to the control and NC@HA groups. Immunohistochemical staining for type II collagen (Col II), a key cartilage-specific ECM component, demonstrated the strongest Col II expression in the Si-A/PUE@HA group (Fig. [109]4D), suggesting superior cartilage matrix production. The Bern score, a histological grading system for cartilage formation, was significantly higher in the Si-A/PUE@HA group than in the other groups (Fig. [110]4E), indicating improved chondrogenic differentiation. Additionally, RT-qPCR analysis showed that the Si-A/PUE@HA hydrogel significantly upregulated the expression of key chondrogenic marker genes, including Acan (Fig. [111]4F), Sox9 (Fig. [112]4G), and Col2a1 (Fig. [113]4H), compared to the control and NC@HA groups. In contrast, the expression of the fibrocartilage marker gene Col1a1 was significantly lower in the Si-A/PUE@HA group (Fig. [114]4I), indicating a reduced tendency toward fibrocartilage formation. These findings collectively demonstrate that the Si-A/PUE@HA hydrogel effectively promotes the chondrogenic differentiation of BMSCs by enhancing cartilage-specific matrix production and gene expression. This highlights its potential as a promising biomaterial for cartilage tissue engineering and regenerative medicine. Fig. 4. [115]Fig. 4 [116]Open in a new tab In vitro chondrogenesis of BMSCs induced by Si-A/PUE@HA hydrogel. (A) H&E staining showing the morphology of BMSC-derived cartilage tissue in the Control, NC@HA, and Si-A/PUE@HA groups. Scale bar: 50 μm. (B) Alcian blue staining indicating glycosaminoglycan (GAG) accumulation in the extracellular matrix. (C) Safranin O staining demonstrating enhanced proteoglycan deposition in the Si-A/PUE@HA group. (D) Immunohistochemical (IHC) staining of type II collagen (COL II), confirming cartilage-specific matrix formation. (E) Quantitative analysis of Bern scores evaluating cartilage tissue quality. (F–I) Relative mRNA expression levels of cartilage-related genes: Acan (F), Sox9 (G), Col2a1 (H), and Col1a1 (I), indicating enhanced chondrogenic differentiation in the Si-A/PUE@HA group. *Statistical significance: ns (not significant), *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 In vitro modulation of macrophage polarization To evaluate the immunomodulatory properties of the Si-A/PUE@HA hydrogel, RAW 264.7 macrophages were cultured with different treatments and stimulated with lipopolysaccharide (LPS) for 24 h to mimic an inflammatory microenvironment (Fig. [117]5A). Immunofluorescence staining revealed that LPS stimulation significantly increased the expression of the M1 macrophage marker CD86 (Fig. [118]5B, D), indicating enhanced pro-inflammatory polarization. However, treatment with the Si-A/PUE@HA hydrogel markedly suppressed CD86 expression compared to both the LPS and LPS + NC@HA groups, demonstrating the hydrogel’s ability to inhibit M1 macrophage polarization. Conversely, the Si-A/PUE@HA hydrogel significantly promoted M2 macrophage polarization, as evidenced by the elevated expression of the anti-inflammatory marker CD163 (Fig. [119]5C, E). Flow cytometry analysis further confirmed these results, showing a substantial decrease in CD86⁺ M1 macrophages (Fig. [120]5F, G) and a significant increase in CD163⁺ M2 macrophages (Fig. [121]5H, I) in the Si-A/PUE@HA group compared to the LPS and LPS + NC@HA groups. Moreover, quantitative real-time PCR (qRT-PCR) analysis revealed that the Si-A/PUE@HA hydrogel significantly downregulated the expression of pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-6 (Fig. [122]5J), while markedly upregulating anti-inflammatory cytokines such as IL-4, IL-10, and IL-1RA (Fig. [123]5K). Collectively, these results demonstrate that the Si-A/PUE@HA hydrogel effectively modulates macrophage polarization by suppressing the pro-inflammatory M1 phenotype and promoting the anti-inflammatory M2 phenotype. This immunoregulatory effect highlights the potential of Si-A/PUE@HA hydrogel as a promising therapeutic strategy for controlling inflammation and enhancing tissue regeneration. Fig. 5. [124]Fig. 5 [125]Open in a new tab Immunomodulatory effects of Si-A/PUE@HA hydrogel on macrophage polarization. (A) Schematic of the experimental design for evaluating macrophage responses. RAW 264.7 cells were treated with Control, LPS, LPS + NC@HA, or LPS + Si-A/PUE@HA for 24 h, followed by fluorescence microscopy, flow cytometry, and qRT-PCR analysis. (B) Immunofluorescence staining of CD86 (green, M1 marker), F4/80 (red, macrophage marker), and DAPI (blue, nuclei) showing macrophage polarization in different groups. (C) Immunofluorescence staining of CD163 (green, M2 marker), F4/80 (red), and DAPI (blue) across groups. (D) Quantification of relative fluorescence intensity of CD86 expression. (E) Quantification of relative fluorescence intensity of CD163 expression. (F) Flow cytometry analysis of CD86 (M1) and F4/80 double-positive cells. (G) Quantification of CD86-positive cells from flow cytometry data. (H) Flow cytometry analysis of CD163 (M2) and F4/80 double-positive cells. (I) Quantification of CD163-positive cells from flow cytometry data. (J) Relative mRNA expression of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6) in different groups. (K) Relative mRNA expression of anti-inflammatory cytokines (IL-4, IL-10, IL-1RA) in different groups. *Statistical significance: ns (not significant), *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 Si-A/PUE@HA hydrogel enhances in vivo cartilage regeneration Building on the promising in vitro results of BMSC chondrogenic differentiation, we further investigated the in vivo cartilage repair potential of the Si-A/PUE@HA hydrogel using a rat osteochondral defect model (Fig. [126]6A). Micro-CT imaging at 6 and 12 weeks post-implantation revealed minimal tissue regeneration in the model group, with distinct boundaries between the defect and native cartilage. In contrast, the Si-A/PUE@HA group showed substantial new tissue formation that was more seamlessly integrated with the surrounding cartilage, especially at 12 weeks, where the defect was nearly filled with smooth, cartilage-like tissue resembling native cartilage (Fig. [127]6B). Quantitative analysis of Micro-CT data provided further confirmation of these findings. The defect area percentage was significantly reduced in the Si-A/PUE@HA group compared to the model and NC@HA groups at both 6 and 12 weeks (Supplementary Fig. [128]2 A). Notably, the Si-A/PUE@HA group demonstrated the most significant reduction in defect area, indicating superior tissue regeneration. Additionally, the bone volume/total volume (BV/TV) ratio, an indicator of bone regeneration, was markedly higher in the Si-A/PUE@HA group than in the other groups at both time points (Supplementary Fig. [129]2B), suggesting enhanced subchondral bone repair. Histological analyses further confirmed these findings. Hematoxylin and eosin (H&E) staining (Fig. [130]6C) and Safranin O/Fast Green staining (Fig. [131]6D) demonstrated limited tissue regeneration in the model group, characterized by fibrous and poorly integrated tissue. In contrast, the Si-A/PUE@HA group exhibited dense, cartilage-like tissue with enhanced matrix deposition and improved integration with native cartilage. Toluidine blue staining (Fig. [132]6E) and type II collagen immunohistochemistry (IHC) staining (Fig. [133]6F) revealed increased glycosaminoglycan content and higher type II collagen expression in the Si-A/PUE@HA group, indicating the formation of hyaline-like cartilage. Quantitative assessments further supported these findings. The International Cartilage Repair Society (ICRS) scores were significantly higher in the Si-A/PUE@HA group at both 6 and 12 weeks compared to the model and NC@HA groups (Fig. [134]6G). Similarly, the Modified O’Driscoll Histological Scores (MOHS) confirmed superior cartilage repair quality in the Si-A/PUE@HA group (Fig. [135]6H). Immunohistochemical quantification of type II collagen (Col2) also showed significantly higher mean optical density (IOD) in the Si-A/PUE@HA group at both time points (Fig. [136]6I), further indicating enhanced cartilage matrix formation. Collectively, these results demonstrate that the Si-A/PUE@HA hydrogel significantly promotes in vivo cartilage regeneration by enhancing new tissue formation, improving tissue integration, and supporting the regeneration of hyaline-like cartilage. These findings highlight the hydrogel’s strong potential for clinical translation in cartilage repair and regenerative medicine. Fig. 6. [137]Fig. 6 [138]Open in a new tab In vivo evaluation of cartilage regeneration promoted by Si-A/PUE@HA hydrogel. (A) Schematic diagram of the experimental design for in vivo osteochondral defect repair in rats, including surgery, treatment, and analysis at 6 and 12 weeks. (B) Micro-CT images showing the repaired osteochondral in the Control, Model, NC@HA, and Si-A/PUE@HA groups at 6 and 12 weeks. (C) Hematoxylin and eosin (H&E) staining displaying tissue morphology and cartilage repair in different groups at 6 and 12 weeks. (D) Safranin O–Fast Green staining indicating proteoglycan deposition in regenerated cartilage. (E) Toluidine blue staining showing glycosaminoglycan (GAG) distribution in repaired cartilage. (F) Immunohistochemical (IHC) staining for type II collagen, confirming cartilage-specific matrix formation. (G) Quantitative analysis of ICRS scores at 6 and 12 weeks, evaluating cartilage repair quality. (H) Quantitative analysis of Modified O’Driscoll Histological Scores (MOHS) at 6 and 12 weeks. (I) Quantification of type II collagen (Col2) expression based on mean integrated optical density (IOD) at 6 and 12 weeks. *Statistical significance: ns (not significant), *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 Si-A/PUE@HA hydrogel modulates immune cell populations and macrophage polarization To investigate the immunoregulatory effects of the Si-A/PUE@HA hydrogel on the local tissue microenvironment, single-cell RNA sequencing (scRNA-seq) was performed on regenerated tissue from the Model and Si-A/PUE@HA groups. UMAP analysis revealed distinct clustering of immune and stromal cell populations, including macrophages, fibroblasts, chondrocytes, B cells, and T/NK cells, with the Si-A/PUE@HA group exhibiting a significant shift towards a more regenerative immune landscape compared to the Model group (Fig. [139]7A, C). Dot plot analysis showed differential expression of key marker genes, with pro-inflammatory markers such as Cd86 highly expressed in the Model group, while anti-inflammatory markers like Cd163 were markedly upregulated in the Si-A/PUE@HA group (Fig. [140]7B). To further elucidate the molecular mechanisms underlying the immunoregulatory and chondrogenic effects of the Si-A/PUE@HA hydrogel, functional enrichment analysis of differentially expressed genes (DEGs) was performed on monocytes/macrophages and chondrocytes. The heatmap analysis revealed distinct expression patterns of key DEGs across various cell types, including chondrocytes, fibroblasts, neutrophils, B cells, monocytes/macrophages, proliferating cells, neural progenitor cells (NPCs), plasmacytoid dendritic cells (pDCs), and endothelial cells (ECs) (Supplementary Fig. [141]3 A). Gene ontology (GO) term enrichment word clouds provided deeper insights into the functional roles of DEGs in monocytes/macrophages and chondrocytes (Supplementary Fig. [142]3B). In monocytes/macrophages, genes associated with inflammation regulation and immune modulation, such as Trem2 and Pycarr, were highly enriched. In chondrocytes, key genes involved in cartilage development and matrix synthesis, including Sox9, Vegfa, and Bmp2, showed prominent enrichment. The larger and warmer-colored gene names in the word clouds represent higher enrichment scores, emphasizing the critical roles of these genes in modulating inflammation and promoting chondrogenesis. Gene set enrichment analysis (GSEA) further highlighted that Si-A/PUE@HA treatment suppressed inflammatory pathways and promoted immune-regulatory and tissue repair pathways (Fig. [143]7D). Macrophage-specific analysis identified four subpopulations: C0 (C6⁺ macrophages), C1 (Tiparp⁺ macrophages), C2 (Mmp12⁺ macrophages), and C3 (Vcam1⁺ macrophages), with the Si-A/PUE@HA group displaying reduced pro-inflammatory C2 macrophages and increased anti-inflammatory C1 and C3 macrophages (Fig. [144]7E). Heatmap analysis confirmed elevated expression of anti-inflammatory and tissue-regenerative genes in the Si-A/PUE@HA group, alongside reduced pro-inflammatory gene expression (Fig. [145]7F, G). Finally, scatter plot analysis demonstrated transcriptional reprogramming of macrophages, with a shift from pro-inflammatory phenotypes to anti-inflammatory, tissue-repairing states induced by Si-A/PUE@HA treatment (Fig. [146]7H). These findings indicate that the Si-A/PUE@HA hydrogel effectively modulates immune cell composition and macrophage polarization, creating a favorable microenvironment for tissue regeneration. Fig. 7. [147]Fig. 7 [148]Open in a new tab Single-cell RNA sequencing analysis reveals the immunoregulatory effects of Si-A/PUE@HA hydrogel. (A) UMAP plot showing the distribution of different cell populations in the Model and Si-A/PUE@HA groups, highlighting distinct immune cell clusters. (B) Dot plot displaying the expression levels of representative marker genes across various cell types and treatment groups. The size and color of the dots represent gene expression levels and the percentage of expressing cells, respectively. (C) Proportional distribution of cell populations between the Model and Si-A/PUE@HA groups. (D) Gene set enrichment analysis (GSEA) of differentially expressed genes (DEGs) between the Model and Si-A/PUE@HA groups, indicating enriched biological pathways. (E) Clustering of macrophage subpopulations, including C0 (C6 + macrophages), C1 (Tgmp + macrophages), C2 (Ympt2 + macrophages), and C3 (Vcam1 + macrophages). (F) Heatmap showing the mean expression of immune-related genes across different macrophage subtypes in both groups. (G) Heatmap of differentially expressed genes related to immune modulation between the Model and Si-A/PUE@HA groups. (H) Scatter plots illustrating the gene expression correlations among distinct macrophage subtypes, highlighting transcriptional shifts induced by Si-A/PUE@HA treatment Modulation of macrophage subtypes and polarization Single-cell RNA sequencing (scRNA-seq) and functional analyses were conducted to investigate the immunomodulatory effects of the Si-A/PUE@HA hydrogel on macrophage polarization and differentiation. Heatmap analysis revealed distinct distributions of macrophage subtypes—C0 (C6⁺ macrophages), C1 (Tiparp⁺ macrophages), C2 (Mmp12⁺ macrophages), and C3 (Vcam1⁺ macrophages)—between the Model and Si-A/PUE@HA groups, with the Si-A/PUE@HA group showing a significant reduction in pro-inflammatory C2 (Mmp12⁺) macrophages and an increase in anti-inflammatory C1 (Tiparp⁺) and C3 (Vcam1⁺) subtypes (Fig. [149]8A). UMAP visualization confirmed that Si-A/PUE@HA treatment effectively suppressed M1 macrophage polarization while promoting M2 macrophage phenotypes, as indicated by reduced pro-inflammatory gene expression and increased anti-inflammatory gene expression (Fig. [150]8B). Pseudotime trajectory analysis further demonstrated that macrophage differentiation in the Si-A/PUE@HA group favored anti-inflammatory lineages, contrasting with the pro-inflammatory trajectory in the Model group (Fig. [151]8C–F), and gene expression dynamics of key markers (C6, Tiparp, Mmp12, Vcam1) validated this shift (Fig. [152]8G). Further analysis revealed lower genomic instability in the Si-A/PUE@HA group, indicated by reduced CNVscore (Supplementary Fig. [153]4 A), along with enhanced stemness properties (higher Cell_Stemness_AUC score) and reduced macrophage proliferation, as shown by decreased G2/M and S phase cell cycle scores (Supplementary Fig. [154]4B–D). Additionally, gene expression profiling showed downregulation of M1 markers and pro-inflammatory genes, with upregulation of M2 markers and anti-inflammatory genes in the Si-A/PUE@HA group (Supplementary Fig. [155]5A–D). GO enrichment analysis further highlighted that immune regulation, cytokine signaling, leukocyte migration, apoptosis inhibition, and stress response pathways were enriched in anti-inflammatory macrophage subtypes (Supplementary Fig. [156]5E). Collectively, these findings demonstrate that the Si-A/PUE@HA hydrogel effectively reprograms macrophage polarization by suppressing pro-inflammatory M1 phenotypes and enhancing anti-inflammatory M2 differentiation, while also improving genomic stability, promoting stemness, and reducing proliferation, thereby fostering a regenerative microenvironment conducive to tissue repair. Fig. 8. [157]Fig. 8 [158]Open in a new tab Analysis of macrophage polarization and lineage trajectory influenced by Si-A/PUE@HA hydrogel. (A) Heatmap showing the distribution of macrophage subtypes (C0 C6+, C1 Tiparp+, C2 Mmp12+, C3 Vcam1+) in the Model and Si-A/PUE@HA groups. (B) UMAP visualization of M1/M2 macrophage polarization and pro-/anti-inflammatory gene expression profiles. (C) Pseudotime trajectory analysis illustrating the differentiation paths of macrophage subtypes. (D) Predicted ordering of macrophage subtypes along the differentiation trajectory. (E) Proportional distribution of cell states in different groups during macrophage differentiation. (F) UMAP plot displaying the lineage trajectory of macrophage subpopulations, with Lineage 1 and Lineage 2 marked. (G) Expression dynamics of marker genes (C6, Tiparp, Mmp12, Vcam1) along pseudotime across different lineages Si-A/PUE@HA hydrogel enhances chondrocyte differentiation and cartilage regeneration Single-cell RNA sequencing (scRNA-seq) analysis was conducted to explore the chondrogenic regulatory effects of the Si-A/PUE@HA hydrogel. UMAP analysis revealed six distinct chondrocyte subpopulations: C0 (Btg2⁺ chondrocytes), C1 (Tiam2⁺ chondrocytes), C2 (Ibsp⁺ chondrocytes), C3 (Col11a2⁺ chondrocytes), C4 (Lyz2⁺ chondrocytes), and C5 (Cd74⁺ chondrocytes) (Fig. [159]9A). Feature plots demonstrated elevated expression of key chondrogenic markers (Fos, Lyz2, Tiam2, Cd74, Col11a2, and Ibsp) in the Si-A/PUE@HA group compared to the Model group, indicating enhanced chondrogenic activity (Fig. [160]9B). Hierarchical clustering and heatmap analyses further confirmed distinct gene expression profiles among these chondrocyte subtypes, with significant upregulation of cartilage-related genes in the Si-A/PUE@HA group (Fig. [161]9C–D). Additionally, the distribution of chondrocyte subpopulations showed an increased proportion of regenerative C3 (Col11a2⁺) and C5 (Cd74⁺) chondrocytes in the Si-A/PUE@HA group, suggesting enhanced cartilage formation (Fig. [162]9E). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses revealed that differentially expressed genes (DEGs) in the Si-A/PUE@HA group were significantly enriched in pathways related to extracellular matrix (ECM) organization, wound healing, and cartilage development (Fig. [163]9F). Pseudotime trajectory analysis demonstrated accelerated chondrocyte differentiation and maturation in the Si-A/PUE@HA group, with dynamic gene expression changes supporting cartilage regeneration (Fig. [164]9G–H). Cell-cell interaction network analysis further highlighted stronger communication between chondrocytes and other regenerative cells in the Si-A/PUE@HA group, facilitating coordinated cartilage repair (Fig. [165]9I–J). Fig. 9. [166]Fig. 9 [167]Open in a new tab Single-cell RNA sequencing analysis reveals the chondrogenic regulatory effect of Si-A/PUE@HA hydrogel. (A) UMAP plot showing distinct chondrocyte subpopulations, including C0 Btg2 + chondrocytes, C1 Tiam2 + chondrocytes, C2 Ibsp + chondrocytes, C3 Col11a2 + chondrocytes, C4 Lyz2 + chondrocytes, and C5 Cd74 + chondrocytes. (B) Feature plots displaying the expression of key marker genes (Fos, Lyz2, Tiam2, Cd74, Col11a2, and Ibsp) across different chondrocyte clusters. (C) Hierarchical clustering and correlation heatmap showing gene expression profiles across chondrocyte subtypes. (D) Heatmap of differentially expressed genes (DEGs) among chondrocyte subpopulations. (E) Heatmap illustrating the distribution of chondrocyte subpopulations between the Model and Si-A/PUE@HA groups. (F) GO (Gene Ontology) and KEGG pathway enrichment analysis of DEGs in different chondrocyte clusters, highlighting pathways involved in extracellular matrix organization, wound healing, and cartilage development. (G) Predicted ordering of chondrocyte subtypes during differentiation. (H) Pseudotime trajectory analysis revealing the dynamic progression of chondrocyte differentiation and gene expression changes over time. (I) Cell-cell interaction networks depicting communication between chondrocyte subtypes and other cell populations in the Model and Si-A/PUE@HA groups Si-A/PUE@HA composite hydrogel promotes cartilage regeneration by activating the PI3K-AKT pathway To investigate the mechanism underlying the cartilage repair promoted by the Si-A/PUE@HA hydrogel, we evaluated inflammatory markers, chondrogenic proteins, and the activation of the PI3K-AKT pathway in the regenerated cartilage. Western blot analysis of cartilage tissue extracts demonstrated a significant reduction in the pro-inflammatory markers iNOS and CD86 in the Si-A/PUE@HA group compared to the Model group, along with increased expression of CD163, a key M2 macrophage marker, indicating a shift toward an anti-inflammatory microenvironment (Fig. [168]10A, B). Immunofluorescence staining of the M1 marker CD86 showed a significant reduction in pro-inflammatory macrophage infiltration in the Si-A/PUE@HA group at both 6 and 12 weeks compared to the Model and NC@HA groups (Fig. [169]10C, D), with quantitative analysis confirming this decrease (Supplementary Fig. [170]6 A, B). Additionally, Si-A/PUE@HA hydrogel treatment enhanced cartilage repair by upregulating the expression of critical chondrogenic markers. Western blot analysis revealed increased levels of SOX9, Col2a1 (type II collagen), and Aggrecan in the Si-A/PUE@HA group, reflecting improved chondrocyte differentiation and extracellular matrix synthesis. Simultaneously, the expression of cartilage matrix-degrading enzymes MMP-13 and Adamts-5 was significantly reduced, suggesting protection against cartilage degradation (Fig. [171]10E, F). Immunofluorescence staining confirmed enhanced expression of ACAN and SOX9 at both 6 and 12 weeks post-implantation, further demonstrating the hydrogel’s role in promoting cartilage matrix formation (Fig. [172]10G, H, Supplementary Fig. [173]7 A, B). To elucidate the molecular mechanism driving these effects, we examined the activation of the PI3K-AKT signaling pathway. Western blot analyses showed upregulation of PI3K, phosphorylated AKT (p-AKT), and mTOR in the Si-A/PUE@HA group compared to the Model group (Fig. [174]10I, J), indicating activation of this pathway. Given the well-established role of the PI3K-AKT pathway in regulating cell proliferation, survival, and chondrogenesis, these findings suggest that the Si-A/PUE@HA hydrogel promotes cartilage regeneration by modulating inflammation and activating PI3K-AKT signaling, thereby enhancing chondrogenic differentiation and matrix production while mitigating cartilage degradation. Fig. 10. [175]Fig. 10 [176]Open in a new tab Si-A/PUE@HA composite hydrogel promotes cartilage regeneration by activating the PI3K-AKT pathway. (A) Western blot analysis of inflammatory markers in cartilage tissue. The expression of pro-inflammatory markers iNOS and CD86 was significantly reduced, while the M2 macrophage marker CD163 was increased in the Si-A/PUE@HA group compared to the Model group. (B) Quantification of relative protein expression levels of iNOS, CD86, and CD163, demonstrating a shift toward an anti-inflammatory microenvironment in the Si-A/PUE@HA group (*p < 0.05, **p < 0.01). (C) Representative images showing CD86 expression in cartilage tissue at 6 weeks post-implantation. CD86-positive areas (red) were reduced in the Si-A/PUE@HA group compared to the Model group. (D) Immunofluorescence images of CD86 at 12 weeks post-implantation, further confirming decreased CD86 expression in the Si-A/PUE@HA group compared to the Control, Model, and NC@HA groups. Scale bars: 200 μm. (E) Western blot analysis of key chondrogenic proteins (SOX9, Col2a1, Aggrecan) and cartilage-degrading enzymes (MMP-13, Adamts-5). The Si-A/PUE@HA group exhibited increased SOX9, Col2a1, and Aggrecan expression, along with decreased MMP-13 and Adamts-5 levels. (F) Quantification of protein expression levels, confirming significant upregulation of chondrogenic markers and suppression of cartilage degradation enzymes in the Si-A/PUE@HA group (*p < 0.05, **p < 0.01). (G) ACAN expression (green) at 6 and 12 weeks post-implantation, demonstrating increased cartilage matrix formation in the Si-A/PUE@HA group compared to the Model and NC@HA groups. (H) SOX9 expression (green) at 6 and 12 weeks post-implantation, confirming enhanced chondrogenic differentiation in the Si-A/PUE@HA group. Scale bars: 200 μm. (I) Western blot analysis of PI3K, phosphorylated AKT (p-AKT), and mTOR in regenerated cartilage. The Si-A/PUE@HA group exhibited increased expression of PI3K, p-AKT, and mTOR compared to the Model group. (J) Quantification of protein expression levels, confirming significant activation of the PI3K-AKT signaling pathway in the Si-A/PUE@HA group (*p < 0.05, ***p < 0.001) In vivo biocompatibility of Si-A/PUE@HA To evaluate the long-term in vivo biocompatibility of the Si-A/PUE@HA hydrogel, major organs—including the heart, liver, spleen, lungs, and kidneys—were examined 12 weeks post-implantation. Hematoxylin and eosin (HE) staining revealed no histopathological abnormalities or structural damage in any of the examined organs across all groups, indicating the absence of tissue inflammation or necrosis (Supplementary Fig. 8 A). Furthermore, Masson’s trichrome staining confirmed no significant collagen deposition or fibrosis in the tissues, further supporting the hydrogel’s non-fibrotic and biocompatible nature (Supplementary Fig. 8B). Immunofluorescence staining for F4/80 (a macrophage marker) and α-SMA (a fibrosis marker) showed minimal immune cell infiltration and no significant fibrotic response in the Si-A/PUE@HA group, comparable to the control group (Supplementary Fig. 8 C, D). These findings collectively demonstrate that the Si-A/PUE@HA hydrogel exhibits excellent in vivo biocompatibility, causing no adverse immune reactions or organ toxicity after long-term implantation. Discussion Osteochondral defects, characterized by damage to both the cartilage and underlying bone, represent a significant challenge in orthopedic medicine. These injuries can arise from trauma, degenerative diseases, or conditions such as osteochondritis dissecans, often leading to debilitating joint pain and reduced mobility. The natural repair capacity of cartilage is limited due to its avascular nature, which compounds the difficulty of achieving effective healing [[177]31]. Consequently, untreated osteochondral defects frequently progress to osteoarthritis, a leading cause of disability worldwide, underscoring the urgent need for optimized therapeutic strategies to restore cartilage function and joint integrity. In this study, we investigated the potential of Si-A/PUE@HA composite hydrogels as a novel scaffold for cartilage tissue engineering. By employing a multi-step synthesis approach, we developed hydrogels that exhibit excellent biocompatibility and mechanical properties conducive to supporting cell proliferation and differentiation. Our research aimed to assess the efficacy of these hydrogels in promoting chondrogenic differentiation of stem cells and facilitating cartilage repair in vivo. The findings from this study demonstrate that Si-A/PUE@HA hydrogels not only enhance cellular activity but also modulate the inflammatory response, thereby contributing to improved healing outcomes in osteochondral defects [[178]32, [179]33]. The Si-A/PUE@HA composite hydrogel has demonstrated significant promise as a bioactive material for cartilage tissue engineering. Its biocompatibility is evidenced by high cell viability and proliferation rates, indicating that it provides a supportive environment for the growth of bone marrow mesenchymal stem cells (BMSCs) which are crucial for cartilage repair. Compared to other hydrogels, such as those derived from chitosan or gelatin, Si-A/PUE@HA hydrogels show superior properties in terms of mechanical strength and elasticity, allowing for better integration and function within the joint environment [[180]34]. The mechanisms underlying the hydrogel’s cytocompatibility likely involve its unique structural composition, which promotes cell adhesion and minimizes inflammatory responses, a critical factor in regenerative medicine [[181]35]. In terms of promoting chondrogenic differentiation, Si-A/PUE@HA hydrogels significantly enhance the expression of key chondrogenic markers such as Acan, Sox9, and Col2a1. This is particularly relevant for addressing cartilage degeneration, as previous studies have identified that the effective promotion of chondrogenesis is vital for restoring cartilage integrity and function in conditions like osteoarthritis [[182]36]. The mechanical properties of hydrogels also play a pivotal role; hydrogels with tunable stiffness and elasticity have been shown to influence stem cell fate, guiding differentiation towards chondrocytes rather than fibrotic pathways [[183]37, [184]38]. Thus, the ability of Si-A/PUE@HA hydrogels to optimize these properties positions them as advanced candidates for clinical applications in cartilage repair. The immunomodulatory properties of Si-A/PUE@HA hydrogels are noteworthy, as they effectively modulate macrophage polarization. This hydrogel reduces pro-inflammatory M1 macrophage presence while promoting the anti-inflammatory M2 phenotype, an essential mechanism for a favorable healing environment [[185]39]. Such polarization has been associated with improved outcomes in tissue engineering, as a balanced inflammatory response can enhance tissue integration and regeneration. The significance of this finding underscores the potential of Si-A/PUE@HA hydrogels not only as scaffolds for cell growth but also as active agents in managing inflammation during the healing process. In vivo studies further support the viability of Si-A/PUE@HA hydrogels for cartilage regeneration, with micro-CT analyses revealing significant new tissue formation and integration with surrounding cartilage. This aligns with findings from other research indicating that hydrogels can enhance cartilage repair by providing a structurally and biochemically favorable environment for tissue growth and integration [[186]40]. The reduction in defect area observed in treated groups illustrates the hydrogel’s effectiveness in promoting regeneration, which is critical for translating these findings into clinical practice for managing osteochondral defects. Long-term biocompatibility studies of the Si-A/PUE@HA hydrogel also indicate its safety for clinical applications, with minimal immune response and no significant histopathological abnormalities noted in major organs after prolonged exposure. This is crucial for any biomaterial intended for implantation, as it highlights the hydrogel’s potential to minimize adverse effects and enhance patient safety [[187]41]. Such positive outcomes from both in vitro and in vivo assessments point towards the hydrogel’s readiness for further evaluation in larger clinical trials aimed at assessing its efficacy in human patients. Overall, the findings from this study substantiate the Si-A/PUE@HA composite hydrogel as a transformative biomaterial in the field of cartilage repair and regenerative medicine. The limitations of this study warrant careful consideration in the context of its findings. Notably, the relatively small sample size may hinder the generalizability of the results, and the absence of clinical validation restricts the applicability of the Si-A/PUE@HA hydrogel in real-world scenarios. Additionally, potential batch effects in the data analysis could introduce variability, leading to discrepancies in the interpretation of the hydrogel’s performance across different experiments. Future investigations should aim to address these limitations by incorporating larger cohorts and clinical trials to substantiate the efficacy and safety of this innovative biomaterial in cartilage repair applications. Furthermore, this study did not directly evaluate the in vitro or in vivo degradation kinetics of the hydrogel, which is an important factor influencing long-term scaffold integration and tissue remodeling. While the inclusion of biodegradable components like hyaluronic acid and puerarin suggests favorable breakdown behavior, future research should incorporate systematic biodegradability assessments under physiological conditions. Future investigations should aim to address these limitations by incorporating larger cohorts and clinical trials to substantiate the efficacy and safety of this innovative biomaterial in cartilage repair applications. It is also important to acknowledge the limitations associated with the use of a rat model for in vivo cartilage repair evaluation. While rats offer advantages such as low cost, reproducibility, and suitability for early-stage biomaterial screening, their articular cartilage is significantly thinner and structurally different from that of larger animals and humans. These differences may limit the translational relevance of observed regenerative outcomes. Furthermore, the rat joint environment does not fully recapitulate the load-bearing and biomechanical complexity of human knees. Future studies should incorporate larger animal models, such as rabbits or goats, which possess thicker cartilage and more closely mimic human joint physiology, to validate the therapeutic efficacy and long-term integration of the Si-A/PUE@HA hydrogel. In summary, the Si-A/PUE@HA hydrogel demonstrates significant promise as a biomaterial for cartilage regeneration, showcasing robust cytocompatibility, enhanced chondrogenic differentiation, and effective immunomodulatory properties. The in vivo results further indicate its potential for substantial cartilage repair, supporting its viability for clinical translation in regenerative medicine. These findings pave the way for future research focused on optimizing the hydrogel formulation and validating its therapeutic benefits in larger clinical populations, ultimately contributing to improved outcomes for patients suffering from osteochondral defects. Supplementary Information [188]Supplementary Material 1^ (2.1MB, docx) [189]Supplementary Material 2^ (19.9MB, mp4) Acknowledgements