Abstract Abnormal accumulation of both intracellular and extracellular free nucleic acids drives chronic inflammation in intervertebral disc degeneration (IVDD). Despite the development of numerous minimally invasive treatments for IVDD, systematic approaches targeting the chronic inflammation mediated by both nucleic acid types are lacking. We propose a dual clearance strategy that inhibits mitochondrial DNA release inside nucleus pulposus cells while removing extracellular DNA from the disc microenvironment. Using single-cell sequencing and clinical samples, we revealed how both nucleic acid types drive inflammation. We then developed a targeted nanovesicle system delivering a mitochondrial membrane-stabilizing small molecule to block DNA leakage and inflammatory signaling, along with a hydrogel that captures extracellular DNA to prevent immune activation. In a rat model, this approach significantly slowed disease progression. This targeted dual nucleic acid clearance strategy provides a approach for treating IVDD and offers a theoretical framework for addressing other nucleic acid-related inflammatory diseases. Subject terms: Nanoparticles, Materials science, Biomaterials __________________________________________________________________ Abnormal accumulation of intra- and extracellular free nucleic acids contributes to the chronic inflammation driving intervertebral disc degeneration (IVDD). Here, the authors report a nanovesicle system to inhibit mitochondrial DNA release in nucleus pulposus cells and remove extracellular DNA from the disc microenvironment to block inflammation for treatment of IVDD. Introduction Low back pain is a global health issue that severely impacts patients’ quality of life and imposes a significant burden on society and the economy^[44]1. IVDD is a leading cause of low back pain, especially in the elderly^[45]2. Current clinical treatments for IVDD—such as nonsteroidal anti-inflammatory drugs (NSAIDs), physiotherapy, and surgical intervention—are largely palliative and fail to halt or reverse disc degeneration^[46]3. Although experimental approaches, including stem cell therapy, gene editing, and biomaterial implants, have shown promise, they primarily aim to repair damaged tissue or suppress inflammation, often overlooking the upstream pathogenic drivers^[47]4–[48]6. In particular, the role of senescent nucleus pulposus (NP) cells and the inflammation they trigger via nucleic acid release remains underexplored. These senescent cells release mitochondrial DNA (mtDNA) into the cytosol and extracellular DNA (exDNA) into the extracellular matrix, activating the cGAS–STING and TLR9 pathways, respectively, to perpetuate sterile inflammation^[49]7,[50]8. Targeting these damage-associated nucleic acid signaling axes thus represents a mechanistically grounded and previously underutilized strategy for treating IVDD. Recent advances in biomimetic nanotechnology have highlighted the potential of drug-loaded cell membrane-derived vesicles as effective platforms for targeted intracellular delivery^[51]9,[52]10. Derived from natural cell membranes, these vesicles inherit bioactive properties such as immune evasion, prolonged circulation time, and homologous cell targeting. Incorporating therapeutic drugs into these vesicles improves drug stability and enables efficient delivery to targeted disease sites, facilitating precision treatment^[53]11. Their inherent biocompatibility and ability to mimic native cell interactions make these vesicles ideal platforms for the targeted modulation of cytosolic nucleic acid-driven inflammatory responses^[54]12,[55]13. Hyaluronic acid (HA) hydrogel, with a composition similar to the extracellular matrix (ECM), exhibits good biocompatibility and excellent drug-loading capacity, allowing sustained release of nanodrugs. It holds great potential for treating IVDD^[56]14,[57]15. Additionally, HA can be easily modified, with its surface functionalized by specific chemical groups or biological ligands to facilitate gel crosslinking reactions^[58]16. Polyamidoamine (PAMAM) dendrimers is a macromolecular network that contains numerous primary amine groups on its surface^[59]17. At physiological pH, these primary amines become protonated, forming positively charged -NH3^+ groups that easily capture negatively charged exDNA^[60]17. This functional hybrid system provides a means to neutralize exDNA and mitigate downstream immune activation. Here, we developed a biomimetic nanovesicle system (BNPMT) derived from NP cell membranes and loaded with BAI1, a small-molecule inhibitor of BAX/BAK-mediated mitochondrial membrane permeabilization (Fig. [61]1). To confer mitochondrial targeting capability, TPP^+ was conjugated to the vesicle surface via copper-free click chemistry. This dual-targeting strategy combines the inherent NP cell-homing ability of the membrane with the mitochondrial affinity of TPP^+, enabling efficient delivery of BAI1 to the NP cell mitochondria. At the target site, BAI1 stabilizes the mitochondrial outer membrane, thereby suppressing the aberrant release of mtDNA into the cytosol and subsequently blocking the production of senescence-associated secretory phenotype (SASP). Metabolomic profiling and mechanistic analyses further revealed that BNPMT stabilizes mitochondrial membrane integrity and restores mitochondrial function and energy metabolism in senescent NP cells. To achieve sustained local delivery and enhance therapeutic efficacy within the intervertebral disc, BNPMT was encapsulated in a dynamic hydrogel matrix crosslinked via Schiff base bonds between oxidized hyaluronic acid and fifth-generation PAMAM dendrimers (Fig. [62]1). Beyond its role as a delivery matrix, the PAMAM hydrogel also acts as a scavenger for exDNA, attenuating TLR9-mediated macrophage activation and downstream inflammatory amplification. Together, this dual-modality platform—simultaneously targeting intracellular mtDNA release and exDNA accumulation—offers effective therapeutic strategy for IVDD. This is the integrated approach that intervenes at both intracellular and extracellular levels of nucleic acid-mediated inflammation. In a rat model of IVDD, BNPMT treatment significantly alleviated disc degeneration and inflammation over an 8-week period, supporting its translational potential for long-term, mechanism-oriented disc repair. Fig. 1. Schematic overview of the hydrogel-nanocomposite design and its mechanism in regulating intra-/extracellular nucleic acids to alleviate intervertebral disc degeneration. [63]Fig. 1 [64]Open in a new tab The biomimetic nanovesicles are constructed by encapsulating BAI1 with NP cell membranes modified with TPP. Benefiting from the homotypic targeting ability of NP cell membranes and the mitochondrial targeting of TPP, the nanovesicles are precisely delivered to the mitochondria of NP cells. BAI1 inhibits the activation of BAX/BAK, thereby suppressing miMOMP, blocking the release of mtDNA into the cytoplasm, and subsequently inhibiting the activation of the cGAS-STING signaling pathway-mediated secretion of SASP factors. The hydrogel system designed for capturing exDNA is cross-linked via Schiff base bonds between oxidized hyaluronic acid (OHA) and PAMAM-G5. The primary amine groups of PAMAM-G5 become protonated to form positively charged -NH3+ under physiological pH, enabling the efficient scavenging of exDNA in the intervertebral disc microenvironment. This prevents the activation of macrophage M1 polarization via the exDNA–TLR9–TRAF6–NF-κB axis and helps improve the inflammatory microenvironment within the disc. In summary, by eliminating both intracellular and extracellular DNA in NP cells, the hydrogel-nanocomposite system effectively remodels the inflammatory microenvironment of the intervertebral disc, thereby delaying disc degeneration. Results NP cell senescence in IVDD: inflammatory activation by cytosolic nucleic acids and extracellular DNA NP degeneration is a major contributor and primary driver of IVDD. Multiple factors—including cellular senescence, apoptosis, mechanical overload, inflammation, and oxidative stress—contribute to NP degeneration, suggesting that its prevention may offer an effective therapeutic strategy for IVDD^[65]18. To further investigate the link between nucleus pulposus degeneration and intervertebral disc degeneration, this study reanalyzed single-cell transcriptomic data from the publicly available [66]GSE165722 database and examined the role of nucleus pulposus cells in this process (Fig. [67]2A). Based on characteristic gene expression, cells from healthy and degenerated intervertebral discs were categorized into 14 groups (Fig. [68]2B, C). The 14 cell groups were further classified into four transcriptionally distinct clusters (Fig. [69]2D). The volcano plot indicates significant upregulation of senescence-related genes (e.g., p21, Tp53) in nucleus pulposus cells from degenerated intervertebral discs (Fig. [70]2E). Gene Ontology (GO) enrichment analysis of differentially expressed genes (DEGs) highlighted significant enrichment in pathways related to mitochondrial function, energy metabolism, and inflammation (Fig. [71]2F). Notably, mitochondrial function, energy metabolism, and inflammation are closely interconnected in senescent cells, and their imbalance collectively accelerates cellular senescence^[72]19,[73]20. In senescent cells, mitochondrial function declines, energy production decreases, and self-repair and stress responses weaken. Meanwhile, ROS accumulates, damaging DNA, proteins, and lipids, ultimately impairing normal cell function^[74]21,[75]22. Gene set enrichment analysis (GSEA) revealed significant upregulation of inflammatory response pathways and downregulation of senescence-inhibition pathways in degenerated intervertebral discs, suggesting a strong association between intervertebral disc degeneration and nucleus pulposus cell senescence (Fig. [76]2G, H). To complement these findings, we characterized the transcriptomic landscapes of young and senescent NP cells in vitro. Rat NP cells were isolated and induced into senescence using doxorubicin (Dox), followed by RNA sequencing (Fig. [77]2I). Principal component analysis (PCA) demonstrated distinct transcriptomic profiles between NC and senescent NP cells (Supplementary Fig. [78]1). Senescent cells showed significant upregulation of genes related to the SASP and downregulation of ECM synthesis genes (Fig. [79]2J). GO enrichment analysis revealed marked enrichment in inflammatory and immune activation pathways, alongside suppression of mitochondrial functional pathways (Fig. [80]2K). GSEA also confirmed elevated immune and inflammatory activity in senescent NP cells compared to normal controls (Supplementary Fig. [81]2). Collectively, these results suggest that inflammatory senescence pathways are activated in NP cells during IVDD. However, the precise mechanisms driving NP cell senescence remain incompletely understood. Under stress conditions, mitochondrial dysfunction may induce minority mitochondrial outer membrane permeabilization (miMOMP), leading to cytosolic leakage of mtDNA and subsequent activation of innate immune responses^[82]23. To test this hypothesis, we extracted the cytoplasmic fraction of normal and senescent NP cells and quantified cytosolic mtDNA via quantitative PCR (qPCR). Senescent NP cells exhibited significant mtDNA accumulation in the cytoplasm (Fig. [83]2L and Supplementary Fig. [84]3). Moreover, histological analyses of human NP tissues from different Pfirrmann grades (Fig. [85]2M) and rat disc samples revealed markedly elevated expression of STING in degenerated discs (Fig. [86]2N and Supplementary Fig. [87]4), implicating activation of the cGAS-STING pathway through cytosolic DNA sensing. In addition to intracellular responses, senescent cells may release mtDNA into the extracellular microenvironment^[88]7, where it can activate macrophages via Toll-like receptors (TLRs), thereby amplifying inflammation. While cytosolic DNA activates inflammation through the cGAS-STING axis, extracellular nucleic acids contribute to immune activation via TLR-mediated pathways^[89]24,[90]25. To assess the relevance of extracellular nucleic acids in IVDD, we isolated NP tissues from healthy and degenerated rats and measured the DNA content in the microenvironment following cell isolation. Degenerated discs exhibited significantly higher exDNA levels than healthy discs (Fig. [91]2O), further supporting the involvement of both intracellular and extracellular nucleic acids in IVDD pathology. In summary, these findings uncover a pathological axis in IVDD, wherein the aberrant accumulation of nucleic acids—both intracellular and extracellular—triggers innate immune activation and drives disease progression. Fig. 2. IVDD is linked to inflammation caused by the accumulation of intracellular and extracellular nucleic acids. [92]Fig. 2 [93]Open in a new tab A Schematic of single-cell RNA-seq in human degenerated disc tissues. B UMAP plot of single-cell transcriptomes from human discs. C Bubble plot of scaled expression of DEGs across clusters. D UMAP plot of human intervertebral disc cells, showing four distinct clusters identified via unsupervised clustering. Each dot represents a single cell, color-coded by subcluster classification. E Volcano plot showing differentially expressed genes in nucleus pulposus cells. F Gene Ontology (GO) analysis was performed to investigate the differentially expressed genes (DEGs) in nucleus pulposus cells. GO enrichment analysis was performed using a hypergeometric distribution algorithm to assess the significance of differentially expressed gene enrichment in each GO term. Fisher’s exact test was applied separately to the Cellular Component. P-values were adjusted for multiple comparisons (FDR < 0.05). G, H GSEA plots showing enrichment analysis for nucleus pulposus cells. GSEA was performed using the default weighted Kolmogorov–Smirnov statistic with FDR < 0.25 as the significance threshold. I Schematic of bulk RNA-seq in rat primary NP cells. J Heatmap of DEGs in rat NP cells (p value < 0.05, |log2FoldChange| >1). K GO enrichment analysis plot for rat nucleus pulposus cells. GO enrichment analysis was performed using a hypergeometric distribution algorithm to assess the significance of differentially expressed gene enrichment in each GO term. Fisher’s exact test was applied separately to the Biological Process, Cellular Component, and Molecular Function categories. P-values were adjusted for multiple comparisons (FDR < 0.05). L Cytoplasmic DNA content in normal vs. senescent rat NP cells (n = 3 biological replicates, mean ± s.d.). M MRI scans of human intervertebral discs from normal and degenerated groups. N Immunohistochemical staining of STING in rat and human NP tissues. Scale bar, 50 μm. Representative images are from three independent experiments. O exDNA content in control vs. degenerated discs (n = 3 biological replicates, mean ± s.d.). Created with BioRender.com (A, I). Statistical significance was assessed using a two-tailed unpaired Student’s t-test. (L, O). Source data are provided as a Source Data file. Development of mitochondrial-targeted nanovesicles (BNPMT) to prevent mtDNA leakage in NP cells As nucleus pulposus cells age, mitochondrial dysfunction occurs, potentially causing mtDNA leakage, which activates the innate immune pathway and triggers inflammation. Therefore, we initially aimed to develop a targeted nanomedicine to prevent mtDNA leakage from mitochondria. BAX-mediated miMOMP is a key driver of cytosolic mtDNA leakage^[94]26. To target this mechanism, we engineered a biomimetic nanovesicle to deliver the BAX inhibitor BAI1, which inhibits miMOMP, prevents abnormal cytosolic DNA accumulation in nucleus pulposus cells, and suppresses SASP factor release (Fig. [95]3A, B). To achieve mitochondrial targeting in nucleus pulposus cells, we modified the nucleus pulposus cell membrane surface with the TPP ligand^[96]27. First, we co-incubated nucleus pulposus cells with Ac4ManNAz, successfully introducing azide groups onto their surface^[97]28. Next, we employed copper-free click chemistry to attach the TPP functional group to the cell surface. To evaluate the efficiency of the click chemistry reaction, we used DBCO-FITC, and fluorescence analysis confirmed the successful attachment of FITC to the nucleus pulposus cell membrane (Fig. [98]3C). Fig. 3. Characterization and targeting effect of BNPMT. [99]Fig. 3 [100]Open in a new tab A Schematic representation of BNPMT synthesis. B 3D representation of BAI1 binding to the BAX protein. C Evaluation of click chemistry efficiency on nucleus pulposus cell surfaces. Scale bar, 50 μm. Representative images are from three independent experiments. D Western blot analysis of Na^+/K^+-ATPase and α-tubulin in nucleus pulposus cell membranes and cytoplasm. Representative images are from three independent experiments. E Average particle sizes of BNPM and BNPMT (n = 6 independent experiments, mean ± s.d.). F Cryo-electron microscopy images of BNPMT (Scale bar, 50 nm) and particle size distribution of BNPM and BNPMT. G Zeta potentials of BNPM and BNPMT (n = 3 independent experiments, mean ± s.d.). H Encapsulation efficiency of BNPMT (n = 3 independent experiments, mean ± s.d.). I Uptake of FITC, FITC@NPM, and FITC@NPMT by RAW264.7 macrophages and NP cells (Scale bar, 50 μm, left). Representative images are from three independent experiments. Statistical analysis of FITC fluorescence intensity in RAW264.7 and nucleus pulposus cells (n = 3 independent experiments, mean ± s.d., right). J Visualization of nanovesicle localization after 4h co-incubation of FITC, FITC@NPM, and FITC@NPMT with nucleus pulposus cells (Scale bar, 10 μm). Representative images are from three independent experiments. K Intensity analysis along the selected white line across the entire cell in (J). L Schematic representation of BNPMT nanovesicles targeting mitochondria to inhibit BAX activation. Created with BioRender.com (L). Statistical analyses were performed using one-way ANOVA followed by Tukey’s multiple comparison test (I). Source data are provided as a Source Data file. We extracted and purified the nucleus pulposus cell membrane, and Western blot (WB) analysis confirmed the successful isolation of NP cell membranes (NPM) (Fig. [101]3D). We then fabricated engineered biomimetic nanovesicles using mechanical extrusion and loaded BAI1 into the vesicles via ultrasonic drug loading, yielding BNPM and TPP-modified BNPMT^[102]29,[103]30. Cryo-electron microscopy confirmed that BNPMT exhibited a typical vesicular morphology. Dynamic light scattering (DLS) analysis showed that BNPM had an average hydrodynamic diameter of ~150 nm with a zeta potential of −16.7 mV, while BNPMT exhibited a larger diameter (~170 nm) and a zeta potential of −14.5 mV (Fig. [104]3E–G). The increased vesicle size and altered surface charge may result from TPP modification. High-performance liquid chromatography (HPLC) analysis determined the drug loading rate of BAI1 to be 17.8% (Fig. [105]3H). The membrane-derived nanovesicles exhibited homotypic targeting ability^[106]31. Incubation of nanovesicles with different cell types revealed that those derived from nucleus pulposus cell membranes exhibited significantly higher fluorescence in nucleus pulposus cells than in macrophages, indicating specific uptake of FITC@NPMT and FITC@NPM by nucleus pulposus cells (Fig. [107]3I). Furthermore, we analyzed the mitochondrial targeting ability of FITC@NPMT. Unlike FITC and FITC@NPM, which primarily localized in the cytoplasm of nucleus pulposus cells, TPP-modified FITC@NPMT showed strong mitochondrial co-localization, confirming its mitochondrial targeting properties (Fig. [108]3J–L). BNPMT prevents mtDNA leakage, thereby mitigating inflammation in aging NP cells Next, we investigated the intracellular anti-inflammatory effects of BNPMT. First, we established an in vitro aging model of nucleus pulposus cells using Dox. Following Dox treatment, BAX(6A7) expression in aging nucleus pulposus cells significantly increased and co-localized with mitochondria, accompanied by a marked rise in cytosolic double-stranded DNA (dsDNA) levels (Fig. [109]4A, B). BNPMT treatment significantly reduced BAX(6A7) expression and cytosolic dsDNA levels in aging nucleus pulposus cells, suggesting that BNPMT effectively inhibited BAX activation, suppressed miMOMP, and reduced mtDNA leakage into the cytoplasm (Fig. [110]4A, B). Furthermore, cytoplasmic fractionation and dsDNA quantification confirmed that BNPMT significantly reduced cytoplasmic dsDNA accumulation (Fig. [111]4C). In conclusion, these findings demonstrate that BNPMT inhibits miMOMP and prevents mtDNA leakage in aging nucleus pulposus cells. The cGAS-STING pathway serves as a key cytosolic DNA sensor, recognizing dsDNA and activating the downstream NF-κB signaling cascade, which promotes the secretion of inflammatory cytokines (e.g., TNF-α and IL-6)^[112]32. To determine whether BNPMT suppresses SASP secretion through the cGAS–STING pathway following the inhibition of cytosolic mtDNA accumulation, we analyzed the expression of key proteins associated with the cGAS–STING signaling pathway across different treatment groups. WB analysis revealed significantly elevated phosphorylation levels of p-STING, p-IRF3, and p-TBK in aging nucleus pulposus cells, whereas BNPMT treatment effectively suppressed cGAS-STING pathway activation (Fig. [113]4D and Supplementary Fig. [114]5). Additionally, qPCR and ELISA analyses demonstrated that BNPMT treatment significantly reduced IL-6 and TNF-α mRNA levels in aging nucleus pulposus cells, along with a marked decrease in IL-6 secretion (Fig. [115]4E, F). These findings suggest that BNPMT mitigates SASP by suppressing the cytosolic DNA sensing pathway cGAS-STING. Fig. 4. BNPMT suppress SASP secretion in NP cells. [116]Fig. 4 [117]Open in a new tab A, B Immunofluorescence staining and quantification of BAX6A7 (A) and dsDNA (B) in aging NP cells treated with BAI1, BNPM, or BNPMT. Scale bars, 10 μm. Representative images are from three independent experiments. Quantification of BAX6A7 fluorescence intensity (n = 3 biological replicates, mean ± s.d.). Quantification of dsDNA fluorescence intensity (n = 3 biological replicates, mean ± s.d.). C Cytoplasmic dsDNA content in NP cells following different treatments (n = 3 biological replicates, mean ± s.d.). D Western blot analysis of STING pathway proteins. Representative images are from three independent experiments. E mRNA expression levels of IL-6 and TNF-α in NP cells from different treatment groups (n = 9 three independent biological replicates with three technical replicates each, mean ± s.d.). F IL-6 cytokine levels in the supernatants of NP cells across different treatment groups (n = 3 biological replicates, mean ± s.d.). G Heatmap of differentially expressed genes in SEN vs. BNPMT groups. H, I KEGG enrichment analysis of upregulated and downregulated gene sets in the BNPMT and SEN groups. KEGG pathway enrichment was performed using a hypergeometric test with Benjamini–Hochberg correction. Pathways with adjusted P-values (FDR) < 0.05 were considered significantly enriched. J GSEA comparing the SEN and BNPMT groups for gene sets involved in the NF-κB, JAK-STAT, and IL-17 signaling pathways. GSEA was performed using the default weighted Kolmogorov–Smirnov statistic with FDR < 0.25 as the significance threshold. K Quantitative qPCR analysis of total mtDNA levels in senescent NP cells post-treatment with CNPMT or BNPMT (n = 3 biological replicates, mean ± s.d.). L ELISA quantification of IL-6 protein secretion in senescent NP cells treated with CNPMT or BNPMT (n = 3 biological replicates, mean ± s.d.). M, N Representative SA-β-gal staining images of senescent NP cells following treatment with CNPMT or BNPMT (N). Scale bar, 100 μm. Quantification of SA-β-gal-positive cells (M) (n = 3 biological replicates, mean ± s.d.). Statistical analyses were performed using one-way ANOVA followed by Tukey’s multiple comparison test (A–C, E, F, K–M). Source data are provided as a Source Data file. To further investigate the therapeutic effects of BNPMT on aging nucleus pulposus cells, we conducted transcriptome sequencing on aging nucleus pulposus cells with and without BNPMT treatment. PCA analysis demonstrated significant mRNA differences between aging nucleus pulposus cells and those treated with BNPMT (Supplementary Fig. [118]6). Differential gene expression (DEG) analysis using DESeq2 revealed that BNPMT treatment significantly downregulated SASP-related genes while upregulating ECM synthesis-related genes, consistent with previous findings (Fig. [119]4G). Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis indicated that, compared to aging nucleus pulposus cells, BNPMT treatment significantly downregulated pathways associated with cell aging and inflammation while upregulating those related to mitochondrial function (Fig. [120]4H, I). GSEA analysis further revealed that BNPMT treatment significantly suppressed inflammation-related pathways, including the NF-κB, JAK-STAT, and IL-17 signaling pathways (Fig. [121]4J). These findings confirm that BNPMT exerts an endogenous anti-inflammatory effect in aging nucleus pulposus cells by preventing cytosolic DNA accumulation and reversing the aging-related inflammatory phenotype. Furthermore, ECM homeostasis, primarily regulated by nucleus pulposus cells, is essential for maintaining normal disc function, and aging nucleus pulposus cells are often linked to an imbalance between ECM synthesis and degradation^[122]33. We analyzed ECM-related gene and protein expression in aging nucleus pulposus cells following BNPMT treatment using WB and qPCR. The results indicated that, compared to untreated aging nucleus pulposus cells, BNPMT treatment significantly increased the protein and mRNA expression of collagen II (COL2), a key ECM component, while also upregulating SOX9 protein expression (Supplementary Figs. [123]7 and [124]8). Meanwhile, the protein and mRNA expression of the ECM-degrading enzyme matrix metalloproteinase-13 (MMP13) was significantly downregulated (Supplementary Figs. [125]7–[126]9). These data further suggest that BNPMT not only suppresses aging-related inflammation but also restores ECM homeostasis by promoting functional repair in nucleus pulposus cells. To further investigate the differences between BNPMT and conventional anti-inflammatory drugs in alleviating NP cell senescence and apoptosis, we constructed biomimetic nanovesicles (CNPMT) by encapsulating curcumin within TPP-modified NP cell membranes. By isolating the cytoplasm of senescent NP cells and quantifying cytosolic dsDNA levels, we found that CNPMT exhibited limited ability to prevent cytosolic mtDNA accumulation compared to BNPMT (Fig. [127]4K and Supplementary Fig. [128]10). Subsequently, we evaluated the anti-inflammatory effects of BNPMT and CNPMT using qPCR and ELISA assays. The results demonstrated that BNPMT significantly outperformed CNPMT in suppressing IL-6 and TNF-α mRNA expression, as well as inhibiting IL-6 cytokine secretion in senescent NP cells (Fig. [129]4L and Supplementary Fig. [130]11), suggesting a superior anti-inflammatory capacity of BNPMT in this context. We then assessed the ability of BNPMT and CNPMT to attenuate NP cell senescence and apoptosis. SA-β-gal staining revealed that BNPMT more effectively reduced NP cell senescence (Fig. [131]4M, N). Furthermore, live/dead staining and flow cytometry analysis showed that BNPMT exhibited markedly enhanced anti-apoptotic effects compared to CNPMT (Supplementary Figs. [132]12 and [133]13). Finally, we investigated ECM homeostasis in NP cells after different treatments. qPCR analysis indicated that BNPMT significantly upregulated the mRNA expression of the ECM component COL2, while effectively downregulating the expression of the ECM-degrading enzyme MMP13 (Supplementary Fig. [134]14). Collectively, these results highlight that, compared with conventional anti-inflammatory drugs, BNPMT demonstrates superior efficacy in mitigating NP cell senescence and apoptosis, as well as in modulating ECM synthesis and degradation. BNPMT-induced metabolic reprogramming in senescent NP cells To further explore the mechanism of BNPMT intervention in senescent nucleus pulposus cells, we conducted metabolomics analysis and examined the metabolic profiles of the NC, SEN, and BNPMT groups using Partial Least Squares Discriminant Analysis. PCA revealed significant metabolic differences among the NC, SEN, and BNPMT groups (Fig. [135]5A). The metabolic profile of BNPMT-treated senescent NP cells closely resembled that of young cells, further confirming that BNPMT partially reverses the senescent phenotype of nucleus pulposus cells, suggesting that metabolic reprogramming may contribute to the functional restoration of senescent NP cells (Fig. [136]5B). BNPMT intervention induced significant metabolic reprogramming in nucleus pulposus cells compared to the SEN group, affecting metabolites such as pyruvate, alanine, glutamate, guanosine, inosine, and uracil (Fig. [137]5B). Pyruvate lies at the intersection of glycolysis and mitochondrial metabolism, and can promote NAD^+ regeneration through lactate production, thereby maintaining mitochondrial function and delaying cellular senescence^[138]34. Glutamate is a precursor of α-ketoglutarate and can enter the TCA cycle to support mitochondrial respiration; it also serves as a substrate for glutathione (GSH) synthesis, contributing to antioxidant defense^[139]35. KEGG pathway enrichment analysis of differentially expressed metabolites revealed that BNPMT primarily reverses metabolic alterations in senescent nucleus pulposus cells through multiple pathways, including alanine, aspartate, and glutamate metabolism; purine metabolism; pyrimidine metabolism; and the biosynthesis of pantothenate and Coenzyme A (Fig. [140]5C). Furthermore, metabolomics analysis allowed us to establish the metabolic profiles of nucleus pulposus cells under different interventions (Fig. [141]5D, E). The biosynthesis of pantothenate and Coenzyme A, along with purine and pyrimidine metabolism, plays a crucial role in regulating cellular energy metabolism, ensuring sufficient energy for basic metabolic and repair functions^[142]36. The biosynthesis of pantothenate and Coenzyme A, glutathione synthesis, and β-alanine metabolism are involved in cellular antioxidant and stress responses, aiding in the elimination of excess free radicals, reduction of oxidative damage, and deceleration of cellular aging^[143]37,[144]38. Additionally, pyrimidine and purine metabolism contribute to ECM synthesis and stability by supplying essential molecules for gene replication, transcription, and repair, thereby supporting the structural and functional integrity of the intervertebral disc^[145]39. To further verify the functional relevance of these metabolic alterations, we examined ATP levels in NP cells (Fig. [146]5F). BNPMT treatment significantly increased ATP production in senescent NP cells, indicating improved energy metabolism. We also assessed mitochondrial membrane potential and mitochondrial Reactive Oxygen Species (ROS) levels. BNPMT-treated NP cells showed restoration of mitochondrial membrane potential and a marked reduction in mitochondrial ROS generation, suggesting enhanced mitochondrial function (Fig. [147]5G and Supplementary Fig. [148]15). Collectively, these results, together with the metabolomic findings, suggest that BNPMT promotes metabolic reprogramming in senescent NP cells, leading to the restoration of mitochondrial function and attenuation of senescence-associated phenotypes. These findings underscore a potential mechanistic link between metabolic regulation and cellular aging in NP cells. Fig. 5. Metabolic alterations in nucleus pulposus cells following BNPMT treatment. [149]Fig. 5 [150]Open in a new tab A PCA plot showing the metabolic profiles of nucleus pulposus cells from the SEN, NC, and BNPMT groups (n = 6 biological replicates). B Heatmap illustrating the differential metabolites in nucleus pulposus cells among the SEN, NC, and BNPMT groups (n = 6 biological replicates). C KEGG pathway enrichment bubble plot depicting metabolite distribution in nucleus pulposus cells from the SEN, NC, and BNPMT groups. KEGG pathway enrichment was performed using a hypergeometric test with Benjamini–Hochberg correction. Pathways with adjusted P-values (FDR) < 0.05 were considered significantly enriched. D Metabolomics network analysis of the SEN, NC, and BNPMT groups. Yellow, blue, and black rectangular borders represent metabolites that increased, decreased, or remained unchanged in BNPMT compared to the SEN group. Brown, blue, and white-filled rectangles indicate metabolites that increased, decreased, or remained unchanged in the SEN group compared to the NC group. Dashed rectangles indicate undetected metabolites. E Schematic diagram illustrating differential metabolic pathways in nucleus pulposus cells between the SEN and BNPMT groups. F Quantification of intracellular ATP levels in NP cells following different treatments (n = 6 biological replicates, mean ± s.d.). G Representative TMRE staining images (Scale bar, 50 μm) and corresponding fluorescence intensity analysis (n = 3 biological replicates, mean ± s.d.) in NP cells from each group. Created with BioRender.com (E). Statistical analyses were performed using one-way ANOVA followed by Tukey’s multiple comparison test (F, G). Source data are provided as a Source Data file. Synthesis and characterization of H-P gel for capturing exDNA Next, we aim to develop a hydrogel system capable of capturing exDNA (Fig. [151]6A). The hydrogel is crosslinked via Schiff base chemistry between oxidized hyaluronic acid and PAMAM, where the positively charged PAMAM effectively captures the negatively charged exDNA. We evaluated the biocompatibility and gelation time of PAMAM dendrimers with different generations (Supplementary Figs. [152]16 and [153]17), and ultimately selected PAMAM-G5 due to its superior stability and biocompatibility, making it a suitable component for constructing the exDNA-capturing hydrogel system. As shown in Fig. [154]6B, when equal volumes of OHA aqueous solution and PAMAM solution are mixed at room temperature, a hydrogel (H-P gel) rapidly forms. Scanning electron microscopy (SEM) imaging of freeze-dried H-P gel reveals a typical porous microstructure, which is conducive to vesicle loading (Fig. [155]6C). Next, we evaluated the injectability and self-healing properties of the H-P gel. The hydrogel was continuously extruded through a 26-gauge needle, successfully forming the letters “NP” without blockage (Fig. [156]6D). Meanwhile, rheological analysis demonstrated shear-thinning behavior, as the viscosity of the H-P gel decreased sharply when the shear rate increased from 0.1 to 1000 s^−1, confirming its injectability (Fig. [157]6E). The self-healing ability of the H-P gel was further examined. In macroscopic tests, two separated pieces of H-P gel fused into a single, continuous gel after 10 min of contact (Fig. [158]6F). Additionally, strain sweep experiments indicated that the storage modulus (G′) and loss modulus (G″) remained nearly constant up to 30% strain, suggesting that the H-P gel can withstand relatively large elastic deformations. However, at 80% strain, the intersection of G′ and G″ marked the critical point at which the hydrogel network collapsed (Fig. [159]6G). To further assess self-healing behavior, continuous alternating strain scanning was conducted. Under a high dynamic strain (200%), the G′ value decreased below G″, indicating network disruption. When the strain was reduced to 1%, the hydrogel fully recovered its G′ and G″ values within seconds, maintaining this recovery over three alternating cycles, thus demonstrating excellent self-healing capability (Fig. [160]6H). The dynamic Schiff base reactions in the H-P gel occur rapidly at room or physiological temperatures. The aldehyde groups in OHA react with the amino groups in PAMAM to form Schiff base bonds, achieving a dynamic equilibrium that imparts both injectability and self-healing properties to the hydrogel. To enable in vivo tracking, indocyanine green (ICG) was covalently conjugated to the hydrogel using ICG-NHS, which forms stable amide bonds with the abundant primary amine groups (–NH[2]) on PAMAM dendrimers. In vivo fluorescence imaging was performed on days 0, 3, 7, 14, and 21 post injection to dynamically monitor the persistence and degradation of the hydrogel within the intervertebral disc space (Supplementary Fig. [161]18). The results demonstrated that the H-P gel exhibited a slow degradation profile, with detectable fluorescence signals remaining up to 21 days after injection. Fig. 6. Characterization and Function of H-P Hydrogel. [162]Fig. 6 [163]Open in a new tab A, B Schematic (A) and image (B) of H-P GEL formation from 1% PAMAM-G5 and 5% HA-CHO (1:1). Representative images are from three independent experiments. C SEM image of H-P GEL. Scale bar, 400 μm. Representative images are from three independent experiments. D Injectability of H-P GEL. E Viscosity curve demonstrating the shear-thinning behavior of the hydrogel as shear rates increase (0.1–1000 s^−1). F Images showing the self-healing ability of the hydrogel. G Storage (G′) and loss modulus (G′′) of the hydrogel in a strain amplitude sweep (γ = 1–1000%) at a fixed frequency. H Rheological behavior of the hydrogel under alternating strains (1–200%). I Schematic of RAW264.7 cell co-culture. J Flow cytometry of CpG uptake (left) and corresponding statistical graph (right) (n = 4 biological replicates, mean ± s.d.). K Flow cytometry of iNOS expression (left) and corresponding statistical graph (right) (n = 3 biological replicates, mean ± s.d.). L Expression levels of TLR9, TNF-α, and IL-6 mRNA in RAW264.7 cells (n = 9 three independent biological replicates with three technical replicates each, mean ± s.d.). M TNF-α and IL-6 (n = 5 biological replicates, mean ± s.d.) cytokine levels in RAW264.7 cell culture supernatant. N Diagram of conditioned medium preparation. O mRNA levels of COL2, ACAN in NP cells after treatment with different conditioned media (n = 9 three independent biological replicates with three technical replicates each, mean ± s.d.). P mRNA levels of MMP3 and MMP13 in NP cells after treatment with different conditioned media (n = 9, three independent biological replicates with three technical replicates each, mean ± s.d.). Q Western blot for COL2 and MMP3. Representative images are from three independent experiments. R, S Immunofluorescence staining of COL2 and MMP3 in NP cells. Scale bars, 100 μm. along with fluorescence intensity statistical graph (n = 4 biological replicates, mean ± s.d.). Created with BioRender.com (I, N). Statistical analyses were performed using one-way ANOVA followed by Tukey’s multiple comparison test (J–M, O, P, R, S). Source data are provided as a Source Data file. exDNA in the intervertebral disc environment activates macrophage TLRs, triggering local inflammation, accelerating nucleus pulposus cell aging, and disrupting the metabolic balance between ECM synthesis and degradation^[164]40. The primary amine groups on the surface of PAMAM can be protonated at physiological pH (pH = 7.4), forming positively charged-NH[3]^+ groups that effectively capture negatively charged exDNA (Fig. [165]6I). Flow cytometry analysis demonstrated that after co-incubation with H-P gel, the uptake of exDNA mimic CpG-FITC by macrophages was significantly reduced, confirming that H-P gel can sequester exDNA from the environment and inhibit CpG-FITC internalization by macrophages (Fig. [166]6J). Upon activation of TLRs on the macrophage surface, M1 polarization occurs, leading to the release of pro-inflammatory cytokines such as TNF-α, IL-6, and IL-1β, thereby initiating and exacerbating local inflammation. Flow cytometry results showed that after H-P gel captured environmental CpG, it significantly downregulated the expression of the M1 macrophage marker inducible nitric oxide synthase (iNOS) (Fig. [167]6K). Furthermore, qPCR and ELISA analyses demonstrated that co-incubation with H-P gel significantly reduced the mRNA expression levels of TLR9, IL-6, and TNF-α, as well as the secretion of the inflammatory cytokines IL-6 and TNF-α in macrophages (Fig. [168]6L, M). These findings indicate that H-P gel effectively suppresses exogenous exDNA-induced inflammation in the intervertebral disc. Chronic exposure of nucleus pulposus cells to an inflammatory microenvironment suppresses their function, disrupts normal metabolic processes, accelerates ECM degradation, and promotes disc degeneration. To investigate whether mitigating exogenous exDNA-induced inflammation could restore ECM homeostasis in nucleus pulposus cells, we collected conditioned media from CpG-treated macrophages and macrophages co-incubated with H-P gel. Nucleus pulposus cells were then cultured in these conditioned media for 24 h to assess ECM-related markers and determine whether alleviating macrophage inflammation could restore the balance between ECM synthesis and degradation (Fig. [169]6N). qPCR analysis revealed that the mRNA expression levels of key ECM components, COL2 and aggrecan (ACAN), were significantly upregulated, whereas the expression levels of ECM degradation enzymes matrix MMP13 and MMP3 were markedly downregulated (Fig. [170]6O, P). WB and immunofluorescence staining results also confirmed this (Fig. [171]6Q–S). Collectively, these results demonstrate that H-P gel effectively mitigates macrophage-driven inflammation, thereby restoring ECM synthesis and degradation homeostasis in nucleus pulposus cells. Therapeutic effect of BNPMT @ H-P in a puncture-induced rat intervertebral disc degeneration model To minimize potential side effects caused by off-target effects of BNPMT in vivo, we adopted a localized delivery strategy. BNPMT was encapsulated within an H-P gel, which confined its bioactivity to the lesion site and reduced systemic exposure. Moreover, the H-P gel enables the sustained release of BNPMT within the intervertebral disc, thereby enhancing its therapeutic efficacy. Subsequently, the therapeutic potential of BNPMT@H-P was evaluated using a puncture-induced rat model of IVDD (Fig. [172]7A). After 8 weeks of treatment, major organs and serum samples were collected for histological and biochemical analysis to assess the biosafety of BNPMT@H-P (Supplementary Figs. [173]19 and [174]20). H&E staining revealed no evident pathological changes in major organs, and serum biochemical parameters showed no significant differences compared with controls. These results indicate that BNPMT@H-P does not elicit systemic immune responses and possesses favorable biocompatibility. Given the near-infrared fluorescence properties of ICG, which offer superior tissue penetration and reduced background autofluorescence in vivo^[175]41, ICG enables more reliable and sensitive imaging outcomes. To simulate the in vivo release behavior of BNPMT, we encapsulated ICG within NP cell membrane–coated nanovesicles and further embedded these nanovesicles into H-P gel for subsequent tracking and imaging. ICG@NPMT and ICG@NPMT@H-P were administered into the rat IVD to mimic the in vivo release profile of BNPMT@H-P. In vivo imaging revealed that ICG@NPMT rapidly diffused and was cleared from the IVD site by day 7, whereas ICG@NPMT encapsulated in H-P gel persisted at the site for up to 21 days. This suggests that H-P gel can sustain a higher local drug concentration within the IVD for an extended duration (Fig. [176]7B, C). BNPMT, H-P gel, and BNPMT@H-P were injected into the rat IVD using a 26G needle. The control group underwent no puncture, while the injury group received no injection. The disc height index (DHI) of the five groups was assessed using radiographic analysis. At four weeks post-surgery, the DHI value in the BNPMT@H-P group was comparable to that of the control group but significantly higher than in the BNPMT, H-P, and non-injection groups. At eight weeks post-surgery, BNPMT@H-P significantly attenuated the puncture-induced reduction in IVD height, whereas BNPMT or H-P treatment alone had a minimal effect (Fig. [177]7D, E). MRI analysis, which assesses IVD water content and serves as a key diagnostic indicator of IVD degeneration, revealed uneven disc signals in the IVDD, BNPMT groups, and H-P groups, suggesting reduced water content^[178]42. These findings were consistent with radiographic assessments, as DHI and MRI results were aligned, further supporting the potential of BNPMT@H-P in delaying IVD degeneration in vivo (Fig. [179]7F, G). The therapeutic efficacy of minimally invasive BNPMT@H-P hydrogel injection in inhibiting IVD degeneration was validated through histological and immunohistochemical analyses at 4 and 8 weeks post-surgery. H&E staining was performed to evaluate the morphology of the NP, fibrous tissue, and their boundaries^[180]43. During the 4- to 8-week observation period, the BNPMT@H-P group exhibited a reduction in NP tissue area; however, the tissue loss was significantly less than that observed in the BNPMT, H-P, and non-injection groups. Furthermore, in the BNPMT, H-P, and non-injection groups, the boundary between the NP and annulus fibrosus (AF) appeared blurred, and the NP region exhibited significant atrophy, indicating IVD fibrosis and the loss of the organized NP-AF structure (Fig. [181]7H). Safranin O-Fast Green staining was performed to assess proteoglycan content in the NP. In the NC group, the NP region was rich in proteoglycans, whereas proteoglycan levels were markedly reduced in the BNPMT, H-P, and non-injection groups. However, proteoglycan depletion was less pronounced in the BNPMT@H-P gel group compared to the other puncture groups (Fig. [182]7I). Histological grading revealed that the BNPMT@H-P group had significantly lower degeneration scores at both 4 and 8 weeks compared to the BNPMT, H-P, and non-injection groups (Fig. [183]7J). Fig. 7. Significant delay of intervertebral disc degeneration progression by BNPMT@H-P in a puncture-induced rat IVD degeneration model. [184]Fig. 7 [185]Open in a new tab A Schematic illustration of puncture-induced IVDD and therapeutic intervention in rat model. B In vivo IVIS imaging at 0, 3, 7, 14, and 21 days post-injection showing dynamic changes in fluorescence intensity at the rat intervertebral disc site (Color scale: blue [low] to red [high]). C Quantification of fluorescence signal intensity in experimental groups over 8 weeks (n = 3 independent rats, mean ± s.d.). D Representative μCT reconstructions of rat tail intervertebral discs across treatment groups. E Quantitative analysis of intervertebral DHI from radiographic measurements (n = 3 independent rats, mean  ± s.d.). F Comparative MRI cross-sections of rat tail intervertebral discs in experimental cohorts. G NP T2-signal intensity quantification at 4- and 8-week timepoints (n = 3 independent rats, mean ± s.d.). H Temporal progression of intervertebral disc morphology demonstrated by H&E staining (Multiple timepoints/treatment groups). I Extracellular matrix remodeling visualized through Safranin O/Fast Green staining (Representative timepoints/groups). J Histopathological grading system scores following surgical intervention (4/8-week post-operation, n = 3 independent rats, mean ± s.d.). Statistical significance between groups at each time point was assessed using an unpaired two-tailed Student’s t-test (C). Created with BioRender.com (A). Statistical analyses were performed using one-way ANOVA followed by Tukey’s multiple comparison test (E, G, J). Source data are provided as a Source Data file. Next, we evaluated the in vivo capability of the BNPMT@H-P dual-mode system to eliminate exDNA. Initially, we performed immunofluorescence staining for dsDNA on intervertebral disc sections (Fig. [186]8A and Supplementary Fig. [187]21), and the results showed a significant reduction in dsDNA fluorescence intensity in NP tissues following BNPMT@H-P treatment. To further quantitatively assess the hydrogel system’s ability to eliminate exDNA from the tissue microenvironment, we conducted a quantitative analysis at the tissue level. We collected NP tissues and isolated NP cells, then extracted the extracellular matrix from the nucleus pulposus to quantitatively analyze the exDNA levels in the NP microenvironment (Fig. [188]8B). The results showed a significant reduction in exDNA levels after BNPMT@H-P administration, consistent with the immunofluorescence findings, suggesting effective exDNA clearance by BNPMT@H-P within the disc microenvironment. In addition, to evaluate the in vivo efficacy of the hydrogel system in suppressing the cytoplasmic release of mtDNA, we conducted qPCR analysis. qPCR analysis of cytoplasmic DNA from isolated NP cells revealed a significant reduction in mtDNA levels compared to the IVDD group (Fig. [189]8C). Collectively, these results indicate that BNPMT@H-P effectively clears exDNA from the NP microenvironment and inhibits the release of mtDNA into the cytoplasm of NP cells. To determine whether the removal of both intracellular and extracellular dsDNA by BNPMT@H-P could suppress STING expression in NP cells and reduce inflammation within the disc, we performed immunohistochemical staining for STING in the intervertebral discs (Fig. [190]8D, E and Supplementary Fig. [191]22), and immunofluorescence staining for IL-6 and IL-1β (Fig. [192]8F, G). The results showed that BNPMT@H-P significantly downregulated STING expression and reduced inflammatory cytokine levels, suggesting that BNPMT@H-P may suppress the release of SASP factors from NP cells via inhibition of the STING pathway. Meanwhile, immunofluorescence analysis of the senescence markers p16 and p21 (Fig. [193]8H, I and Supplementary Figs. [194]23 and [195]24) was performed to assess whether BNPMT@H-P could reverse NP cell senescence. Post-injection, a marked reduction in the average fluorescence intensity of p16 and p21 was observed, indicating that BNPMT@H-P can alleviate cellular senescence in NP cells, thereby contributing to IVDD treatment. Furthermore, IHC analysis revealed a significant increase in the expression of the key ECM marker COL2 and a decrease in the ECM degradation marker MMP3 in the BNPMT@H-P group compared with other puncture groups (Fig. [196]8J, K and Supplementary Figs. [197]25 and [198]26). These findings suggest that BNPMT@H-P promotes intervertebral disc regeneration by remodeling the disc microenvironment and mitigating NP cell senescence. Fig. 8. Analysis of treatment effects after injection of BNPMT@H-P. [199]Fig. 8 [200]Open in a new tab A Representative immunofluorescence images of dsDNA staining in IVD tissues at 8 weeks post-treatment. Scale bar, 100 μm. Representative images are from three independent experiments. B Quantification of exDNA levels in the IVD microenvironment across different treatment groups (n = 3 independent rats, mean ± s.d.). C Cytoplasmic mtDNA levels in NP cells isolated from IVD tissues of different treatment groups (n = 3 biological replicates, mean ± s.d.). D Representative Immunohistochemistry images of STING expression in IVD tissues at 8 weeks post-treatment across different experimental groups. Scale bar, 100 μm. E Quantitative analysis of STING expression levels at week 8. (n = 3 independent rats, mean ± s.d.). F Representative immunofluorescence images of IL-6, IL-1β staining in IVD tissues at 8 weeks post-treatment. Scale bar, 100 μm. G Quantitative analysis of fluorescence intensities for IL-6, IL-1β at week 8. (n = 3 independent rats, mean ± s.d.). H Representative immunofluorescence images of p16, p21 staining in IVD tissues at 8 weeks post-treatment. Scale bar, 100 μm. I Quantitative analysis of fluorescence intensities for p16, p21 at week 8. (n = 3 independent rats, mean ± s.d.). J Representative Immunohistochemistry images of COL2 and MMP3 expression in IVD tissues at 8 weeks post-treatment across different experimental groups. Scale bar, 100 μm. K Quantitative analysis of COL2 and MMP3 expression levels at 8-week. (n = 3 independent rats, mean ± s.d.). Statistical analyses were performed using one-way ANOVA followed by Tukey’s multiple comparison test (B, C, E, G, I, K). Source data are provided as a Source Data file. Discussion In this study, we developed a hydrogel-based nanocomposite system (BNPMT@H-P) that implements an organelle–environment synergistic strategy for the treatment of IVDD. By simultaneously targeting intracellular mtDNA release and capturing exDNA, this platform interrupts the self-amplifying inflammatory loop driven by pathogenic nucleic acids. The biomimetic nanovesicles derived from NP cell membranes enabled homologous targeting and mitochondrial delivery of BAI1, restoring mitochondrial integrity, suppressing SASP factor release, and improving metabolic function in senescent NP cells. Meanwhile, the PAMAM-based hydrogel matrix not only enabled sustained local drug release but also neutralized pro-inflammatory exDNA through electrostatic binding, thereby modulating the extracellular immune microenvironment. Importantly, this dual-modality approach not only achieved robust anti-inflammatory effects but also enhanced the disc’s intrinsic capacity for self-repair by regulating NP cell fate and function. The therapeutic benefits were sustained over an 8-week course in a rat model of trauma-induced IVDD, demonstrating the translational potential of this organelle–environment synergistic strategy as a paradigm for nucleic acid-targeted IVDD therapy. Although our model was based on injury-induced disc degeneration, which replicates key pathological features such as mitochondrial dysfunction, sterile inflammation, and ECM breakdown, the mechanisms targeted by our therapy are broadly relevant to other clinically prevalent IVDD subtypes^[201]44,[202]45. In age-related IVDD, mitochondrial instability and progressive accumulation of senescent NP cells are well-established hallmarks^[203]18,[204]46,[205]47. Similarly, in mechanical stress-induced degeneration—common in physically demanding occupations or spinal deformities—chronic compression accelerates NP cell senescence, elevates mtDNA and exDNA levels, and drives pro-inflammatory cascades^[206]48. Given that BNPMT@H-P system specifically targets these conserved degenerative mechanisms, we believe it holds promise beyond acute injury contexts and may offer broader applicability across the heterogeneous clinical spectrum of IVDD. Despite encouraging preclinical results, several challenges remain before clinical translation can be realized. The injectability and mechanical compatibility of the hydrogel in human intervertebral discs need thorough evaluation, as human discs are larger and endure greater mechanical loads than rodent models^[207]49; thus, the hydrogel must sustain structural integrity and controlled release under physiological stress without compromising disc biomechanics. Optimizing viscoelastic properties and enabling minimally invasive delivery via fine-gauge needles are critical^[208]50. Long-term biocompatibility and safety must also be established, since repeated injections and potential immune responses over extended periods in human discs remain uncharacterized despite generally favorable safety profiles of PAMAM dendrimers and hyaluronic acid derivatives^[209]51. Moreover, given the multifactorial nature of human IVDD—including age-related degeneration, mechanical overload, metabolic and genetic factors—it is essential to evaluate efficacy across diverse, clinically relevant models such as age-associated and mechanically induced degeneration^[210]52. Scalable production of biomimetic nanovesicles with consistent quality and stability is also vital for regulatory approval and commercialization^[211]29. Importantly, our platform leverages biocompatible, tunable components—cell membrane-derived vesicles for homotypic targeting, BAI1 as a mitochondrial stabilizer, and PAMAM/HA hydrogels for localized, sustained drug release and exDNA scavenging—offering adaptability to different disease stages and administration routes (e.g., image-guided injection). Future work should assess long-term safety, biodistribution, and regenerative outcomes in spontaneous or age-related IVDD models, while optimizing vesicle formulations and dosing regimens tailored to patient-specific factors to enhance therapeutic precision and clinical feasibility. Together, our work provides a mechanistically informed and clinically adaptable framework for treating IVDD by modulating nucleic acid–driven inflammation. This strategy opens new avenues for developing non-surgical, cell-free therapies that preserve disc function and delay disease progression in diverse patient populations. Methods Data collection and reanalysis Single-cell RNA sequencing (scRNA-seq) data from degenerated and non-degenerated intervertebral discs of a single individual were obtained from the Gene Expression Omnibus database ([212]GSE165722). The scRNA-seq analysis was conducted using the 10× Genomics platform, and data processing was performed with the Seurat package. Low-quality cells were filtered out based on the following criteria: cells expressing fewer than 200 or more than 6000 genes were excluded, as abnormal gene counts may indicate dying cells, cell membrane damage, or doublets. Additionally, cells with mitochondrial gene content exceeding 40% were removed, as this suggests excessive cytoplasmic leakage and mRNA escape. After quality control, high-quality cells were normalized and scaled using the “NormalizeData” and “ScaleData” functions to achieve linear transformation. PCA was conducted on the top 2000 variable genes, and the 30 most significant principal components were selected for clustering analysis. To correct for batch effects, the “RunHarmony” function from the Harmony package was applied to integrate scRNA-seq data across multiple samples. Clustering results were visualized using Uniform Manifold Approximation and Projection (UMAP). Finally, cells were classified based on characteristic markers, and DEGs were identified. GO enrichment analyses were performed for each cell group. mRNA sequencing experimental method and analysis Total RNA was extracted using the TRIzol reagent (Invitrogen, USA) following the manufacturer’s protocol. RNA purity and concentration were measured with a NanoDrop 2000 spectrophotometer (Thermo Scientific, USA), while RNA integrity was assessed using an Agilent 2100 Bioanalyzer (Agilent Technologies, USA). Library preparation was performed using the VAHTS Universal V6 RNA-seq Library Prep Kit according to the manufacturer’s instructions. Transcriptome sequencing and analysis were conducted by OE Biotech Co., Ltd. (Shanghai, China). Bioinformatic analysis was performed using the OECloud tools at ([213]https://cloud.oebiotech.com/task/). The volcano map (Or other graphics) was drawn based on the [214]R on the OECloud platform ([215]https://cloud.oebiotech.com/task/). Metabolomics sequencing and analysis NP cell samples from each experimental group were collected and washed 2–3 times with pre-cooled PBS at 4 °C. After removing the PBS, the cells were immediately snap-frozen in liquid nitrogen. For metabolite extraction, each sample was treated with a total of 1 mL pre-chilled methanol:water (v:v = 4:1), added in two aliquots into 1.5 mL EP tubes. The samples were thoroughly dispersed using a pipette and subjected to ultrasonication in an ice bath (1620 W, 10 min, 6 s on / 4 s off cycles). This was followed by an additional 20 min of ultrasonic extraction in an ice-water bath and incubation at –40 °C overnight. Subsequently, the samples were centrifuged at 12,000 × g for 10 min at 4 °C. A total of 800 μL of supernatant was collected (in two 400 μL portions), transferred to LC-MS vials, and evaporated to dryness. The dried residue was reconstituted in 300 μL of methanol:water (v:v = 1:4) containing internal standards (4 μg/mL), vortexed for 1 min, ultrasonicated for 10 min, and incubated again at –40 °C overnight. After a second centrifugation at 12,000 × g for 20 min at 4 °C, 150 μL of the supernatant was transferred to LC-MS vials equipped with inserts for analysis. All extraction reagents were pre-cooled to −20 °C prior to use. Quality control (QC) samples were prepared by pooling equal volumes of extracts from all samples. Untargeted metabolomics analysis was performed using a Waters ACQUITY UPLC I-Class Plus system coupled with a Thermo Scientific Q Exactive high-resolution mass spectrometer. Chromatographic separation was carried out on an ACQUITY UPLC HSS T3 column (100 mm × 2.1 mm, 1.8 μm) maintained at 45 °C. The mobile phases consisted of solvent A (water with 0.1% formic acid) and solvent B (acetonitrile), delivered at a flow rate of 0.35 mL/min. The injection volume was 5 μL. Raw data were processed and analyzed by OE Biotech Co., Ltd. (Shanghai, China). Bioinformatic analysis was performed using OECloud tools ([216]https://cloud.oebiotech.com/task/). Volcano plots and other visualizations were generated using R software ([217]https://www.r-project.org/) via the OECloud platform. Cell culture Four-week-old male Sprague-Dawley rats were obtained from the Experimental Animal Center of Zhejiang Chinese Medical University, China. NP tissue was isolated from the rat tail intervertebral discs, minced, and digested with type II collagenase at 37 °C for 4 h. The resulting cell suspension was filtered twice through a 70 μm single-cell strainer to obtain a single-cell suspension, followed by centrifugation at 1000 × g for 5 min to collect primary NP cells. The collected NPCs were cultured in Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (Sigma-Aldrich, USA) and 1% streptomycin-penicillin solution (Gibco, USA). NP cells were cultured at 37 °C and 5% CO[2]. The culture medium was refreshed the following day, and cells were passaged upon reaching 80% confluence. NP cells in the second passage were regarded as normal state and used to perform experiments. RAW264.7 cell lines were purchased from CCTCC (Wuhan, China). The macrophage was cultured in DMEM containing 10% FBS. Membrane surface modification of nucleus pulposus cells with TPP The isolated nucleus pulposus cells were seeded in 10 cm culture dishes and cultured in DMEM supplemented with 10% fetal bovine serum (FBS), 1% penicillin/streptomycin (PS), and 10 μM non-natural mannose derivative (Ac4ManNAz) for 48 h. After incubation, the cells were washed twice with PBS, and the medium was replaced with serum-free medium containing 10 μM DBCO-TPP for a 2-h treatment. In the experiment characterizing the efficiency of the click chemistry reaction, DBCO-TPP was replaced with equal concentration of DBCO-FITC. Preparation and characterization of biomimetic nanovesicles NP cell membranes and TPP-modified NP cell membranes were extracted using a Membrane and Cytosol Protein Extraction Kit (Beyotime, China) according to the manufacturer’s instructions. The membrane protein concentration was adjusted to 1 mg/mL using a BCA Protein Assay Kit (Beyotime, China). The isolated membranes were then extruded through a 220 nm polycarbonate membrane at least five times to ensure uniform vesicle dispersion. BAI1 (HY-103269, MCE, USA) was dissolved in DMSO at a concentration of 10 mM and mixed with an equal volume of PBS containing either unmodified NP membranes nanovesicles (NPM) or TPP-modified NP membranes nanovesicles (NPMT). The resulting mixture was sonicated on ice using an ultrasonic processor (10 s on, 5 s off, repeated for 10 cycles) to obtain BAI1-loaded nanovesicles, termed BNPM and BNPMT. The nanovesicles were subsequently collected by ultracentrifugation at 20,000 × g for 1 h at 4 °C. The BAI1 loading efficiency was determined by HPLC (Agilent 1260, USA). The morphology of the nanovesicles was characterized using cryogenic transmission electron microscopy (Cryo-EM), and their hydrodynamic diameter and zeta potential were measured using a Zetasizer Nano ZS90 DLS instrument (Malvern Panalytical, UK). BNPMT targeting nucleus pulposus cells mitochondria Confocal microscopy was used to assess the targeting efficiency of nanovesicles. FITC-encapsulated nanovesicles were prepared using the same method as BNPM and BNPMT to evaluate their targeting capabilities. For cellular uptake experiments, equal numbers of NP cells and RAW264.7 macrophages were incubated with free FITC, FITC@NPM, or FITC@NPMT (with the equal FITC concentration of 10 µg/mL) for 4 h. After incubation, nuclei were stained with Hoechst dye (Yeasen, China). For mitochondrial targeting experiments, MitoTracker (Beyotime, China) was used to label mitochondria in NP cells during BNPM and BNPMT uptake assays. Live-cell imaging was performed using a Nikon confocal laser scanning microscope to observe nanovesicle uptake under different treatment conditions. Cells treatment To induce senescence in primary NPCs, cells were treated with 200 nM Dox for 24 h, followed by culture in fresh medium for an additional 4 days. In the NC group, NPCs were treated with an equal volume of PBS under the same conditions. In the experimental group, BAI1, BNPM, BNPMT (2.5 μM based on BAI1) was co-cultured with NPCs for 24 h. Cytoplasmic DNA isolation After collecting the NP cells with different treatments, they were placed in 500 μL of buffer (Containing 150 mM NaCl, 50 mM HEPES, pH 7.4, and 25 μg/ml digitonin (Sigma-Aldrich, USA) and incubated at room temperature for 10 min. The cells were then centrifuged at 150 × g for 5 min at 4 °C, and the supernatant was collected. This supernatant was further centrifuged twice at 150 × g at 4 °C, followed by a final centrifugation at 17,000 × g for 10 min to obtain the cytoplasmic fraction. DNA was extracted from the cytoplasmic fraction using the DNeasy Blood & Tissue Kit (Qiagen, 69504) according to the manufacturer’s instructions. Finally, the concentration of the extracted DNA was measured using the Qubit™ dsDNA Assay Kit (Invitrogen, USA). Co-culture of macrophages with H-P gel RAW264.7 macrophages were seeded into the lower chambers of 12-well Transwell plates for co-culture with H-P hydrogel. The experimental groups were designed as follows: (1) NC group, with no hydrogel added; (2) CpG group, with CpG added to the lower chamber at a final concentration of 1 μg/mL in DMEM; and (3) CpG + H-P group, with H-P hydrogel placed in the upper chamber and CpG (1 μg/mL) added to the lower chamber medium. After 24 h of incubation, macrophages and their supernatants were collected for flow cytometry, qPCR, and ELISA analyses. Preparation of Conditioned Medium: The macrophage supernatants were centrifuged at 1000 × g for 5 min, and the resulting supernatants were mixed with an equal volume of fresh complete DMEM to generate conditioned media for each group. NP cells were then cultured in the respective conditioned media for 24 h, followed by immunofluorescence staining, ELISA, WB, and qPCR analysis. Immunocytochemistry After washing the cells three times with PBS, we added 4% paraformaldehyde to completely cover and fix the cells at room temperature for 15 min. Next, the cells were washed three times with PBST, and then 0.1% Triton ×-100 was added to permeabilize the cells, followed by a 20-min incubation at room temperature and three additional PBST washes. The cells were then blocked with 5% BSA at room temperature for 60 min. After blocking, primary antibodies (BAX6A7 antibody, Santa Cruz, USA; TOMM20 antibody, Abcam, UK; dsDNA marker, Santa Cruz, USA; Collagen Type II Polyclonal antibody, Proteintech, China; MMP3 antibody, Proteintech, China) were added, and the cells were incubated overnight at 4 °C. Following the removal of primary antibodies, the cells were washed three times with PBST and incubated with secondary antibodies Alexa Fluor® 488 (Abcam, UK) or Alexa Fluor® 594 (Abcam, UK) at room temperature for 1 h. After this, the cells were washed three times with PBST and stained with DAPI (Solarbio, China) to visualize the nuclei. Finally, cell images were captured using a confocal microscope to assess the localization and expression of the target proteins. Western blotting Cells were lysed using a lysis buffer (Fude, China) supplemented with phosphatase inhibitors (Beyotime, China) and protease inhibitors (Beyotime, China). The protein concentration of the lysate was determined using the BCA protein assay kit (Beyotime, China). Next, protein samples were mixed with protein loading buffer (Fude, China) and boiled in a metal bath at 100 °C for 5 min. Proteins were then separated by SDS-PAGE and transferred onto a PVDF membrane (Millipore, USA). To prevent nonspecific binding, the membrane was blocked with 5% BSA at room temperature for 1 h. After blocking, primary antibodies (ATP1A3 Polyclonal antibody, Proteintech, China; Alpha Tubulin Polyclonal antibody, Proteintech, China; Phospho-TMEM173/STING (Ser366) Antibody, Affinity, USA; TMEM173/STING Antibody, Affinity, USA; Phospho-IRF3 (Ser396) Antibody, Affinity, USA; IRF3 Antibody, Affinity, USA; Phospho-TBK1 (Ser172) Antibody, Affinity, USA; TBK1 Antibody, Affinity, USA; GAPDH Polyclonal antibody, Proteintech, China; Collagen II Antibody, Affinity, USA; MMP3 Antibody, Affinity, USA) were added, and the membrane was incubated overnight at 4 °C. The membrane was then washed three times with TBST to remove any unbound primary antibodies, followed by incubation with secondary antibodies (HRP-conjugated Goat Anti-Rabbit IgG(H + L), Proteintech, China; HRP-conjugated Goat Anti-mouse IgG(H + L), Proteintech, China) at room temperature for 1 h. A final wash with TBST was performed, and target protein signals were detected using the ECL chemiluminescence method (Fude, China). Protein band intensities were semi-quantified using ImageJ software. qRT-PCR and ELISA analyses Total RNA from cells was extracted using an RNA extraction kit (Accurate Biology, China). RNA concentration was measured using the NanoDrop ND-1000 spectrophotometer. Subsequently, RNA was reverse-transcribed into cDNA using the Evo M-MLV reverse transcription kit (Accurate Biology, China). Real-time qPCR was performed using qPCR SYBR Green Master Mix (Yeasen, China), with specific primer sequences provided in Supplementary Table [218]1. For cytosolic DNA analysis, 20 ng of dsDNA was used for qPCR. Cell supernatants were collected, and cytokine concentrations were determined using ELISA kits (Boster, China). Flow cytometry Macrophages were co-incubated with CpG-FITC and HP hydrogel for 24 h. After incubation, the cells were collected and analyzed by flow cytometry (Beckman, USA) to assess the FITC fluorescence intensity, which reflects their uptake of CpG-FITC. To evaluate macrophage M1 polarization, a similar co-incubation was performed, after which the cells were collected and resuspended in 0.1% Triton ×-100, then incubated at room temperature for 10 min. Following centrifugation at 1000 × g for 5 min to discard the supernatant, the cells were resuspended in 5% BSA and incubated at room temperature for 30 min. After another centrifugation at 1000 × g for 5 min, PE-anti-iNOS antibody (BioLegend, USA) was added and the cells were incubated for 30 min. Finally, the cells were analyzed by flow cytometry to determine their fluorescence, indicating the level of M1 polarization. After treatment, NP cells from different groups were collected and stained using an Annexin V-FITC apoptosis detection kit (Beyotime, China) according to the manufacturer’s instructions. The stained cells were then analyzed by flow cytometry. Preparation and characterization of H-P Gel/BNPMT-loaded H-P Gel HA-CHO was synthesized via the sodium periodate oxidation method^[219]53. To form the H-P hydrogel, 5 wt% lyophilized HA-CHO and 1 wt% PAMAM solution were each dissolved in equal volumes of PBS and mixed at room temperature. For the preparation of BNPMT-loaded H-P hydrogel, HA-CHO was first dissolved in PBS containing BNPMT, followed by mixing with an equal volume of PAMAM solution. Gel formation was visually confirmed by observing the vial for signs of gelation. To enhance visualization, a non-reactive dye was added to the hydrogel, and its injectability and self-healing properties were documented using photography. The dynamic rheological properties of the H-P hydrogel were evaluated using a strain-controlled rheometer (Anton Paar MCR302, Austria). Rat IVDD model construction Eight-week-old male Sprague-Dawley (SD) rats were obtained from the Animal Experiment Center of Zhejiang Chinese Medical University (China). All animal experiments were approved by the Animal Experiment Ethics Committee of Zhejiang Chinese Medical University. The rats were housed in a pathogen-free environment under a 12-h light/dark cycle, with ad libitum access to food and water. They were randomly assigned to different groups for in vivo experiments. The rats were anesthetized by intraperitoneal injection of sodium pentobarbital (45 mg/kg) and placed in a prone position. A 20-gauge needle was used to puncture the Co6/7 intervertebral disc from the dorsal side; the needle was inserted from the center of the disc to the opposite side, rotated 180°, and held for 10 s. The Co5/6 or Co7/8 intervertebral discs were preserved intact as internal controls. Evaluations were performed at 4 and 8 weeks post-treatment. The control group received no puncture or injection. The BNPMT group was injected with 20 μL of BNPMT nanovesicles (Containing 0.5 mM BAI1). The H-P group received 20 μL of H-P hydrogel alone. The BNPMT@H-P group was administered 20 μL of BNPMT-loaded H-P hydrogel (Containing 0.5 mM BAI1). Radiological evaluation analysis Rats were euthanized at 4 and 8 weeks after treatment, and their tail intervertebral disc specimens were collected and fixed in formaldehyde. μCT (SkyScan, Belgium) and MRI (Universal Corporation, USA) were used to evaluate intervertebral disc degeneration in the rat tail, focusing on changes in nucleus pulposus signal intensity and intervertebral space height. X-ray images were analyzed using ImageJ software to calculate the Disc Height Index (DHI%). T2-weighted images were also obtained through coronal scans. Pathological damage to the tail intervertebral discs was assessed by three experienced radiologists and three orthopedic surgeons based on the modified Thomson classification. In vivo imaging of rats ICG was encapsulated into nanovesicles derived from nucleus pulposus cell membranes (ICG@NPMT with a final ICG concentration of 10 µg/mL) using an ultrasonic co-extrusion method to evaluate their in vivo release behavior. The ICG@NPMT nanovesicles were subsequently encapsulated within the H-P hydrogel and injected into the rat coccygeal intervertebral disc at day 0. This approach enabled real-time tracking of the nanovesicles’ release profile in vivo.In parallel, to evaluate the in vivo degradation behavior of the H-P hydrogel itself, ICG was covalently conjugated to the hydrogel using ICG-NHS, which forms stable amide bonds with the abundant primary amine groups (–NH[2]) on PAMAM dendrimers. The ICG-labeled hydrogel was similarly injected into the rat intervertebral disc at day 0. Fluorescence signals were captured at predetermined time points (Day 0, 3, 7, 14, and 21) using an in vivo imaging system (IVIS Lumina III, PerkinElmer, USA) to monitor the dynamic release of nanovesicles and the degradation of the hydrogel matrix, respectively. Histological evaluation, immunohistochemistry, and immunofluorescence At 4 and 8 weeks after model induction, rats in each group were sacrificed, and caudal intervertebral disc specimens were collected and fixed in 4% paraformaldehyde. The specimens were then decalcified in 15% EDTA for at least 4 weeks and embedded in paraffin to prepare 5 μm continuous sections. H&E and Safranin O/Fast Green staining were performed to observe changes in the intervertebral disc tissue structure and collagen content. To further assess ECM synthesis and degradation in the disc, as well as the expression of inflammatory factors, immunohistochemistry and immunofluorescence staining were performed. The sections were deparaffinized with xylene and a gradient of ethanol (100–80%) and underwent antigen retrieval using citrate buffer. Blocking was carried out with a 5% BSA solution, followed by overnight incubation with primary antibodies at 4 °C. Secondary antibodies (HRP or fluorescence-labeled) were incubated at 37 °C for 1 h. DAB was used for color development, followed by counterstaining with hematoxylin. Finally, sections were mounted with neutral resin or anti-fade mounting medium and observed using a scanning system. Statistical analysis All experiments were repeated at least three times. Semi-quantitative analysis was performed on at least three samples, and the results are expressed as the mean ± standard deviation (s.d.). Statistical analysis was conducted using GraphPad Prism 10 software (GraphPad Software Inc.). Comparisons between two groups were made using an unpaired two-tailed Student’s t-test, and for multiple group comparisons, one-way ANOVA followed by Tukey’s post hoc test was applied. A P-value of less than 0.05 was considered statistically significant. Ethics approval All ethical aspects of the human-related components of this study, including the consultation of patient medical data and the collection and use of human nucleus pulposus (NP) tissue samples, were approved by the Ethics Committee of Hangzhou Hospital of Traditional Chinese Medicine (Grant number: 2022KY039). Normal human NP tissue samples were collected from patients aged 27–31 years with burst fractures. Based on MRI T2-weighted imaging and the Pfirrmann grading system, the non-degenerated intervertebral discs were classified as grade I. Degenerated human NP tissue samples were collected from patients aged 56–62 years with lumbar disc herniation, with the corresponding discs graded as Pfirrmann grade IV. All selected patients had no history of cardiovascular or cerebrovascular diseases, cancer, infections, immune or endocrine disorders, or organ dysfunction. In total, three pairs of samples (three healthy and three degenerated) were used for immunohistochemical analysis. Written informed consent was obtained from all patients and volunteers for the collection and use of their tissue samples in scientific research. In addition, separate written informed consent was obtained to publish information that could potentially identify individuals (e.g., age, sex). All relevant approvals from China’s Ministry of Science and Technology regarding the use and export of human genetic resources relevant to this study have also been obtained. All animal-related procedures, including animal acquisition, surgical operations, and sample collection, were approved by the Animal Ethics Committee of Zhejiang Chinese Medical University (Grant number: 20250331-08). Reporting summary Further information on research design is available in the [220]Nature Portfolio Reporting Summary linked to this article. Supplementary information [221]Supplementary information^ (2.8MB, pdf) [222]41467_2025_63194_MOESM2_ESM.pdf^ (4.7KB, pdf) Description of Additional Supplementary Files [223]Supplementary Data 1^ (163.8KB, xlsx) [224]Reporting Summary^ (124.6KB, pdf) [225]Transparent Peer Review file^ (3MB, pdf) Source data [226]Source Data^ (1MB, xlsx) Acknowledgements