Abstract Background Osteoarthritis (OA) is a degenerative joint disease with an immense unmet medical need. FGF18 protein is a potential regenerative factor for cartilage repair. However, traditional protein delivery methods have limited efficacy due to the short lifetime and shallow infiltration. Results In this work, we discovered that lipid nanoparticle (LNP) can infiltrate and deliver FGF18 mRNA deeper in the cartilage than proteins. After mRNA UTR optimization and chemical modification, the expression of FGF18 can last up to 6 days in the cartilage. Furthermore, delivering FGF18 mRNA activates FOXO3a-autophagy pathway, which protects against chondrocyte degeneration and senescence. Local intra-articular injection of FGF18 mRNA-LNP significantly alleviates OA symptoms in DMM and senile OA models. Sustained expression and accessibility of FGF18-mRNA to deeper chondrocytes makes LNP-mRNA more effective than FGF18 recombinant protein. Conclusions In summary, this study presents a novel approach superior to recombinant protein alone and holds promise as a new therapeutic strategy for OA. Graphical abstract [50]graphic file with name 12951_2025_3103_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12951-025-03103-9. Keywords: Osteoarthritis, FGF18, mRNA, Lipid nanoparticles, Autophagy Introduction Osteoarthritis (OA) is a prevalent joint disease worldwide, affecting over 250 million people and imposing a significant burden on the economy and society [[51]1, [52]2]. Currently, most clinical treatments aim to alleviate pain symptoms in the early stages of the disease. Several biologic therapies [[53]3–[54]6] have shown promising efficacy in the osteoarthritis treatment. Despite encouraging preclinical results, a series of drugs failed in clinical development [[55]7]. Numerous clinical deficiencies in drug efficacy can be attributed to insufficient drug delivery [[56]8]. Rapid clearance and inadequate penetration depth are the main barriers for the effective drug delivery in cartilage. Even using intra-articular injections, the half-lives of many compounds are only 2–4 h [[57]8]. The recombinant protein is also rapidly degraded, and only 7.7% of the recombinant protein remains on the surfaces of the femur and tibial cartilage after 24 h of injection [[58]9]. The short half-life reduces the function time of the therapy and restricts the overall efficacy. In addition, the majority of chondrocytes reside in the middle or deep zones of cartilage and covered by the negatively charged cartilage matrix, therefore, biological macromolecules are difficult to reach the chondrocytes [[59]10]. Thus, these unmet medical needs require novel strategies to improve the lifetime and penetration depth of recombinant proteins within the joint cavity. Recently, mRNA therapy has emerged as a promising approach [[60]11, [61]12]. In addition to the success of vaccines, mRNA has been applied in protein replacement therapy to rescue the protein deficiency, such as CoA carboxylase, Methylmalonyl-coenzyme A mutase (MUT), ornithine transcarbamylase (OTC) [[62]13]. The mRNA does not integrate into the genome. Once it enters the cytoplasm, it can be directly translated into proteins. Therefore, mRNA-based therapeutics have shown good safety profiles [[63]11]. However, negative charge and instability of mRNA, coupled with its susceptibility to degradation by nucleases and immunogenicity, highlight importance of a suitable delivery system [[64]14–[65]17]. Lipid nanoparticles (LNPs) can encapsulate the mRNA and neutralize the negative charge to extend its life in the patient [[66]18]. LNP are usually composed of four lipids, i.e. cationic ionizable lipids, phospholipids, PEG lipids, and cholesterol, which are widely used as an efficient delivery system for mRNA [[67]11, [68]19–[69]21]. The ionizable cationic lipids significantly extends the half-life of mRNA, protecting mRNA from nuclease-mediated degradation. In body fluids with a neutral pH, LNPs do not have any charge and therefore, do not interact with negatively charged red blood cells. However, after they are taken up by cells, LNPs become positively charged inside the acidic endosomal environment, helping mRNA to escape from the endosomes. In addition, modifying antibodies on the surface of LNP can also enhance the targeted delivery of mRNA [[70]22]. Overall, using LNP for mRNA delivery can overcome physiological barriers and enable the targeted delivery and translation of mRNA at the site of interest [[71]11]. While LNPs have been extensively studied in soft tissues [[72]19, [73]23–[74]25]. delivering drugs to cartilage is a challenging task due to its unique inorganic and organic composition. Recent studies have revealed that FGF18 plays an important role in the development and progression of osteoarthritis [[75]26–[76]29]. The FGF family and its receptors, known as FGF receptors (FGFRs), aid in the differentiation of chondrocytes and protect articular cartilage from degeneration during bone and cartilage development [[77]30, [78]31]. A multi-cohort genome wide association meta-analysis of osteoarthritis reported that single nucleotide polymorphism (SNP) of FGF18 gene positively correlate with osteoarthritis [[79]32]. Studies on FGFR3 conditional knockout mice in chondrocytes showed increased severity of osteoarthritis after DMM (destabilization of medial meniscus) modeling [[80]33–[81]35]. Recombinant FGF18 protein has shown clinical potential for the treatment of osteoarthritis [[82]3, [83]36–[84]39]. OA is prevalent among the elderly, and cellular senescence is a significant characteristic of OA chondrocytes [[85]40, [86]41]. Autophagy, a normal cellular metabolic process, mediates the travelling of cellular components to lysosomes, promoting cell survival under stress conditions. Impaired autophagy is a key contributor to aging [[87]42]. In comparison to young mice, the expression of autophagy-related genes is significantly reduced in the cartilage of senile mice [[88]43]. These changes are associated with increased levels of apoptosis and cartilage degeneration. Similar findings have been observed in post-traumatic OA mouse models [[89]44], suggesting the importance of autophagy in both senescence and OA. Therefore, targeting chondrocyte autophagy is a promising direction for rescuing cartilage from senescence. In this study, we are motivated to investigate the efficacy of LNP encapsulated FGF18 mRNA in the OA treatment. Interestingly, we found that the LNP was able to penetrate deeper into the cartilage than FGF18 protein alone, resulting in better delivery to the joint cavity. By optimizing the UTR region and chemical modification, we were able to increase FGF18 expression by 10-fold compared to the intrinsic UTR of FGF18. LNP-FGF18 mRNA showed protective effects against chondrocyte degeneration and senescence in different models, including inflammatory chondrocytes and two mouse OA models. In summary, our study presents a novel approach to effectively treat chondrocyte senescence and degeneration in the cartilage by delivering LNP-encapsulated FGF18 mRNA in vitro and in vivo. Results FGF18 expression level decreases in cartilage inflammation and senescence microenvironment We conducted a comprehensive investigation into the expression levels of FGF18 in various osteoarthritis models. Our study included cellular cultures, murine models, and clinical specimens. Initially, we performed RNA sequencing on primary chondrocytes in an inflammatory environment. The decreased expression of cartilage matrix synthesis-related genes (SOX9, COL2A1 and ACAN) and the increased expression of matrix degradation-related genes (MMPs) confirm the successful stimulation by inflammatory factors. Results showed a 75% reduction in the expression level of FGF18 (Fig. [90]1A). We also stimulated ATDC5 chondrocytes with varying concentrations of IL-1β and TNF-α for 24 h. Results revealed an 80% decrease in mRNA expression of SOX9 and COL2A1, a 10-fold increase in MMP13 expression, and concomitant 60% reductions in FGF18 expression level (Fig. [91]1B). In the H[2]O[2]-induced cellular senescence model, the expression level of FGF18 decreased by 60% (Supplementary Fig. [92]1A). Results from the GSE database also indicate that the expression level of FGF18 in chondrocytes of elderly individuals is only 25% of that in young individuals (Supplementary Fig. [93]1B), confirming the downregulation of FGF18 in senescent microenvironment. To investigate the role of FGF18 in osteoarthritis, we established two distinct mouse models: destabilization of the medial meniscus (DMM) and a naturally aging model. Histological examinations (Safranine O/Fast Green) validated the successful creation of the arthritis models, with apparent cartilage abrasion in DMM and aging mice. (Fig. [94]1C-D, F-G). Immunohistochemical analyses revealed a 50% decrease in FGF18 positive cell proportion in both inflamed and senescent cartilage (Fig. [95]1C, E, F, H). Finally, we collected tibial plateau samples from patients undergoing total knee arthroplasty, which revealed a 50% decrease in FGF18^(+) positive cell proportion within damaged osteoarthritic cartilage (Fig. [96]1I-K). These results substantiate the proposition that the FGF18 experiences diminished expression within the microenvironment of joint cartilage inflammation and senescence while FGF18 supplementation could promote cartilage repair and delay the progression of OA. Fig. 1. [97]Fig. 1 [98]Open in a new tab FGF18 expression level decreases in inflammatory and senescent microenvironments. (A) Heatmap of relevant differential expression genes (DEGs) in chondrocyte RNA sequencing after TNF-α treatment. (B) mRNA expression levels of SOX9, COL2A1, MMP13, and FGF18 after stimulation with different concentrations of inflammatory factors (n = 3). (C-E) Safranin O/Fast Green staining, and FGF18 immunohistochemical staining and corresponding quantitative analysis in knee joints of DMM model (n = 6). Scale bar: 200 μm. (F-H) Safranin O/Fast Green staining, immunohistochemical staining of FGF18 and quantitative analysis in knee joint cartilage of senile model (n = 6). Scale bar: 200 μm. (I-K) Safranin O/Fast Green staining, immunohistochemical staining of FGF18, and quantitative analysis of clinical specimens (n = 6). Scale bar: 500 μm and 250 μm. Data are shown as mean ± SD. P value was calculated by one-way ANOVA, followed by Tukey’s post hoc tests or Student’s t test. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001 LNP-FGF18 mRNA delivers FGF18 to the deep zone of cartilage Penetration depth is a key factor the successful delivery of chondrocyte repair. We are wondering about the penetration depth of LNP and FGF18 proteins. We encapsulated the FGF18 mRNA within lipid nanoparticles (LNPs) (Fig. [99]2A). The prepared LNPs have an average size of 128 nm with a polydispersity index (PDI) of 0.019, showing a uniform particle size distribution (Fig. [100]2B). The Zeta potential is -4.03 ± 1.89 mV under physiological pH 7.4 conditions, and this value shifts to 22.0 ± 4.76 mV at pH 5.0, which is the pH value in endosomes. The regular spherical shape of LNPs was analyzed using cryo-transmission electron microscopy (Fig. [101]2C). Importantly, cell viability staining and CCK8 assays provided compelling evidence that 8 μg/ml of LNPs were non-toxic to the chondrogenic cell line, i.e. ATDC5 cell and 293T cells (Supplementary Fig. [102]2). Furthermore, we labeled FGF18 with Cy3 fluorescence and LNP with Dil, respectively, to trace their infiltration depth in the cartilage. Interestingly, recombinant FGF18 protein remains on the surface of the cartilage layer, whereas LNP-mRNA can penetrate five times deeper into the cartilage up to nearly 50 μm in both young and old mice cartilage (Fig. [103]2D, E). Furthermore, collagen fibril network pore size of young and old mice was observed and measured with SEM (Fig. [104]2F). Although collagen network in aged mice is denser and has smaller pore sizes compared to young mice (ranging from 102 nm to 260 nm), it remains similar in diameter to prepared LNPs. These results suggest that the LNP-mRNA system possesses superior penetration capabilities which enable the LNP to act on deeper chondrocytes. Fig. 2. [105]Fig. 2 [106]Open in a new tab LNP-mRNA penetrates into cartilage and expresses the target protein. (A) Flowchart of the LNP-mRNA System Preparation. (B) Particle size of LNP. (C) Representative cryo-transmission electron microscopy images of LNP, Scale bar: 100 nm. (D, E) Representative immunofluorescence images of the penetration depth of Cy3-labelled FGF18 and Dil-labelled LNP in the cartilage of mice of different ages and quantitative analysis (n = 4). White dashed line represents the boundary between the cartilage and the subchondral bone. Scale bar: 10 μm. (F) Representative SEM images of collagen fiber networks in the cartilage of mice of different ages. Scale bar: 1 μm. (G-H) Luminescence intensity at different time points after transfection with lipo3000 and LNP systems in 293T and ATDC5 cells (n = 4). (I) Representative fluorescent images of ATDC5 cells transfected with varying doses of EGFP mRNA-LNP at different time points. Scale bar: 50 μm. (J-K) Bioluminescence imaging and quantitative analysis of signal flux at different time points after intra-articular injection of LNP-Fluc mRNA in mouse knee joints (n = 3). Data are shown as mean ± SD. P value was calculated by one-way ANOVA, followed by Tukey’s post hoc tests. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001 LNP-FGF18 mRNA enables considerable expression of FGF18 in joint cavity We further compared the transfection efficiency between LNPs and Lipofectamine 3000 in the chondrogenic cell (ATDC5) and 293T cells. We used different concentrations of Fluc mRNA and our findings revealed that the LNP system had a superior transfection efficiency in both cell types, especially in ATDC5 cells, when compared to Lipofectamine 3000 (Fig. [107]2G, H). Additionally, we observed that mRNA signals from LNP transfection at the 72-hour time point were approximately half of what was observed at the 24-hour point, whereas Lipofectamine 3000-mediated transfection showed a more significant decline at the 72-hour time point. Representative fluorescence microscopy images were applied to evaluate the transfection of 293T cells and ATDC5 cells with EGFP mRNA-LNP at varying doses and time points. The fluorescence results align with the signal trends observed for Fluc mRNA (Fig. [108]2I and Supplementary Fig. [109]3). These results suggest that the LNP-mRNA system is highly effective and has a long-lasting release capability in cells. Subsequently, our investigation explored the duration of the LNP mRNA system’s expression in a murine model. We administered 3 μg of LNP-Fluc mRNA into the knee joint of mice and monitored Fluc expression at various intervals using bioluminescence imaging. Our findings confirmed that the LNP mRNA system can sustain localized knee joint expression for up to 6 days (Fig. [110]2J, K). Monitoring bioluminescence in other organs of the mice revealed minimal evidence of LNP metabolism through the liver. The signal diminished after 48 h, indicating no side effects of LNP on other organs (Supplementary Fig. [111]4). On the other hand, injecting mRNA directly into the knee joint resulted in a significantly reduced Fluc signal. The signal persisted for only 48 h (Supplementary Fig. [112]5). These results collectively validate the efficacy and protracted release capability of the LNP-mRNA system. To enhance the expression and secretion of FGF18 protein, we optimized the 5’ untranslated region (UTR) of FGF18 mRNA. Plasmids containing varying UTRs, including mouse UTR (mUTR), synthetic UTR (SynUTR) and human UTR (hUTR), were transfected into ATDC5 and 293T cells, respectively. RT-qPCR analysis definitively demonstrated 400-fold increase in FGF18 plasmids (Fig. [113]3A). SynUTR, in particular, had the most significant impact on enhancing FGF18 protein expression, resulting in a 1000-fold increase (Fig. [114]3B-C, Supplementary Fig. [115]7A, B). Dot blot experiments on cell culture supernatants also confirmed that synUTR plasmid transfection resulted in the highest concentration of secreted FGF18. regardless of whether it was in ATDC5 or 293T cells (Fig. [116]3D, Supplementary Fig. [117]6A, Supplementary Fig. [118]7C, D). We then encapsulated UTR-modified FGF18 mRNA within LNPs and measured the particle size, zeta potential, and encapsulation efficiency of LNPs containing different UTRs. The results indicate that the particle sizes of LNPs with various UTRs were all within the range of 120–130 nm, with zeta potentials between − 10 mV and 0 mV, and encapsulation efficiencies exceeding 90%. These findings suggest that while different UTRs of FGF18 mRNA can have some impact on LNP characterization, the effects are relatively minor. Transfection experiments in ATDC5 cells were then conducted at different time points. Notably, the results showed that FGF18 expression and secretion in the culture medium were positively correlated with the concentration of LNPs and negatively correlated with transfection time (Fig. [119]3E-K, Supplementary [120]6B, C). Specifically, 3 μg/mL LNP-FGF18 mRNA would generate 100 ng/ml FGF18 after 24 h. This specific concentration of LNP-FGF18 mRNA with SynUTR was subsequently utilized in all subsequent experiments. Fig. 3. [121]Fig. 3 [122]Open in a new tab Optimization of UTR regions of FGF18 mRNA to obtain sustained secretion of FGF18. (A-C) Detection and quantitative analysis of FGF18 mRNA and protein after transfection with plasmids containing different UTRs (n = 3). (D) FGF18 secretion was quantified in the cell culture supernatant of cells, which were tranfected with plasmids containing different UTRs (n = 3). (E-G) Detection and quantitative analysis of FGF18 mRNA and protein after transfection with different concentrations of LNP-FGF18 mRNA (n = 3). (H-I) Time course of FGF18 expression after transfection with LNP-FGF18 mRNA and quantitative analysis (n = 3). (J, K) Measurement of FGF18 content in the cell culture supernatant after transfection with different concentrations or durations of LNP-FGF18 mRNA (n = 3). Data are shown as mean ± SD. P value was calculated by one-way ANOVA, followed by Tukey’s post hoc tests. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001 LNP-FGF18 mRNA promotes the proliferation of chondrocytes by rescuing them from degeneration and senescence FGF18 protein is known for its ability to mitigate cartilage extracellular matrix degradation and stimulating matrix synthesis [[123]36], but the effects in the LNP-mRNA format is unclear. Motivated by the superior infiltration depth and duration, we compared the effects of recombinant FGF18 protein and LNP-FGF18 mRNA in cells cultured in inflammatory (10ng/mL IL-1β) or non-inflammatory conditions. Both of them enhanced the mRNA expression and protein translation of genes associated with cartilage matrix synthesis, among which COL2A1 and SOX9 increased by up to 2-fold, while simultaneously suppressed the mRNA linked to matrix degradation and fibrosis, such as MMP3, MMP13, and COL1A1, by over 50% compared to solely IL-1β group (Fig. [124]4A-C). Meanwhile, immunofluorescence results show a 30% increase in SOX9 positive cell proportion and COL2A1 fluorescent intensity after FGF18 or LNP-FGF18 mRNA treatment (Fig. [125]4D-F). Micromass culture experiments consistently demonstrated that both LNP-mRNA and FGF18 protein effectively augmented the secretion of cartilage cell matrix and rescued matrix depletion under inflammatory conditions after 4, 6, and 8 days (Fig. [126]4G-J). IOD value of toluidine blue staining increases by 20% after FGF18 or LNP-FGF18 mRNA treatment (Fig. [127]4G, H) and that of alcian blue also elevates 20–50% depending on different duration compared to control group or solely IL-1β group (Fig. [128]4I, J). Our findings suggest that the LNP mRNA exhibited similar effects to those of FGF18 recombinant protein in cells. Fig. 4. [129]Fig. 4 [130]Open in a new tab Recombinant FGF18 protein and LNP-FGF18 mRNA alleviate chondrocyte inflammation-induced degeneration. (A-C) mRNA transcription and protein expression levels of COL2A1, SOX9, MMP3, MMP13, and COL1A1 after treatment with inflammatory factors, FGF18 or LNP-FGF18 mRNA (n = 3 or 4). (D-F) Immunofluorescence and quantitative analysis of COL2A1 and SOX9 expression levels in ATDC5 chondrocytes treated with FGF18 or LNP-FGF18 mRNA (n = 4). Scale bar: 50 μm. (G-J) Alcian Blue and Toluidine Blue staining of ATDC5 chondrocyte micromass cultures at different time points treated with FGF18 or LNP-FGF18 mRNA and quantitative analysis (n = 4). Data are shown as mean ± SD. P value was calculated by one-way ANOVA, followed by Tukey’s post hoc tests. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001 To comprehensively explore the mechanism by which LNP-FGF18 mRNA affects chondrocytes and its role in preventing cartilage degeneration, we applied RNA sequencing to analyze ATDC5 chondrocytes treated with LNP-FGF18 mRNA for 24 h (Supplementary Fig. [131]8). The genes related to cell cycle and senescence such as E2F1 and CCND1 were enriched (Fig. [132]5A-C). We then conducted further senescence-related studies on primary murine chondrocytes. RT-qPCR and western blot experiments revealed that LNP and FGF18 could downregulate the elevated expression of P16^INK4a, P21, and P53 by 40–60% compared to solely IL-1β group (Fig. [133]5D-F). In line with these results, SA-β-gal staining experiments showed that treatment with FGF18 mRNA and FGF18 protein under inflammatory conditions reduced the number of senescent primary murine chondrocytes by 50%. Moreover, EdU assays and Ki67 immunofluorescence experiments conclusively demonstrated that both FGF18 and LNP-FGF18 mRNA could enhance the proliferation potential of primary chondrocytes, with proportions of EdU-positive and Ki67-positive cell proportions increased by approximately 100% compared to the solely IL-1β group (Fig. [134]5G-J). Fig. 5. [135]Fig. 5 [136]Open in a new tab Recombinant FGF18 protein and LNP-FGF18 mRNA inhibit chondrocyte senescence induced by inflammatory stimulation. (A, B) KEGG pathway enrichment analysis and GSEA(Gene Set Enrichment Analysis) of transcriptome sequencing data of ATDC5 chondrocytes stimulated with LNP-FGF18 mRNA. (C-E) mRNA and protein expression levels, along with quantitative analysis, of senescence-related genes in chondrocytes treated with FGF18 or LNP-FGF18 mRNA (n = 3 or 4). (F-I) Immunofluorescence staining for Ki67, EdU assays, and senescence-associated β-galactosidase staining after stimulation with FGF18 or LNP-FGF18 mRNA, along with quantitative analysis (n = 4). Scale bar: 50 μm and 200 μm. Data are shown as mean ± SD. P value was calculated by one-way ANOVA, followed by Tukey’s post hoc tests. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001 Afterwards, we attempted to understand the molecular mechanisms behind the recovery of cartilage senescence and degeneration with the help of FGF18. The RNA sequencing data showed that there was a significant increase in autophagy and FOXO signaling pathway after FGF18 LNP treatment (Fig. [137]5A). Notably, FGF18 is recognized as a gene associated with longevity and autophagy [[138]45], prompting us to explore into the expression patterns of autophagy-related genes. Our findings indicated that the addition of FGF18 mRNA led to an increase in FOXO3a and BECLIN1 expression, which in turn increased the LC3B II/I ratio (Fig. [139]6A, B). We also observed a decrease in the number of and autolysosomes in an inflammatory environment through immunofluorescence and transmission electron microscopy (Fig. [140]6C). Importantly, the presence of FGF18 protein and FGF18 mRNA significantly improved these changes. Fig. 6. [141]Fig. 6 [142]Open in a new tab Recombinant FGF18 and LNP-FGF18 mRNA activate chondrocyte autophagy pathway to counteract aging and degeneration. (A, B) Protein expression and quantitative analysis of FOXO3a and autophagy-related genes in ATDC5 chondrocytes after stimulation with FGF18 or LNP-FGF18 mRNA (n = 3). (C) Immunofluorescence staining of LC3 and representative images of autophagosomes by transmission electron microscopy after stimulation with FGF18 or LNP-FGF18 mRNA. Scale bar: 10 μm, 500 nm. (D, E) Protein expression and quantitative analysis of FOXO3a in FOXO3a knockdown cells (n = 3). (F, G) Expression and quantitative analysis of BECLIN1, LC3B, P16, SOX9 and COL2A1 in ATDC5 chondrocytes under the influence of IL-1β (10ng/mL), FOXO3a knockdown, 3-MA (5mM), and LNP-FGF18 mRNA (n = 3). (H-J) Alcian blue and toluidine blue staining for collagen in chondrocytes along with quantitative analysis (n = 4). (K-L) SA-β-gal staining, LC3B immunofluorescence, transmission electron microscopy representative images and related quantitative analysis (n = 4). Scale bar: 200 μm, 10 μm, 500 nm as shown in the figures. Data are shown as mean ± SD. P value was calculated by one-way ANOVA, followed by Tukey’s post hoc tests. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001 To elucidate the relationship between autophagy, chondrocyte senescence, and degeneration, we constructed FOXO3a knockdown cell lines (Fig. [143]6D-E) and imposed autophagy inhibition using 3-MA as inhibitor [[144]46]. We used a range of techniques, including western blot, micromass culture, LC3B immunofluorescence, SA-β-gal staining, and transmission electron microscopy to examine the effects of FGF18 and LNP treatment on chondrocyte autophagy. we found that FGF18 mRNA offered protection to chondrocytes from degeneration and senescence by enhancing autophagy. The expression levels of cartilage matrix synthesis-related genes COL2A1 and SOX9 increased by 100%. The expression of BECLIN1 and LC3B II/I ratio was restored, while the expression of the senescence gene P16 decreased by 50%. The proportion of senescent cells decreased by 60%, significantly enhancing the matrix secretion function and autophagosome number in chondrocytes. When we applied FOXO3a knockdown and 3-MA treatment to these cells, the protective effects of FGF18 mRNA were reversed under inflammatory conditions, as seen through SA-β-gal staining and micromass culture (Fig. [145]6F-J). These results suggest that FGF18 mRNA protects cartilage cells from degeneration and senescence by enhancing autophagy. LNP-FGF18 mRNA alleviates the progression of osteoarthritis in both DMM and senile osteoarthritis models To further evaluate the efficacy of LNP-FGF18 mRNA in vivo, we established two distinct osteoarthritis models, i.e. natural senility and DMM. Weekly intra-articular injections of 500ng FGF18 protein or 6 μg LNP-FGF18 mRNA were administered to the knee joint cavity (Fig. [146]7A). Biosafety experiments confirmed that there was no side effect in other organs after injection (Supplementary Fig. [147]9). Von Frey and hotplate experiments confirmed that both FGF18 and LNP-FGF18 mRNA significantly reduced lower limb pain hypersensitivity induced by DMM (Fig. [148]7B, C). However, the LNP-mRNA group has significantly higher Von Frey threshold and response time in hotplate compared to the protein group. Gait analyses revealed a 20% improvement in parameters such as max contact area, print area, mean intensity and duty cycle in mice treated with FGF18 protein or LNP-FGF18 mRNA compared to the DMM or senile groups (Figs. [149]7D and E and [150]8A and B). Fig. 7. [151]Fig. 7 [152]Open in a new tab Rescue of osteoarthritis induced by DMM using FGF18 and LNP-FGF18 mRNA. (A) Experimental flowchart of the animal study. (B, C) Quantitative analysis of Von Frey and hotplate experiments at different time points (n = 15 or 18). (D, E) Representative gait trajectory and quantitative analysis of max contact area, print area, mean intensity, and duty cycle in different mice groups. (F) Representative images of X-ray, micro-CT reconstruction and subchondral bone reconstruction, synovial HE staining, cartilage Safranine O/Fast Green staining, and immunohistochemical staining for Ki67, LC3 and FGF18. Scale bar: 100 μm. (G) Quantitative analysis of subchondral bone BV/TV, trabecular number, thickness, and separation (n = 6 or 7). (H) Quantitative analysis of osteophyte number (n = 6 or 7). (I-L) Quantitative analysis of synovial scores, OARSI scores, cartilage thickness, and area (n = 6 or 7). (M-O) Quantitative analysis of FGF18 (M), Ki67 (N), LC3 (O) positive cells (n = 6 or 7). Data are presented as mean ± SD. P value was calculated by one-way ANOVA, followed by Tukey’s post hoc tests, Kruskal-Wallis test and Mann-Whitney test. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001 Fig. 8. [153]Fig. 8 [154]Open in a new tab Rescue of osteoarthritis symptoms in senile mice by FGF18 and LNP-FGF18 mRNA. (A, B) Representative gait trajectory and quantitative analysis of max contact area, print area, mean intensity, and duty cycle in different mice groups (n = 15 or 18). (C) Representative images of X-ray, synovial HE staining, cartilage Safranin O/Fast Green staining, and immunohistochemical staining for Ki67, LC3, and FGF18. Scale bar: 100 μm. (D-G) Quantitative analysis of synovial scores (D), OARSI scores (E), cartilage thickness (F), and area (G). (H-J) Quantitative analysis of FGF18 (H), Ki67 (I), LC3 (J) positive cells (n = 4 or 6). Data are presented as mean ± SD. P value was calculated by one-way ANOVA, followed by Tukey’s post hoc tests, Kruskal-Wallis test or Mann-Whitney test. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001 Radiological results indicate that intra-articular injections of FGF18 protein and LNP-FGF18 mRNA could reduce subchondral bone sclerosis caused by the DMM model. This treatment results in a decrease in BV/TV and bone trabecular thickness compared to the DMM group. However, the trabecular number remained unchanged after FGF18 or LNP administration (Fig. [155]7F, G). Additionally, the number of osteophytes was reduced by 60% reduction after LNP-FGF18 mRNA injection, superior to 30% reduction after FGF18 protein injection (Fig. [156]7H). Furthermore, we applied HE staining to examine synovitis severity, Safranin O/Fast Green to evaluate the proteoglycan stained by Safranin-O (red) in cartilage matrix, and alcian blue staining to check stain acidic polysaccharides in cartilages. These staining results indicated a significant alleviation of synovitis and cartilage wear in both DMM and senile models after injection with FGF18 and LNP-FGF18 mRNA, with lower synovitis scores and OARSI scores compared to DMM and senile groups. Notably, the LNP-FGF18 mRNA group induced thicker cartilage compared to FGF18 protein group (Figs. [157]7F and I-L and [158]8C-G, Supplementary Figs. [159]10–[160]11). Since we have shown that FGF18 is able to protect cartilage cells from degeneration via enhancing autophagy, we evaluated the expression levels of Ki67 and LC3 in chondrocytes after the injection of FGF18 and LNP-FGF18 mRNA. Local injection of LNP-FGF18 mRNA generate more FGF18 positive cells, increasing by 2-fold in chondrocytes of knee joint in DMM model (Fig. [161]7F, M) and Senile model (Fig. [162]8C, H). FGF18 mRNA group has significantly more LC3 and Ki67 positive cells in DMM model (Fig. [163]7F, N, O) and Senile model (Fig. [164]8C, I, J). Discussion Osteoarthritis (OA) is a degenerative joint disease that is caused by multiple factors. Although many biomolecules have shown therapeutic potential for the treatment of OA, their efficacy is often limited by the depth of penetration and their lifespan in cartilage cavity. In this study, we discovered superior penetrating capability of LNP in cartilage, which can deliver the mRNA to the deeper zone of the cartilage compared to the protein alone. This system is capable of achieving chondrocytes in the deeper zones about 50 μm beneath mice cartilage, which is superior to recombinant proteins by about 5-fold (Fig. [165]2). By optimizing the untranslated region and chemical modification of FGF18 mRNA, we constructed a synFGF18 mRNA delivery system that can stably express therapeutic doses of FGF18 protein and sustain expression in the knee joint cavity of mice for more than 6 days. LNP-FGF18 mRNA can alleviate the degeneration and senescence of chondrocytes induced by inflammatory factors in vitro, while promoting chondrocyte proliferation (Figs. [166]4 and [167]5). Mechanistically, LNP-FGF18 mRNA can promote FOXO3a expression and activate chondrocyte autophagy (Fig. [168]6). We established two OA models, i.e., DMM-induced OA and aging induced OA, to evaluate their efficacy. The injection of LNP-FGF18 mRNA into knee joint cavity of mice significantly alleviated symptoms of osteoarthritis, such as osteophyte formation, cartilage damage, synovial swelling, and decreased pain threshold (Figs. [169]7 and [170]8). Due to the advantages of LNP-FGF18 mRNA over FGF18 recombinant protein, such as greater penetration depth and longer lifespan, our in vivo animal experiments demonstrated that LNPs had superior effects compared to recombinant proteins, including osteophyte number, cartilage thickness, Ki67-positive cell proportion, Von Frey test results, and hotplate test results. These results indicate the clinical potential of LNP-FGF18 mRNA for the OA treatment. Currently, mRNA research focuses on infectious diseases and cancer vaccines [[171]12, [172]47], which only need a small amount of antigen protein to activate the immune response [[173]18]. However, protein replacement therapies, which need recombinant proteins to substitute the deficient protein, high level of protein expression is required, which is 50-1000 times higher than in vaccine applications [[174]13]. Further improvements in the mRNA technology are necessary to reduce its immunogenicity and improve its translation efficiency [[175]48]. We replaced the uridine bases of mRNA with N1-methyl pseudouridine (m1ψ), performed Cap1 capping and Poly(A) tailing on the mRNA and optimized the UTR segments of the mRNA. These optimizations improved the translation efficiency of FGF18 protein by 1000 times compared to the natural FGF18 mRNA. In contrast to the relatively short lifetime of recombinant proteins [[176]9], in vivo bioluminescence results demonstrated that the LNP-mRNA system could locally express the protein in the knee joint cavity for up to one week, thus surpassing the therapeutic efficacy of one-injection FGF18 protein in mouse models. Additionally, the LNP-mRNA system demonstrated good biocompatibility and low immunogenicity, with no significant inflammatory response observed in the mice’s visceral organs or local synovial tissue. Although mRNA requires a carrier to extend its lifetime, the carrier offers the potential to overcome certain drug delivery barriers, such as penetration depth. The dense, negatively charged extracellular matrix (ECM) generates a barrier for traditional drugs to reach the chondrocytes. The collagen fibril network (mostly type II collagen with some type IX and XI collagen) has an approximate pore size of 60–200 nm [[177]10], in consistence with our SEM analyses (Fig. [178]2F), which is comparable to the diameter of our LNP systems. Although the collagen network in aged mice is denser and has smaller pore sizes compared to young mice due to calcification and increased cross-linking of collagen molecules that occurs as a result of AGE (advanced glycation end- products)-formation [[179]49], it remains the same in diameter to prepared LNPs. Our findings suggest that compared to recombinant protein, our LNP system can penetrate deeper into chondrocytes, resulting in superior therapeutic effects. Further development in LNP-mRNA platform, such as lipid structure [[180]50], composition and lipid ratios, will improve the penetration and transfection efficiency. Compared to other disease, knee joint is a sealed cavity, so it has minimal off-tissue side effects. In vivo imaging results have shown that only a small amount of LNP leaks out of the joint cavity and undergoes hepatic metabolism. After 48 h, there is no detectable signal (Supplementary Fig. [181]4). However, in addition to chondrocytes, other cell types such as macrophages, synovial cells, and fibroblasts exist in the knee joint cavity. LNP-FGF18 mRNA system will also enter other types of cells and produce FGF18 protein. However, these proteins face the same issue as recombinant proteins, which are difficult to reach deep cartilage cells. Additionally, FGF18 mRNA does not only acts chondrocytes but also affects other cells. Kuang et al.’s study [[182]26] has shown that the lack of FGF18-FGFR3 signaling can enhance the CXCL12-dependent chemotaxis of macrophages by upregulating CXCR7, thereby exacerbating joint damage in mice. Therefore, the LNP-FGF18 mRNA system may alleviate arthritis by inhibiting macrophage chemotaxis and other mechanisms, which will be investigated in the following work. Previous studies have shown that recombinant FGF18 protein can reverse degeneration, apoptosis, and reduce intracellular ROS production induced by the inflammatory factor IL-1β [[183]36]. However, little attention has been paid to the effect of FGF18 protein on chondrocyte senescence. This study discovered, for the first time, a significant decrease of FGF18 expression in the knee joint cartilage of naturally aging (24-month-old) mice. We further demonstrated that FGF18 protein and FGF18 mRNA can effectively rescue cellular senescence induced by inflammatory factors. Furthermore, we injected the LNP-mRNA into knee joints of mice with naturally aging osteoarthritis. Results showed that FGF18 mRNA not only alleviated DMM-induced post-traumatic osteoarthritis model but also improved local inflammation and senescence in naturally aging osteoarthritis, which simulates the characteristics of most patients in clinical settings. This discovery may expand the use of FGF18 in age-related ailments across multiple systems. In terms of molecular mechanism of FGF18, traditional studies have suggested that FGF18 activates downstream MAPK and Pi3k-Akt pathways by binding to FGFR3, thereby promoting chondrocyte proliferation. However, this cannot explain the anti-inflammatory and anti-senescence effects of FGF18, because MAPK and Pi3k-Akt signaling pathways are already overactivated in the inflammatory and senescence environment [[184]51, [185]52]. This phenomenon suggests a more comprehensive exploration of the FGF18 mechanism of action. Cellular autophagy is a self-protective mechanism in which cells form autophagosomes to encapsulate damaged organelles, which then fuse with lysosomes for degradation, thereby maintaining cellular homeostasis [[186]53]. Autophagy is inhibited in osteoarthritis and senescent fibroblast-like synoviocytes [[187]40, [188]46]. Our study found that addition of LNP-FGF18 mRNA system significantly increased the mRNA level of FOXO3a based on transcriptome sequencing results. FOXO3a is a well-known gene and an upstream gene of autophagy [[189]54, [190]55]. Further experiments using western blotting, immunofluorescence, and transmission electron microscopy confirmed that FGF18 can reactivate the autophagy that is suppressed by inflammation, restoring normal autophagy levels. The increase autophagosomes engulf aging and damaged organelles, thereby alleviating cellular inflammation and aging. In conclusion, this study developed the LNP-FGF18 mRNA system to penetrate the cartilage and stably express FGF18 protein in chondrocytes. This system promotes the expression of FOXO3a protein that induces autophagy, ultimately reducing inflammation and senescence. The LNP-FGF18 mRNA system can slow down the occurrence and progression of osteoarthritis both in vitro and in vivo. This strategy holds promise as a new therapeutic strategy for OA. Methods Human knee cartilage specimen collection The sample collection procedure was approved by the ethics committee of Shanghai Ninth People’s Hospital affiliated to Shanghai Jiaotong University School of Medicine (SH9H-2021-T401-4). Human cartilage samples were collected from patients who provided signed informed consent. During total knee arthroplasty (TKA) procedures, samples were collected from human tibial plateaus. Osteochondral plugs with a size of 0.5 cm × 0.5 cm were drilled from the damaged (OA) or intact (relatively healthy) parts of the tibial plateau. These removed plugs were then fixed in 4% paraformaldehyde for subsequent histological assessment using Safranine O/Fast Green staining and FGF18 immunohistochemistry. Chemically modified mRNA (mRNA) synthesis and formulation mRNA encoding FGF18, Fluc and EGFP gene were synthesized with a T7 RNA Polymerase in vitro transcription (IVT) kit bought from Yeasen, China (10673ES). The UTR information was listed in the Supplementary Table [191]2. During mRNA transcription, uridine was replaced by N1-methyl pseudouridine (m^1ψ). IVT reaction details, capping and tailing of mRNA and subsequent purification of mRNA was performed according to the protocols of kits bought from Yeasen, China (10673ES) and Beyotime, China (R7075). Concentration of mRNA were quantified by Nanodrop spectrophotometers (Thermo Fisher Scientific, USA). A chimeric UTR is designed based on the globin and gp130 associated protein, which is named as synUTR. Lipid nanoparticles production and characterization Lipid nanoparticles were composed of four different lipids including SM-102, 1,2-distearoyl-sn-glycero-3-PC (1,2-DSPC), cholesterol and 1,2-dimyristoyl-rac-glycero-3-methoxypolyethylene glycol 2000 (DMG-PEG2000) bought from Cayman Chemical (LNP-102, USA) at lipid molar ratios of 50:10:38.5:1.5 respectively, dissolved in absolute ethanol. mRNA was dissolved in 50mM sodium acetate, pH 5.0, and mixed with ethanolic lipid mixture with an ethanol: aqueous ratio of 1:3. Mixing procedure was performed with a microfluidic device with a Y-channel design. After mixture, LNPs were dialyzed in neutral buffer against 1000 volumes of buffer overnight. Encapsulation efficiency was more than 90% for all LNP-mRNAs in this study determined by Ribogreen. Particle size and zeta potential of LNPs were evaluated using dynamic light scattering (DLS) instrument from Malvern Instruments Ltd. (ZS-90) and Bettersize Instrument (BeNano 90). A certain amount of sample was dispersed in deionized water (0.1% w/v), followed by sonication for 3 min. The measurements were performed three times, and the average values were calculated. Cryo-transmission electron microscopy (Cryo-TEM), TEM and scanning electron microscopy (SEM) analyses For cryo-TEM, a polymer-coated carbon-reinforced copper grid was treated with a small drop (approximately 3 μl) of mRNA-LNPs solution for 1 min, blotted for 5 s, and swiftly plunged into liquid ethane cooled by liquid nitrogen. This process was carried out at 8 °C and 100% humidity using a Vitrobot Mark IV from FEI, Thermo Fisher Scientific. The grids were subsequently imaged with a Talos Arctica system at 200 kV, utilizing a Falcon 3EC detector and EPU for data collection. For TEM, the samples were fixed with 2.5% glutaraldehyde and kept at 4 °C for 12–24 h. Subsequently, fixed samples were dissolved in 1% osmium tetroxide for 1–2 h and washed three times with PBS for 15 min each time. The samples were then dehydrated and infiltrated with a gradient concentration of acetone for embedding. The resin blocks were cut into 70–90 nm ultrathin sections using an ultramicrotome, followed by retrieval of the sections using copper grids. Sections were stained with uranyl acetate for 8–10 min and lead citrate for 8–10 min. After drying, the samples were observed and images were captured for analysis using a transmission electron microscope. For SEM, samples were fixed, dehydrated and coated with 10 nm of gold before SEM scans (Gemini300, Carl Zeiss). Cartilage infiltration assay Recombinant FGF18 was labeled by Cy3 (BRC-15, Conflure Biology) with protocol “NHS Ester Labeling of Amino-Biomolecules, [192]www.lumiprobe.com”. In short, Cy3 NHS ester was mixed with FGF18 in a 0.1 M sodium bicarbonate buffer and incubated in dark for 8 h. LNP was labeled with Dil (MedChemExpress, HY-D0083) solution with a concentration of 100 μM and incubated for 8 h in the dark. Cy3-FGF18 and Dil-LNPs were purified using size exclusion chromatography columns to remove free dyes (Thermo Fisher Scientific, 89882). For cartilage infiltration assay, Cy3-FGF18 and Dil-LNPs were intra-articularly injected to young (3 m) and old (18 m) mice. Knee joints were collected after 24 h for subsequent frozen sections. Nuclei was stained with DAPI and images were taken with a confocal microscopy. Animals All animal experiments were conducted in accordance with the guidelines approved by the ethics committee of the Shanghai Ninth People’s Hospital affiliated to Shanghai Jiaotong University School of Medicine (SH9H-2023-A856-1). C57BL/6J mice of wild-type were obtained from the animal center of Shanghai Ninth People’s Hospital. The mice were housed in specific pathogen-free (SPF) cages under standard conditions, with a temperature of 24℃, a humidity of 60%, and a 12-hour light-dark cycle. They were provided with a standard diet. For the DMM surgery, 10-week-old C57BL/6J mice were selected and anesthetized with isoflurane. The right knee joint was exposed, and the medial meniscotibial ligament was transected to induce destabilization of the medial meniscus, following a previously established protocol [[193]56]. 500ng FGF18 protein (20 μL, 25 μg/mL) or 6 μg LNP-FGF18 mRNA (20 μL, 300 μg/mL) was injected intraarticularly in the right knee once a week after surgery. After eight weeks post-surgery, the mice were euthanized, and the right knee joint was collected. The joints were fixed in 4% paraformaldehyde (PFA) for 48 h and then transferred to 75% ethanol for subsequent micro-computed tomography (micro-CT) and histological analysis. For the senile osteoarthritis (OA) model, C57BL/6J mice were fed a normal diet and allowed to age until 24 months, with 3-month-old mice serving as controls. 500 ng FGF18 protein (20 μL, 25 μg/mL) or 6 μg LNP-FGF18 mRNA (20 μL, 300 μg/mL) was injected intra-articularly in knee joints of 18-month-old mouse once a week for two months. Right lower extremities were collected and fixed for further radiological and histological analysis. Gait analyses Gait analyses was evaluated with the CatWalk gait analysis system (Noldus Technology, Netherlands) after eight weeks post-surgery or senile mice at 24-month-old [[194]57]. Mice were pretrained to get used to crossing the walkway for thirty minutes before gait recording. The recordings were carried out in a dark environment. Gait analysis was recorded using a video-based system. The paw, upon contacting the glass plate, reflected green light to a video camera, which recorded the entire run at a frequency of 100 Hz. The same investigator conducted the entire gait analysis experiments with blindness to grouping. Both hind and front paws were assessed for data analysis. Compliant runs were defined with a maximum speed variation of 60%. Runs with backing, sniffing, cleaning, or long stops were excluded. Three compliant runs were required for statistical analysis. CatWalk software will automatically label all areas and identify and assign them to the respective paw. The following parameters were analyzed: max contact area, print area, mean intensity, and duty cycle (percentage of stance phase in a step cycle). Von-frey test and hotplate test The Von Frey test was conducted to measure the pain threshold in the hindlimbs of mice using an electronic Von Frey filament (Xinruan Technology, China). Prior to the test, each mouse was allowed to acclimate to the test chamber for 30 min. A positive response was recorded if the animal exhibited any nociceptive behavior, such as paw withdrawal, shaking, or licking. The threshold force exerted by the electronic filament was recorded, with at least three replicative forces measured for each mouse in each test. The Von Frey test was performed every two weeks after the DMM surgery. For hotplate test, mice were subjected to the hotplate test at a temperature of 55 °C. The response time was recorded as the duration between the hindlimbs touching the plate and response behaviors such as paw shaking, paw licking, or jumping. The hotplate pain assay was performed every two weeks following the DMM surgery. At least three replicative response times were recorded for each mouse. Both Von Frey or hotplate tests were conducted by an observer who was blinded to the grouping of mice. Micro-CT Knee joints from mice were fixed for two days and then scanned using a high-resolution micro-CT scanner (Skyscan 1072, Belgium) with a pixel size of 9 μm, a 55 kVp source, and a 145 μAmp current. Osteophyte volume, bone volume/tissue volume (BV/TV), trabecular number (Tb. N), trabecular thickness (Tb. Th) and trabecular separation (Tb. Sp) of subchondral bone were calculated with CTAn software (Bruker, Germany). Three-dimensional reconstructions of subchondral bone sections were produced with CT-Vox software (Bruker, Germany), as described in our previous study [[195]58]. Histological analysis For histologic analysis, knee joints and human specimens were fixed, decalcified, dehydrated, and embedded into paraffin. Serial tissue sections of a thickness of 5 μm were cut in a sagittal or coronal plane. Safranine O/Fast Green staining was performed to assess cartilage degeneration of tibial and femur cartilage using the Osteoarthritis Research Society International (OARSI) scoring system [[196]59]. Hematoxylin and eosin (HE) staining was also performed to evaluate synovial inflammation using the synovitis score [[197]60, [198]61]. For immunohistochemistry (IHC) staining, the sections were incubated with a primary antibody against FGF18 (11495-1-AP, 1:200, Proteintech), Ki67 (1:200, ab16667, Abcam) and LC3 (14600-1-AP, 1:100, Proteintech). Images were captured using a Zeiss DM4000B high-resolution microscope, and quantitative analysis was performed using the ImageJ software. Positive cells were counted by two different researchers (KK and BL). Average number of positive cells calculated by the two researchers was used as the final count of positive cells. This number was then divided by the total number of cells in the section to determine the proportion of positive cells. In vivo bioluminescence On days 1, 2, 3, 4, and 6 after injecting LNP-Fluc mRNA into the right knee joint cavity of C57BL/6J mice, bioluminescent imaging was performed with an IVIS Spectrum system (PerkinElmer, USA). To prevent interference from dark-colored fur, the hair on the lower limbs of the mice was removed. D-luciferin was diluted in phosphate-buffered saline (PBS) and injected intraperitoneally at a dose of 150 mg/kg. After 20 min, the mice were anesthetized using isoflurane. Photon emissions from the right lower limb were collected with a 1-minute exposure time. Luminescent signal was overlaid onto the mouse photo, and the regions of interest were delineated using Living Image software (PerkinElmer, USA) to calculate the signal flux within the regions. For assessing the metabolic pathway of LNP-mRNA, bioluminescent imaging was performed on mice 24 and 48 h after injecting LNP-Fluc mRNA into the knee joint cavity of C57BL/6J mice. D-luciferin was injected intraperitoneally at a dose of 150 mg/kg. After 20 min, the mice were euthanized, and their hearts, livers, spleens, lungs, and kidneys were collected and imaged using the IVIS imaging system with a 1-minute exposure time. Living Image software was used to detect the signal flux within the regions of interest. Cell culture and reagents Murine primary chondrocytes were isolated from newborn C57BL/6J mice following a previously established protocol [[199]62]. Primary chondrocytes were cultured in Dulbecco’s Modified Eagle Medium/Nutrient Mixture F12 (DMEM/F12) with 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin. The ATDC5 cell line, an immortalized mouse chondrocyte cell line, and HEK293T cells were obtained from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China). ATDC5 chondrocytes were cultured in DMEM with 4.5 g glucose/L, 5% FBS, and 1% penicillin-streptomycin. 293T cells were cultured in DMEM with 4.5 g glucose/L, 10% FBS, and 1% penicillin-streptomycin. IL-1β and TNF-α protein was purchased from Genscript. Recombinant human FGF18 protein was purchased from Sino Biological (13206-H08H). 3-Methyladenine (3-MA) was purchased from TargetMol (T1879). Lipofectamine 3000 was purchased from Thermo Fisher Scientific, United States. 10 ng/mL IL-1β, 10 ng/mL TNF-α and 5 mM 3-MA were used to incubate chondrocytes. Dot blot Dot blot assay was used to evaluate the protein expression of FGF18 in cell culture supernatants. After transfecting ATDC5 or 293T cells with LNP or plasmid for 24 h, cell culture supernatants were collected and centrifuged at 3000 rpm for 15 min to obtain the supernatant. Then, 30 μl of the supernatant was spotted onto a nitrocellulose membrane, and 10 μg and 1 μg of rhFGF18 were spotted as positive controls. After complete absorption of the liquid on the membrane, it was blocked with 5% BSA (Bovine Serum Albumin) for 60 min. After washing three times with TBST (tris-buffered saline and Tween 20), the membrane was incubated overnight at 4 °C with the primary antibody against FGF18 (1:1000, 11495-1-AP, Proteintech). On the following day, after washing three times with TBST, the membrane was incubated with the corresponding fluorescent secondary antibody at room temperature for 2 h. The membrane was then visualized using the Odyssey V3.0 image scanner (Li-COR. Inc., Lincoln, NE, USA). The fluorescence intensity of the supernatant dots and the rhFGF18 dots were measured using ImageStudio Lite software (Li-COR) to determine the expression level of FGF18 protein in the supernatant. Luciferase reporter gene assay Cells were seeded in a black-bottom 96-well plate (BS-MP-96B, Biosharp) and transfected with different concentrations of LNP-Fluc mRNA. At different time points after transfection (24 h, 48 h, 72 h), the firefly luciferase activity was measured using the firefly luciferase reporter gene assay kit (RG051, Beyotime). 100 μl of the assay reagent was added to each well of the 96-well plate and incubated at room temperature for 5 min. Subsequently, chemiluminescent signals were detected using a multifunctional microplate reader. Plasmid and lentivirus transfection FGF18 plasmids with different UTRs were synthesized by Genecefe Biotechnology Inc. China. Sh-FOXO3a lentivirus was purchased from Obio technology, China. Plasmid was transfected with the protocol of Lipofectamine 3000 and the protein expression was detected at 48 h after transfection. For lentiviral infection, we added lentivirus at a multiplicity of infection (MOI) of 10 and incubated the cells. After 48 h of infection, the media was replaced, and the successfully transfected cells were selected using puromycin to establish a stable FOXO3a knockdown cell line. Micromass culture After digestion, centrifugation, and suspension, a suspension of ATDC5 chondrocytes was prepared at a concentration of 1.5 × 10^^7 cells per mL. Then, 10 μL of the cell suspension was added to the center of each well in a 24-well culture plate. The plate was incubated in a cell incubator for two hours to allow cell attachment. 1 mL of cell medium containing 1% insulin-transferrin-selenium (ITS) (41400045, Thermo Fisher) and either 100 ng of FGF18 protein or 3 μg of LNP-FGF18 mRNA was gently added to each well. Cell medium was changed every two days. After different time periods, the cell masses were fixed and stained with Alcian blue or toluidine blue dyes for 30 min. The integrated optical density (IOD) value of each micromass was calculated using Image J software. Cell toxicity To test the toxicity of the LNP-mRNA system on cells, we used cell counting kit-8 (cck8) (Dojindo, Japan) and calcein/pi staining kit (C2015, Beyotime, China) to test the toxicity of LNP. In the cck8 experiment, we treated cells with different concentrations of LNP system for 24 h, 48 h, and 72 h. Then, at different time points, we added 10% cck8 reagent (Dojindo, Kumamoto, Japan) to the cells and incubated them in a cell incubator for 2 h. Finally, an Infinite M200 pro multimode microplate reader (Tecan Life Sciences, Switzerland) was used to detect the absorbance at 450 nm of each well. For calcein/pi staining, we added calcein/pi working reagent to cells and incubated them at 37 °C in the dark for 30 min. Then calcein and pi positive cells were observed and quantified under a laser confocal microscope. EdU assay EdU assay was utilized to assess cell proliferation by incorporating the thymidine analogue, EdU (5-ethynyl-2’-deoxyuridine), into DNA replication. The EdU assay protocol followed the instructions provided with the kits purchased from Beyotime, China (C0075). Primary chondrocytes were incubated with a 10 μM EdU working solution at 37 °C for 2 h to label the cells with EdU. Following this, the cells were fixed, washed, and permeabilized. To prepare the click reaction solution, we combined components such as click reaction buffer, CuSO4, azide 555, and click additive solution. The prepared Click reaction solution was then added to the cells and incubated in the dark for 30 min. Finally, we captured representative images using a laser confocal microscope and further quantitative analysis was done with Image J. Senescence-associated-β-galactosidase (SA-β-gal) staining Cellular senescence was tested with a SA-β-gal staining kit bought from Beyotime, China (C0602). Primary chondrocytes were cultured with FGF18, LNP-FGF18 or IL-1β for 7 days. The SA-β-gal staining procedure was performed following the instructions provided with the kit. Random microscopy fields were selected for image acquisition. ImageJ software was utilized to count the number of β-gal staining-positive cells for subsequent quantitative analysis. Immunofluorescence (IF) For cell immunofluorescence, we treated ATDC5 chondrocytes with different drugs for 24 h. Then, we fixed, permeabilized, blocked, and washed the cells. Next, we incubated the cells with the corresponding primary antibodies overnight at 4 °C. After that, we incubated the cells with Alexa Fluor-conjugated secondary antibodies (1:1000) at room temperature for 1 h and stained the cell nuclei with DAPI. Finally, we observed and captured representative images under a laser confocal microscope and quantified the number of positive cells using ImageJ software. COL2A1 antibody (1:100, ab34712), SOX9 antibody (1:100, ab185230), and Ki67 antibody (1:200, ab16667) were all purchased from Abcam. Western blot After treating cells with different conditions for 24 h, we lysed the cells using RIPA lysis buffer (P0013, Beyotime) supplemented with 1% protease/phosphatase inhibitor and purified and quantified cell proteins through centrifugation and bicinchoninic acid assay (BCA). 20 μg protein was loaded into each well of SDS-PAGE gel, and Tris-Glycine was used as the electrophoresis buffer. After electrophoresis at 140 V for 1 h, protein was then transferred to a PVDF membrane and blocked with a 5% BSA solution. Subsequently, the membrane was incubated with the corresponding primary and secondary antibodies and the protein bands were imaged using an Odyssey V3.0 image scanner (Li-COR. Inc., Lincoln, NE, USA). COL2A1 antibody (1:1000, ab34712), SOX9 antibody (1:1000, ab185230), MMP3 antibody (1:1000, ab52915), and MMP13 antibody (1:1000, ab39012) were purchased from Abcam. β-actin antibody (4970, 1:2000), BECLIN1 antibody (3495, 1:1000), FOXO3a antibody (12829, 1:1000), and LC3B antibody (43566, 1:1000) were purchased from CST. FGF18 antibody (11495-1-AP, 1:1000) were purchased from Proteintech. RNA sequencing Chondrocytes were seeded in six-well plates at a density of 4 × 10^6 cells per well. After 24 h of incubation with 10 ng/mL TNF-α (C600052, Sangon) or 3 μg/mL LNP-FGF18 mRNA, total RNA was extracted using the Total RNA Extraction Kit (R6812-01HP, Omega, United States). RNA sequencing was performed by Wuhan Huada Gene Technology Co., Ltd. (China). Gene Ontology (GO) enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, and gene set enrichment analysis (GSEA) were conducted using the Mybgi platform (Wuhan Huada Gene Technology, [200]https://mybgi.bgi.com/tech/login). Heatmap analysis was performed in GraphPad Prism 8.0 (GraphPad Software Inc., San Diego, CA, USA) based on the Z-score of TPM counts for each gene. Quantitative real-time PCR (qRT-PCR) Total RNA was extracted from cells using a Total RNA Extraction Kit (R6812-01HP, Omega, United States). 1000 ng cDNA was synthesized with the aid of a cDNA synthesis kit (RR036A, Takara, Japan), diluted in 200 μL of ddH[2]O. A total of 10 μL of the qRT-PCR mix, consisting of 3.6 μL of double distilled H[2]O, 1 μL of cDNA, 0.2 μL of upstream and downstream primers respectively, and 5 μL of SYBR premix ([201]B21202, Selleck, USA), was added to 384-well plate. qRT-PCR process was performed on an ABI 7500 Sequencing Detection System (Applied Biosystems, USA). The primer sequences used for the target genes are listed in Supplementary Table [202]1. Statistical analysis Statistical analyses were conducted using SPSS version 25.0 (SPSS Inc., Chicago, IL, USA). For data that followed a normal distribution, comparisons between two groups were performed using the unpaired Student’s t-test. One-way ANOVA followed by Turkey’s post hoc tests were used for comparisons among three or more groups. Non-normally distributed data were analyzed using the Mann-Whitney and Kruskal-Wallis tests. GraphPad Prism 8.0 (GraphPad Software Inc., San Diego, CA, USA) was utilized for the design and creation of statistical charts. All P values reported in this study are two-sided, and statistical significance was defined as P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. Electronic supplementary material Below is the link to the electronic supplementary material. [203]Supplementary Material 1^ (5.5MB, docx) Acknowledgements