Abstract Mesenchymal’ stem cells (MSC) are widely used for transplantation to treat various diseases due to their strong immune regulatory and tissue repair abilities. Magnetic nanoparticles (MNPs) can be used to track transplanted MSC. However, the potential impact of the MNPs we developed on MSC function remains unclear. In this study, we treated MSC with poly-L-lysine (PLL)-modified MNPs (MSC-MNPs) at a concentration of 0.1 µg/µL to assess their effects on MSC. The results showed that there were no significant effects on cell morphology, differentiation potential, proliferation, apoptosis, or the cell cycle after MSC were treated with MNPs. Interestingly, further experiments revealed that MNPs significantly enhanced the migratory capacity of MSC. Bio-Plex analysis revealed that MNPs promoted the expression of anti-inflammatory cytokines while inhibiting the secretion of pro-inflammatory factors. Flow cytometry also detected a significant increase in the number of MSC subpopulations, including CD184⁺MSC, CD106⁺MSC, and CD55⁺MSC, after MNPs labeling, which was further supported by proteomic analysis. Moreover, MSC-MNPs promoted the phenotypic transition of reactive astrocytes from A1 to A2 after coculture with activated astrocytes. In conclusion, MNPs have no cytotoxic effects on MSC labeling and significantly enhance their anti-inflammatory functions, offering new possibilities for the clinical application of MSC. Supplementary Information The online version contains supplementary material available at 10.1038/s41598-025-13083-3. Keywords: Mesenchymal stem cells, Magnetic nanoparticles, Migration, Cytokines, Anti-inflammatory Subject terms: Immunology, Neuroscience, Stem cells, Nanoscience and technology Introduction Personalized therapies utilizing allogeneic or autologous cells are becoming increasingly common in the medical domain^[32]1–[33]3. Notably, mesenchymal stem cells (MSC) have been extensively employed in the treatment of various diseases because of their broad availability, ease of procurement, low immunogenicity, robust proliferation capacity, and roles in immune regulation, anti-inflammation and tissue repair^[34]4,[35]5. These unique properties make them highly promising candidates for the treatment of autoimmune diseases and central nervous system injuries^[36]6. Nevertheless, the clinical application of cell transplantation therapies is challenging: At present, it is difficult to noninvasively monitor the migration, homing, implantation efficiency and functional ability of transplanted cells in vivo. In recent years, magnetic nanomaterials (MNPs) have been increasingly used in the biomedical field. MNPs are renowned for their magnetism, biocompatibility, and biodegradability, and have a variety of potential applications, such as drug delivery, specific cell labeling, and magnetic resonance imaging (MRI), especially for cell tracking via MRI^[37]7,[38]8. The distinctive physicochemical properties of MNPs confer considerable advantages in medical imaging. Labeling mesenchymal stem cells with MNPs and conducting magnetic resonance imaging detection can significantly enhance the success rate of stem cell transplantation. Currently, histopathological techniques are limited in providing information about the fate of implanted cells after animal euthanasia or via biopsy surgery. To facilitate real-time tracking of the cellular position, viability, and functional status, recent advancements have introduced imaging techniques that involve the internalization of MNPs by cells^[39]9. These internalized MNPs function as imaging agents and are monitored via MRI. The utilization of MRI for cell tracking presents several significant advantages, including high spatial resolution, excellent anatomic background contrast, and the absence of ionizing radiation exposure^[40]10. Additionally, MRI enables long-term cell tracking, with the ability to monitor cells over periods of months. Some MNPs have already been employed to investigate the fate of labeled cells in animal models and specific clinical contexts^[41]11–[42]13. For instance, fluorescent magnetic nanoparticles have been used to label B10 cells, facilitating continuous magnetic resonance imaging for up to four weeks post-injection and transplantation to assess the survival and differentiation of labeled cells. Current research indicates that cells labeled with MNPs generally exhibit non-cytotoxic behavior and do not affect cell proliferation and differentiation abilities^[43]14. However, a few studies have reported that MNPs-labeled stem cells may partially lose their ability to differentiate in a concentration-dependent manner^[44]15. Therefore, before utilizing MNPs-labeled MSC, it is imperative to confirm that these cells continue to fulfill the established criteria for MSC and to evaluate whether MNPs have any impact on the biological functions of mesenchymal stem cells. MSC show great therapeutic potential in regenerative medicine^[45]16,[46]17. The International Society for Cell Therapy has established the following minimum criteria for MSC^[47]18: (1) MSC must adhere to uncoated plastic culture dishes under standard culture conditions; (2) MSC must simultaneously express CD105, CD73, and CD90, but not CD45, CD34, CD14, CD11b, CD79, CD19, or lineage markers such as HLA-DR; (3) MSC must be able to differentiate into osteoblasts, adipocytes, and chondrocytes in vitro. MNPs-labeled MSC must adhere to the aforementioned standards. The biological function of MSC may be influenced by the size, concentration, particle shape and coating type of MNPs^[48]15,[49]19,[50]20. Therefore, the impact of each type of MNPs on the biological functions of MSC should be assessed prior to their application. Currently, research on the biological effects of MNPs on MSC primarily focuses on aspects such as cell viability, proliferation, apoptosis, migration, and cell cycle regulation. However, in central nervous system diseases, transplanted MSC exert their effects mainly by secreting inflammation-related factors to improve the inflammatory microenvironment^[51]21. Yet, whether the uptake of MNPs by MSC affects the secretion of inflammatory cytokines and the underlying mechanisms remains unknown. In this study, we treated MSC with PLL-modified MNPs and comprehensively evaluated the effects of MNPs on the biological functions of MSC, including cell viability, cell phenotype, directed differentiation, proliferation, apoptosis, cell cycle, migration, and cytokine secretion. Our results indicate that PLL-modified MNPs can not only serve as tracers for biological imaging of MSC, but also significantly enhance the migration and anti-inflammatory capabilities of MSC. This dual functionality can greatly improve the transplantation success rate and overall functional performance of MSC. Results MNPs did not affect the differentiation potential or the expression of intrinsic surface markers of MSC Our previous studies demonstrated that MSC transplantation can effectively promote spinal cord injury repair^[52]21. However, the survival and migration of transplanted MSC cannot be tracked in vivo. Magnetic nanomaterials, known for their excellent biocompatibility, have been widely used in the medical field, particularly as tracers in clinical applications^[53]22. MNPs possess strong magnetic resonance imaging capabilities and can effectively track cells after being ingested by MSC^[54]13,[55]23. Nevertheless, the potential effects of MNPs on MSC need to be thoroughly tested before their clinical use. We developed zinc-doped magnetic nanoparticles (Zn[0.4]Fe[2.6]O[4]), which exhibited a cubic shape with an average diameter of approximately 60 nm (Fig. [56]1A). These MNPs were modified with poly-L-lysine (PLL) to enhance their biocompatibility. The PLL-modified MNPs (PLL-MNPs) exhibited a high saturation magnetization of 76 emu g⁻¹ (Fig. [57]1B), indicating their rapid response to external magnetic fields. After surface modification, the MNPs showed excellent hydrophilicity and dispersibility. The hydrodynamic diameters of the MNPs and PLL-MNPs were 490 ± 50 nm and 253 ± 10 nm, respectively (Fig. [58]1C). The zeta potentials of the MNPs and PLL-MNPs were − 6.14 ± 0.32 mV and 34.82 ± 0.52 mV, respectively (Fig. [59]1D). The PLL coating significantly improved the dispersibility of the MNPs through electrostatic repulsion and steric hindrance generated by the high density of amino groups. Additionally, PLL provided MNPs with the ability to interact with MSC. Fig. 1. [60]Fig. 1 [61]Open in a new tab Characterization of MNPs and identification of MSC. (A) Morphology of MNPs under TEM. (B) Curve displaying the magnetization of dry MNPs in the magnetic field. (C, D) Hydrodynamic diameter and zeta potential of MNPs and MNPs-PLL (n = 3). (E) Morphology of MSC and MSC-MNPs. (F) MSC-MNPs can induce differentiation into bone, cartilage and fat through alizarin red, alcian blue, oil red O staining. (G, H) Identification of the expression of MSC markers via flow cytometry. In accordance with the International Society for Cellular Therapy (ISCT) criteria for MSC identification, we assessed MSC that had ingested MNPs. We found that the morphology of MSC did not significantly change after uptake of PLL-MNPs (MSC-MNPs, 0.1 µg/µL, Fig. [62]1E). Moreover, the capacity of the MSC-MNPs for lipogenesis, osteogenesis, and chondrogenesis was the same as that of the untreated MSC(Fig. [63]1F). We also examined cell surface markers and found that MSC-MNPs expressed CD90 and CD105, but did not express CD34, CD11b, CD19, CD45, or HLA-DR, similar to untreated MSC(Fig. [64]1G, H). These results indicated that MNPs uptake by MSC does not affect their differentiation potential or the expression of intrinsic cell surface markers. The migration ability of MSC was enhanced after MNPs uptake Normally, tracers are not expected to affect normal cell functions. To determine whether the uptake of MNPs by MSC has any effect on their function, we examined cell proliferation, apoptosis, the cell cycle, and migration. Prussian blue and Eosin staining revealed that iron oxide-containing blue nanoparticles were taken up by MSC (Fig. [65]2A). We assessed cell proliferation via the CCK-8 assay and found no significant differences between the MSC-MNPs group and the MSC group at 6,12,24,48 h (Fig. [66]2B). These findings suggest that the uptake of MNPs by MSC did not induce cytotoxicity. To investigate whether MNPs influence cell apoptosis, we performed FACS analysis on MSC treated with MNPs for 24 h. There were no significant differences in PI and Annexin V double-positive populations between the MSC-MNPs group and the MSC group (Fig. [67]2C, D), indicating that MNPs uptake did not affect cell apoptosis. We also examined the cell cycle distribution of MSC after 24 h of MNPs treatment. Similarly, no significant changes were observed between the MSC-MNPs group and the MSC group (Fig. [68]2E, F). Subsequently, we utilized DCFH-DA assays to detect reactive oxygen species (ROS). Our findings indicated that MNPs did not augment ROS production, as evidenced by both immunofluorescence staining and flow cytometry (Supplementary materials Figure [69]S1). Interestingly, to assess whether MNPs are involved in MSC migration, we conducted a wound scratch assay. After 12 h, the coverage of migrating MSC in lesion areas was significantly greater in the MSC-MNPs group than in the MSC group (Fig. [70]2G, I). In the transwell assay, cultured macrophages were stimulated with LPS to produce chemokines in the lower chamber, while MSC or MSC-MNPs were seeded in the upper chamber. Crystal violet staining showed that a greater number of MSC migrated through the membrane pores in the MSC-MNPs group than in the MSC group (Fig. [71]2H, J). These results suggest that the uptake of MNPs promotes the migration of MSC. Fig. 2. Fig. 2 [72]Open in a new tab Effects of MNPs uptake by MSC on cell proliferation, apoptosis, the cell cycle and migration. (A) Prussian blue and Eosin staining revealed MNPs uptake by MSC. All MSC in the MSC-MNPs group were blue. (B) The proliferation of MSC after uptake of MNPs was detected via CCK-8 assay. (C, D) Using annexin V and propidium iodide (PI) staining, FACS analysis revealed a comparable percentage of apoptotic cells in the MSC-MNPs group and the MSC group. (E, F) FACS analysis of PI-stained cells showed no significant change in the cell cycle in the MSC-MNPs group or the MSC group. (G, I) MSC migration was studied via a wound scratch assay. At 12 h after scratching, the coverage of migrating MSC in lesion areas was significantly greater in the MSC-MNPs group than in the MSC group. (H, J) Transwell assays showed that more MSC migrated through the membrane pores in the MSC-MNPs group than in the MSC group. The data are represented as the mean ± SEM. *P < 0.05, **P < 0.01, Two-way repeated measures ANOVA with Bonferroni’s post hoc correction in B and Student’s t-test in others, n = 3 in B, F, I, J; n = 5 in D. MNPs uptake by MSC promoted their anti-inflammatory function and affected the number of cell subtypes related to migration and immune regulation To investigate the effects of MNPs uptake on the anti-inflammatory function of MSC, we collected the supernatant from MSC after 24 h of MNPs treatment and measured the cytokine levels using the Bio-plex assay (Fig. [73]3A). Compared with those in the MSC group, we observed significant decreases in TNF-α and MCP-1 in the MSC-MNPs group (Fig. [74]3A, B; MSC and MSC-MNPs in pg/ml: 271.4 ± 12.83 and 150.7 ± 8.708 for TNF-α, 115.1 ± 6.836 and 85.46 ± 5.722 for MCP-1, P < 0.01 and P < 0.05, respectively). Conversely, the levels of the anti-inflammatory cytokines IL-10, IL-13, RANTES and VEGF were significantly increased in the MSC-MNPs group (Fig. [75]3A, B; MSC and MSC-MNPs in pg/ml: 14.55 ± 0.98 and 24.39 ± 1.33 for IL-10, 47.62 ± 1.546 and 74.25 ± 7.341 for IL-13, 41.97 ± 3.13 and 63.51 ± 6.49 for RANTES, 14.66 ± 1.86 and 22.88 ± 0.95 for VEGF, P < 0.01, P < 0.05, P < 0.05, P < 0.05). These findings suggest that MNPs uptake by MSC promote the secretion of anti-inflammatory factors while reducing the secretion of pro-inflammatory factors. Fig. 3. Fig. 3 [76]Open in a new tab MNPs affect the cytokine secretion and the number of cell subsets of MSC. (A) Heatmap of the Bio-plex analysis showing the relative expression of 27 cytokines in the MSC and MSC-MNPs groups at 24 h after MNPs treatment. (B) Quantitative analysis indicates that, compared with the MSC group, the level of TNF-α and MCP-1 decreased, and the level of IL-10, IL-13, RANTES and VEGF increased in the MSC-MNPs group. (C-E) FACS analysis showed that the numbers of CD184^+ MSC, CD106^+ MSC and CD55^+MSC were significantly greater in the MSC-MNPs group than in the MSC group. *P < 0.05; **P < 0.01; Student’s t-test, n = 3 in each group. Single-cell sequencing revealed the presence of distinct functional subgroups of MSC^[77]24,[78]25. Specifically, CD184⁺ MSC exhibited stronger migration capabilities, facilitating cell movement to injury sites. The CD106⁺ MSC subgroup expressed high levels of immunosuppressive factors, regulated cytokine secretion, and demonstrated robust immunomodulatory abilities. CD55, a complement activation suppressor, protected MSC from cell death caused by complement activation. Given that MNPs uptake by MSC enhances migration and anti-inflammatory cytokine secretion, we used flow cytometry to detect the expression of CD184, CD106, and CD55 on the surface of MSC. Under normal conditions, MSC express low levels of CD184 and CD55, with only a small amount of CD55 detected. After MNPs uptake, the number of CD184⁺, CD106⁺, and CD55⁺ subsets significantly increased (MSC and MSC-MNPs percentage of cell subtypes: 0.8800 ± 0.1818 and 7.367 ± 1.691 for CD184⁺, 3.81 ± 0.53 and 13.20 ± 2.65 for CD106⁺, 2.39 ± 1.07 and 30.40 ± 5.39 for CD55⁺, P < 0.05, P < 0.05, P < 0.01, Fig. [79]3C-E). MNPs uptake by MSC alters proteomic profiles To further explore the potential molecular mechanisms by which MNPs affect cytokine secretion and the number of cell subsets of MSC, we performed data-independent acquisition (DIA) proteomics using protein extracts from cultured MSC treated with or without MNPs. We identified 237 differentially expressed proteins (DEPs; FDR < 0.05, |FoldChange|>1.5) between the MSC and MSC-MNPs groups, including 93 upregulated and 144 downregulated DEPs (Fig. [80]4A). The expression of multiple proteins related to the cytoskeleton and axon orientation was significantly modified in the MSC-MNPs groups. These include RhoU GTPase (RHOU), Slit Guidance Ligand 2 (SLIT2), and Slit Guidance Ligand 3 (SLIT3). Additionally, the upregulation of tissue repair-related proteins, such as Midkine(MDK), Hepatocyte Growth Factor Activator(HGFAC); cell migration related proteins, such as Wnt Family Member 5 A(WNT5A), Semaphorin 3 C(SEMA3C); and anti-inflammatory and antioxidant stress-related proteins, such as Apolipoprotein A1(APOA1), IL-13, Ferritin Light Chain(FTL) was observed. In contrast, pro-inflammatory associated proteins, such as Chitinase 3-like protein 1(CHI3L1), Bromodomain Containing 3(BRD3), and TNF Receptor Associated Factor 3(TRAF3) were downregulated. These changes were identified in the volcano diagram of 237 DEPs(Fig. [81]4A). We also found that the expression of many cytokines, such as connective tissue growth factor(CCN2), insulin like growth factor(IGF), CD55, was significantly elevated in the MSC-MNPs group(Fig. [82]4B). To confirm DIA results, we performed western blotting with antibodies against SLIT2, HGFAC, MDK, APOA1 and CD55. These proteins were confirmed to be upregulated in the MSC-MNPs group (Fig. [83]4C, D, original blots are presented in Supplementary materials Figure S2). Additionally, Western blot results confirmed the downregulation of CHI3L1 and TRAF3 in the MSC-MNPs group (Fig. [84]4C, D). We performed KEGG pathway enrichment analysis of the 237 DEPs and found that these DEPs were clustered in signaling pathways related to complement and coagulation cascades and axon guidance pathway (Fig. [85]4E). Specifically, ten DEPs were identified in the complement and coagulation cascades pathway (F2, F3, F5, F10, uPAR, TPA, C3, Clusterin, DAF, fibrinogen), and seven DEPs were related to the axon guidance pathway (SLIT2, SLIT3, SEMA3A, WNT5A, EPHA, RGS3, PAR3). To further explore the relevant mechanisms, we used Western blot to detect the expressions of PD-L1 and FasL and found that MNPs did not affect the expressions of PD-L1 and FasL in MSCs(Figure S3 A-C). Fig. 4. [86]Fig. 4 [87]Open in a new tab MNPs alters proteomic profiles of MSC. (A) Volcano plot showing the proteins that were upregulated and downregulated in the MSC-MNPs group compared to the MSC group. DIA proteomics revealed 93 upregulated DEPs and 144 downregulated DEPs in the MSC-MNPs group compared with the MSC group (FDR < 0.05, |FoldChange|>1.5), including polarization and axon guidance related proteins, pro-inflammatory related proteins and anti-inflammatory related proteins. (B) Heatmap showing that MNPs uptake by MSC promoted the expression of cytokine-related proteins. (C, D) Western blot analysis verified the upregulation of SLIT2, HGFAC, CD55, APOA1 and MDK and the downregulation of TRAF3 and CHI3L1 in the MSC-MNPs group compared with those in the MSC group. The first three lanes correspond to three samples from the MSC group, while the last three lanes correspond to three samples from the MSC + MNPs group, as indicated in the figure. (E) KEGG pathway enrichment analysis revealed that these DEPs were clustered in the top 15 signaling pathways related to Complement and coagulation cascades and Axon guidance pathway. *P < 0.05, **P < 0.01, ***P < 0.001, triplicate experiments in each group, Student’s t-test. The inflammatory regulatory function of MSC-MNPs promotes the phenotypic switch of reactive astrocytes to alleviate neuroinflammation The above results revealed that MSC-MNPs possess anti-inflammatory and axonal orienting functions. Typically, MSC-MNPs exert their effects through paracrine mechanisms. To verify the functional impact of MSC-MNPs, we designed an experiment involving the coculture of MSC-MNPs with reactive astrocytes. Reactive astrocytes undergo complex morphological, molecular, and functional changes in response to injury^[88]26. In earlier studies, astrocytes were categorized into A1 and A2 phenotypes based on their gene expression, secreted cytokines and function^[89]27. Using a similar classification, we further investigated the effect of MSC-MNPs on the astrocyte response in vitro, focusing on phenotype, synapse length, and angle. Cultured reactive astrocytes were subjected to scratch injury and coculture with MSC for 24 h (Fig. [90]5A). The scratch induced the formation of astrocyte protrusions (Fig. [91]5B), but there was no significant difference in protrusion length between the MSC-MNPs group and the MSC group (Fig. [92]5C). Additionally, there was no significant change in the percentage of astrocytes with protrusions oriented perpendicularly to the lesion (45°−90°) in the MSC-MNPs group compared with the MSC group (Fig. [93]5D). Reactive astrocytes were cocultured with MSC for 24 h, after which their mRNA was collected for RT-qPCR analysis. In the MSC-MNPs group, the levels of A1-phenotype genes C3 (MSC: 1.00 ± 0.10, MSC-MNPs: 0.35 ± 0.14, P < 0.01) and Serping1 (MSC: 1.00 ± 0.13, MSC-MNPs: 0.34 ± 0.08, P < 0.01) were significantly lower, while the levels of A2-phenotype genes Tm4sf1 (MSC: 1.00 ± 0.15, MSC-MNPs: 1.85 ± 0.23, P < 0.05) and Ptx3 (MSC: 1.00 ± 0.73, MSC-MNPs: 4.27 ± 0.65, P < 0.05) were significantly higher compared to the MSC group (Fig. [94]5E). Meanwhile, we measured the levels of neurotrophic factors—GDNF, NGF, CTNF, and BDNF—in the supernatant of astrocytes using ELISA experiments (Fig. [95]5F-I). We found that, compared with the MSC group, the levels of GDNF in MNP-MSC were significantly increased (MSC and MSC-MNPs in pg/ml: 85.46 ± 5.72 and 115.1 ± 6.836, P < 0.05). These results suggest that MSC-MNPs promote the phenotypic switch of reactive astrocytes, thereby alleviating neuroinflammation. Fig. 5. [96]Fig. 5 [97]Open in a new tab Effects of MSC-MNPs on the polarization and phenotype of reactive astrocytes. (A) Schematic diagram of the coculture of MSC-MNPs and astrocytes. (B, C) Astrocyte reactivity was studied in cultured cells via a wound scratch assay. At 24 h after scratching, GFAP immunostaining revealed protrusions of polarized astrocytes, and there was no significant difference in protrusion length between the MSC-MNPs group and the MSC group. (D) Protrusion orientation was evaluated by the angles between the protrusion axes and the scratch line (indicated in B), which revealed that the number of astrocytes with protrusions preferentially oriented perpendicular to the lesion (45°−90°) was comparable between the MSC-MNPs group and MSC group. (E) RT-qPCR quantification of C3, Serping1, Clcf1, B3gnt5, Tm4sf1 and Ptx3 transcripts in cultured spinal cord astrocytes. The mRNA levels are normalized to that of GAPDH. (F-I) The levels of neurotrophic factors, including GDNF, NGF, CNTF, and BDNF, in the supernatant of astrocytes were detected using an ELISA experiment. *P < 0.05, **P < 0.01; n = 100 in C and D, n = 4 in E, n = 3 in F-I; Student’s t-test in C and E; Chi-square test in D. Discussion In this study, we selected umbilical cord-derived mesenchymal stem cells (UCMSC) because of their robust immune regulatory and neuroprotective properties^[98]21. Additionally, UCMSC help to alleviate the limitations associated with the difficulty of generating sufficient cell numbers, making them an important source for transplantation^[99]28. Animal model studies have demonstrated that UCMSC transplantation holds great potential for improving the local microenvironment following central nervous system injury, as well as for promoting neural remodeling and functional recovery^[100]21. At present, a few types of MSC transplants have advanced to Phase I/II clinical trials^[101]29,[102]30. However, monitoring the spatiotemporal migration and homing of MSC, as well as assessing their implantation efficiency and functional capacity, remain challenging problems that need to be addressed. Currently, the use of the magnetic resonance imaging (MRI) capabilities of MNPs to track the fate and distribution of transplanted cells has been applied in animal models and some clinical settings^[103]7,[104]31,[105]32. From the perspective of future clinical applications, MNPs offer several significant advantages. However, detailed information regarding their impact on the biological function of MSC at the cellular level remains limited. Unmodified MNPs are difficult for MSC to take up and may adversely affect the proliferation and migration of MSC. To address this issue, various strategies have been developed to enhance the uptake of MNPs by MSC. These strategies include coating MNPs with a variety of substances, such as surfactants, precious metals, silica, and chitosan. Currently, the three main types of SPION coatings—polyethylene glycol (PEG), starch, and dextran—appear to be the most beneficial and have been approved for human use^[106]33. However, different MNPs or MNPs with different coatings exert varying effects on MSC. For example, Ferucarbotran—a SPION with a crystalline nonstoichiometric Fe^2+ and Fe^3+ iron oxide core—stimulates in vitro MSC proliferation^[107]33. Meanwhile, MNPs coated with polyglucose-sorbitol-carboxymethylether (PSC) have been shown to enhance osteogenic differentiation in human bone marrow-derived mesenchymal stem cells (hBMSC) in vitro^[108]34. In contrast, no significant effects were observed when MSC were treated with silica-coated MNPs^[109]20. Clearly, more research is needed in this area to establish the effects of different MNPs at various concentrations on specific MSC functions and differentiation pathways. Studies have shown that the concentration of MNPs can influence cellular biological functions, which may be related to the oxidative stress induced by high concentrations of MNPs^[110]15. For instance, the optimal concentration for MNPs@SiO₂-RITC in vitro is 0.1 µg/µL, while 1.0 µg/µL represents the plateau concentration for cellular uptake. When MSC were treated with 1.0 µg/µL MNPs@SiO₂-RITC their migration was significantly inhibited, ROS production was promoted, and cell viability was affected. In contrast, no significant cytotoxic effects were observed when MSC were treated with 0.1 µg/µL MNPs@SiO₂-RITC^[111]20. Ideally, the use of MNPs as tracers should have no significant effect on these functions of MSC, indicating that they have no cytotoxic effect. In our study, we developed zinc-doped magnetic nanoparticles (Zn[0.4]Fe[2.6]O[4]), which were confirmed to possess excellent capabilities for MRI^[112]35. We utilized MNPs at a concentration of 0.1 µg/µL, which is suitable for normal MRI^[113]32to evaluate their effects on MSC morphology, phenotype, targeted differentiation ability, proliferation, apoptosis, the cell cycle, and migration. Additionally, the MNPs used in our study were only modified with PLL, without any other coatings, to avoid the possible effects of surface coating on cellular biological functions^[114]36. Interestingly, we found that MNPs can enhance the migratory function of MSC, which is beneficial for their homing. Given that transplanted MSC primarily exert their effects through exocrine mechanisms and can improve the inflammatory microenvironment by regulating cytokine secretion^[115]21we investigated the impact of MNPs on MSC cytokine secretion. Specifically, through multi-cytokine analysis, we discovered that the expression of pro-inflammatory cytokines was reduced in the supernatant of MSC treated with MNPs, and the expression of anti-inflammatory factors was significantly increased. These finding suggest that MNPs uptake by MSC may alter their exocrine function, thereby enhancing their anti-inflammatory capabilities. Single-cell RNA sequencing (scRNA-seq) datasets have revealed the presence of distinct subpopulations of MSC^[116]24,[117]25. These MSC subpopulations exhibit specific surface proteins, different biological activities, and corresponding therapeutic effects. In our study, flow cytometry revealed that after MSC internalized MNPs, the number of CD184^+ MSC subpopulations associated with migration increased significantly. This finding is consistent with those reported in previous studies^[118]35. Similarly, the numbers of CD106^+ and CD55^+ MSC subpopulations, which are related to immunosuppression, also increased. These results further support the potential role of MNPs in promoting MSC migration and improving the inflammatory microenvironment. In our DIA proteomics analysis, we discovered that MNPs not only enhanced the anti-inflammatory function of MSC but also affected proteins associated with cell polarization and axon guidance. To further validate these findings, we conducted coculture experiments with MSC-MNPs and activated astrocytes. These results indicate that MSC-MNPs can indeed promote the conversion of astrocytes from the A1 phenotype to the A2 phenotype and enhance the secretion of the neurotrophic factor GDNF, thereby mitigating neuroinflammation^[119]37. However, MSC-MNPs did not influence the synaptic orientation of astrocytes, which may be attributed to the fact that the axon-guiding functional proteins of MSC do not directly act on astrocytes. However, further experimental verification is still needed to elucidate the axon-guiding function of MSC. In summary, this study demonstrated that the MNPs(Zn[0.4]Fe[2.6]O[4]) we developed at a concentration of 0.1 µg/µL had no cytotoxic effect on MSC and could serve as effective tracers for these cells. Furthermore, the MNPs were found to enhance the migration function of MSC and increase their anti-inflammatory capabilities. However, the comprehensive impact of MNPs on the biological functions of mesenchymal stem cells still requires further verification through animal model experiments. Therefore, in the future, we plan to investigate the effects of MNPs on the biological functions of MSC from different sources and to evaluate the in vivo MRI imaging efficacy of MSC-MNPs, thereby ensuring their safety and effectiveness. Materials and methods Synthesis of PLL-Modified magnetic nanoparticles Zinc acetylacetonate hydrate (97%) and oleic acid were purchased from Aladdin. Iron (III) acetylacetonate (97%) and dibenzyl ether (> 98%) were obtained from Sigma. ε-Poly-L-lysine (PLL, Mw = 4000) was sourced from MACKLIN (Shanghai). The zinc-doped magnetic nanoparticles (Zn[0.4]Fe[2.6]O[4], MNPs) were synthesized via high-temperature pyrolysis, based on a previously published method^[120]38. To impart a positive surface charge to the MNPs, they were modified with PLL through a surface double-exchange process, which enables the MNPs to interact with the MSC. Briefly, 5 mg of MNPs were dispersed in 10 ml of ethanol. Then, 50 mg of PLL was dissolved in 5 ml of deionized water. The ethanol solution of MNPs was exposed to an ultrasonic probe at an amplitude of 30%. The PLL aqueous solution was added dropwise to the MNPs solution and sonicated for 90 min in an ice bath. The resulting product (PLL-MNPs) was washed three times with deionized water and collected by magnetic separation. Identification of MNP characterization The morphology of the MNPs was examined via transmission electron microscopy (TEM, JEM-1230, JEOL Ltd.). The particle size was statistically analyzed with Nano Measurer 1.2 software ([121]https://nano-measurer.software.informer.com/1.2/). The zeta potential and hydrodynamic size of both the MNPs and PLL-MNPs were assessed via dynamic light scattering (DLS, Mastersizer 2000, Malvern, Worcestershire, UK). The static magnetic properties of the dry MNPs were measured via a vibrating sample magnetometer (VSM, Lakeshore 7407, US). Mesenchymal stem cell culture and identification Human umbilical cord-derived MSC were obtained from Saliai Stem Cell Science and Technology (Guangzhou, China). The MSC were cultured in Dulbecco’s Modified Eagle Medium-F12 (DMEM-F12) supplemented with 10% fetal bovine serum (FBS, Gibco) and 1% penicillin/streptomycin (Gibco). The cells were maintained in standard incubators at 37 °C with 5% CO₂. To assess cell identity, surface antigens were analyzed via flow cytometry using the Human MSC Analysis Kit (BD Biosciences, NJ, USA). To evaluate the differentiation potential of MSC, cells at passages 3–4 were treated with MNPs (0.1 µg/µL) overnight and then induced to differentiate into osteogenic, adipogenic, and chondrogenic lineages using complete induction and differentiation media (OriCell, CA, USA). Differentiation was confirmed by staining with Alizarin Red (for osteogenesis), Oil Red O (for adipogenesis), and Alcian Blue (for chondrogenesis). Flow cytometry analysis Flow cytometry was employed to monitor cell surface antigens, cell apoptosis, cell cycle distribution, and ROS levels. Cell surface antigens were detected via the Human MSC Analysis Kit (BD Biosciences, NJ, USA), mouse anti-human CD184-PE antibodies (eBioscience, California, USA), mouse anti-human CD106-PE antibodies (eBioscience), and mouse anti-human CD55-FITC antibodies (eBioscience). Briefly, MSC treated with MNPs (0.1 µg/µL) overnight were collected (5 × 10⁵ cells) and incubated for 30 min with the added antibodies at room temperature in the dark. After washing three times, the cells were analyzed via a flow cytometer (Beckman Coulter, CA, USA). Cell apoptosis and the cell cycle were assessed via the Apoptosis and Cell Cycle Analysis Kit (Invitrogen). For apoptosis detection, 1 × 10⁶ cells were collected and stained with Annexin V-FITC and propidium iodide (PI). For cell cycle analysis, 1 × 10⁶ cells were fixed with 70% ethanol and stained with PI. The detection of ROS levels was performed using the ROS Assay Kit (Beyotime, S0033S), following the provided instructions. The data were analyzed via FlowJo software v11 ([122]https://www.flowjo.com/flowjo/download). MSC chemotaxis assay Transwell assay Transwell chambers with 8-µm-pore membranes (Beyotime, Shanghai, China) were used in 6-well plates. MSC (5 × 10³ cells/ml), treated with or without MNPs, were seeded in the upper chambers in 10% FBS-DMEM/F12 medium (Gibco). The lower chambers contained the supernatant of the macrophages treated with LPS and maintained for 12 h at 37 °C with 5% CO₂. After incubation, the membranes were fixed with 4% paraformaldehyde for 10 min and then stained with Crystal Violet solution (Beyotime). The cells on the upper side of the membrane were removed via a cotton swab, and the cells on the lower side were imaged using a microscope equipped with a 5x objective. Wound scratch assay MSC were seeded in 6-well plates at a density of 5 × 10⁴ cells/well. After reaching 80% confluence, the cells were treated with MNPs (0.1 µg/µl) overnight. The monolayers were then scratched with a 200 µL pipette tip, and the debris was removed by washing. To assess cell migration, images were taken with a microscope equipped with a 5x objective lens in the bright field every 4 h after scratching. The images were analyzed via ImageJ V1.8.0 software ([123]https://imagej.net/downloads). Cell proliferation assay The Cells (1 × 10³) were seeded in 96-well plates. Cell proliferation activity was assessed at 0, 6, 12, 24, and 48 h via the Cell Counting Kit-8 (CCK-8, Dojindo Molecular Technologies, CK04-11), following the manufacturer’s protocol. RT-qPCR Total RNA was extracted from cells via TriZol Reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s instructions. cDNA was synthesized from 1 µg of total RNA using the Reverse Transcription System (Promega). Subsequently, 1 µL of the cDNA was used for PCR amplification via the Eco™ Real-Time PCR System (Illumina). The sequences of the primers used are listed in Supplementary materials. The expression levels of the target genes were evaluated via the 2⁻ΔΔCt method. Cell staining MSC staining MSC treated with MNPs (0.1 µg/µL) overnight were stained with a Prussian Blue Stain Kit (Abcam, ab150674), Eosin (Solarbio, G1100) or ROS Assay Kit (Beyotime). The staining procedures followed the manufacturers’ instructions. Astrocyte staining Astrocytes were incubated with rat anti-glial fibrillary acidic protein (GFAP; 13–0300, Thermo Fisher Scientific) overnight at 4 °C. Next, the cells were incubated with the appropriate secondary antibody for 1 h at room temperature. Cytokine analysis The supernatant of MSC treated with MNPs for 24 h was collected for cytokine analysis. The levels of cytokines were measured via the Bio-Plex system (Bio-Rad) with a 27-plex cytokine kit (M500KCAF0Y, Bio-Rad, CA, USA). This kit includes antibodies against the following cytokines: FGF basic, Eotaxin, TNF-α, MCP-1, MIP-1α, RANTES, IFN-γ, G-CSF, GM-CSF, IL-1ra, IL-1β, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12 (p70), IL-13, IL-15, IL-17 A, IP-10, PDGF-BB, and VEGF. The detection of neurotrophic factors was performed using ELISA kits (BDNF ELISA Kit: Beyotime, PB070; GDNF ELISA Kit: Thermo Fisher Scientific, EEL117; NGF ELISA Kit: Epizyme, HJ265; CNTF ELISA Kit: Cusabio, CSB-E07312m,) in accordance with the reagent instructions. Three samples were analyzed in each experimental group. Data-Independent acquisition (DIA) proteome analysis Total protein was extracted from MSC treated with or without MNPs, via Radio Immunoprecipitation Assay Lysis buffer (RIPA). Protein sequencing and the construction of protein libraries were carried out by Guangzhou Jidiao. Data analysis was performed via OmicShare and Omicsmart tools([124]https://www.omicshare.com/). The criteria for identifying differentially expressed proteins included coefficient of variation < 0.5, average ratio-fold change ≥ 1.5 or ≤ 0.67, and P value < 0.05, as determined by Student’s t-test. Western blots Proteins extracted from MSC-MNPs or MSC, were separated on 10% sodium dodecyl sulfate polyacrylamide gels and transferred to 0.45 μm polyvinylidene fluoride membranes. The following primary antibodies were used: anti-APOA1 (1:1000, Zenbio, 680033), anti-HGFAC (1:1000, Zenbio, 670841), anti-CHI3L1 (1:1000, Zenbio, 820410), anti-TRAF3 (1:1000, Zenbio, 160776), anti-MIDKINE (1:1000, Zenbio, 381110), anti-SLIT2 (1:1000, Zenbio, 862267), anti-β-tubulin rabbit polyclonal antibody (1:1000, Abcam, ab18207), and anti-GAPDH mouse polyclonal antibody (1:1000, Abcam, ab8245), anti-PD-L1 rabbit monoclonal antibody (1:1000, CST, #13684), and anti-FasL rabbit monoclonal antibody (1:1000, Abcam, ab302905). The secondary antibodies used were peroxidase anti-rabbit IgG (1:10000, Abcam, ab6721) and peroxidase anti-mouse IgG (1:10000, Vector Laboratories). Immunoreactivity was detected by a chemiluminescence detection kit (Epizyme, SQ202). Images were captured via the ChemiDoc™ Touch Imaging System (Vilber, France), and the signals were quantified using ImageJ V1.8.0 software ([125]https://imagej.net/downloads). All the data were normalized to the corresponding control average ratios. Animals Animal experiments were approved by the Laboratory Animal Ethics Committee at the University of Health and Rehabilitation Sciences, China (Approval No. 2023 − 2012), and all methods are reported in accordance with ARRIVE guidelines. C57BL/6 mice obtained from Beijing Huafukang Biotechnology Co., LTD. During the experiment, animals were housed at a temperature of 23 ± 1 °C and maintained on a 12-h dark/light cycle. Isoflurane was used to anesthetize the animals when required. The method of animal euthanasia was cervical dislocation after isoflurane anesthesia. Primary spinal astrocyte culture and activation Astrocyte cultures were prepared from neonatal C57BL/6 mice at postnatal days 1–3, based on the previously published method^[126]37. Briefly, spinal cord tissues were dissected from neonatal mice, and then subjected to trypsinization (0.15% trypsin, 7 min, 37 °C). The dissociated spinal cord cells were suspended in astrocyte culture medium, which consisted of DMEM-F12(Gibco) supplemented with 10% FBS(Gibco). When the cells reached 90% confluence, the flasks were shaken at 200 rpm for 20 h to detach the neurons, microglia, and oligodendrocytes. The purity of the astrocytes was assessed by GFAP immunostaining and DAPI nuclear staining, and only astrocytes with a purity greater than 95% were used in subsequent experiments. Astrocytes were activated via the supernatant from macrophages treated with LPS, as previously described^[127]37. Briefly, macrophages (RAW 264.7) were treated with lipopolysaccharide (LPS, 200 ng/ml, Thermo Fisher Scientific) for 24 h. The supernatant was then collected and added to the astrocyte cultures to induce activation. Astrocyte phenotype and polarization assay Transwell chambers with 0.4-µm-pore membranes (Beyotime, Shanghai, China) were used in 6-well plates. MSC (5 × 10^3 cells/ml) treated with MNPs or not, were seeded into the upper chambers in 10% FBS-DMEM/F12 medium (Gibco). Purified astrocytes were plated in poly-D-lysine-coated 6-well plates at a density of 5 × 10⁴ cells/well and served as the lower chambers. Prior to adding the upper chamber, the astrocytes were pre-cultured for 12 h with the supernatant from the macrophages treated with LPS. After coculture for 24 h, some astrocytes were collected for RNA extraction. The remaining astrocytes were used for further analysis of the protrusion length and angle via a wound scratch assay. To study the protrusion length and angle of extension relative to the scratch line, astrocytes were fixed in 4% paraformaldehyde and subjected to anti-GFAP immunostaining. Images were captured with microscope (Zeiss, Germany) and analyzed by ImageJ V1.8.0 software ([128]https://imagej.net/downloads). Statistical analysis The data are presented as mean ± SEM. For comparisons of cell proliferation at different time points, two-way repeated measures ANOVA with Bonferroni’s post-hoc correction was employed. The chi-square test was used for the statistical analysis of protrusion orientation. Other data were analyzed via Student’s t-test. All the statistical analyses were performed using GraphPad Prism 7.04 ([129]https://www.graphpad.com/features). The significance level was set at P < 0.05. *P < 0.05, **P < 0.01, and ***P < 0.001. Electronic supplementary material Below is the link to the electronic supplementary material. [130]Supplementary Material 1^ (2.5MB, docx) Author contributions A. L. designed the research project. X. L. performed studies and data analysis. S. Q., X. L. and Y. L. performed data analysis. A. L. and X. L. drafted the first version of the manuscript with input from S. Q. and X.L. A.L. reviewed and edited the manuscript. All authors read and approved the final manuscript. Funding The work is supported by: National Natural Science Foundation of China (82401622, A. Liu), Natural Science Foundation of Shandong Province (ZR2023QH131, A. Liu), Natural Science Foundation of Qingdao Municipality (23-2-1-148-zyyd-jch, A. Liu), and Guangdong Provincial Key Laboratory of Laboratory of Spine and Spinal Cord Reconstruction(NO. 2023B121203001, A. Liu). The funding sources had no role in study conception and design, data analysis or interpretation, paper writing or the decision to submit this paper for publication. Data availability The datasets generated and/or analysed during the current study are available in the PRIDE repository and project accession: PXD062622. Declarations Competing interests The authors declare no competing interests. Ethical approval Animal experiments were approved by the Laboratory Animal Ethics Committee at the University of Health and Rehabilitation Sciences, China (approval No. 20232012), and all methods are reported in accordance with ARRIVE guidelines. Footnotes Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Xinyue Li and Sheng Qin contributed equally to this work. References