Abstract Spinal cord injury (SCI) is a critical condition affecting the central nervous system that often has permanent and debilitating consequences, including secondary injuries. Oxidative damage and inflammation are critical factors in secondary pathological processes. Selenium nanoparticles have demonstrated significant antioxidative and anti-inflammatory properties via a non-immunosuppressive pathway; however, their clinical application has been limited by their inadequate stability and functionality to cross the blood-spinal cord barrier (BSCB). This study proposed a synthesis method for ultra-small-diameter lentinan Se nanoparticles (LNT-UsSeNPs) with significantly superior reactive oxygen species (ROS) scavenging capabilities compared to conventional lentinan Se nanoparticles (LNT-SeNPs). These compounds effectively protected PC-12 cells from oxidative stress-induced cytotoxicity, alleviated mitochondrial dysfunction, reduced apoptosis. In vivo studies indicated that LNT-UsSeNPs efficiently penetrated the BSCB and effectively inhibited the apoptosis of spinal neurons. Ultimately, LNT-UsSeNPs directly regulated the PI3K-AKT-mTOR and Ras-Raf-MEK-ERK signaling pathways by regulating selenoproteins to achieve non-immunosuppressive anti-inflammatory therapy. Owing to their ultra-small size, LNT-UsSeNPs exhibited strong spinal barrier penetration and potent antioxidative and anti-inflammatory effects without compromising immune function. These findings suggest that LNT-UsSeNPs are promising candidates for further development in nanomedicine for the effective treatment of SCI. Graphical abstract [48]graphic file with name 12951_2024_3054_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12951-024-03054-7. Keywords: Selenium, Spinal cord injury, Antioxidant, Anti-inflammatory, Immune protection Introduction Spinal cord injury (SCI) with the high rates of disability and tissue damage is a prevalent and serious disorder of the central nervous system, ultimately and is associated with considerable incidence and mortality. Approximately 700,000 new cases of SCI are reported annually worldwide [[49]1]. SCI is typically caused by high-intensity trauma, including vehicular falls, accidents, violence, but can also result from infections, tumors, degenerative spinal diseases, ischemia-reperfusion injury, and vascular abnormalities [[50]2, [51]3]. SCI effect motor, sensory, and autonomic functions with the temporary or permanent impairment, imposing a substantial psychological and economic burden on patients and their families. Pathologically, the primary injury is an initial contusion or transection caused by immediate or sustained mechanical compression or traction on the spinal cord [[52]4–[53]8]. Secondary injury is the response of the body and cells to primary injury and involves complex changes in the immune system, nervous system, and blood circulation. These changes include spinal cord ischemia, neuronal death, oxidative stress, inflammatory responses, neuronal apoptosis, axonal demyelination, and glial scar formation. Secondary injury is a key factor leading to spinal cord dysfunction and impedes recovery, with inflammation and oxidative stress playing critical roles in the process. Many studies have showed that oxidative stress is crucial for the onset and progression of SCI. Oxidative stress at the injury site results from a disparity between the production and neutralization of ROS, which encompasses reactive oxygen molecules like H[2]O[2], O[2]^-, ^1O[2], and O[3] [[54]9, [55]10]. Research has confirmed that oxidative stress and inflammation mutually promote each other in an inflammatory environment, where inflammatory cells release significant amounts of ROS, exacerbating oxidative damage [[56]11]. Following SCI, necrotic neurons and other cells release high levels of ROS. Owing to high levels of polyunsaturated fatty acids, oxidative metabolic activity, and limited antioxidant capacity, neurons and glial cells are prone to oxidative stress, damaging adjacent surviving neurons. Additionally, local ischemia and hypoxia following SCI lead to mitochondrial dysfunction in neurons, further increasing ROS production and increasing local oxidative stress levels. Multiple studies have demonstrated that inhibiting oxidative stress can improve neuronal survival rates, restore neurological function, suppress glial cell activation, and reduce the expression of inflammatory factors [[57]12–[58]16]. Thus, ROS, oxidative stress, and inflammation are closely related to SCI, and the inhibition of secondary oxidative stress post-SCI is crucial for the recovery of neurological function. Currently, clinical approaches primarily use glucocorticoids, such as methylprednisolone (MP), to inhibit the production of inflammatory factors after SCI, prevent secondary neuronal death, and inhibit lipid peroxidation to reduce ROS generation and alleviate edema [[59]17, [60]18]. However, high-dose hormone therapy can cause immune system disorders and increase the risk of infection and other complications, thereby affecting patient recovery. With recent technological advancements, nanomaterials have been extensively studied in disease treatment applications and have consistently demonstrated significant efficacy. Particularly in nanomedicine, nanoparticles often exhibit high catalytic effects owing to their unique particle sizes and rich surface-active sites. Hence, developing nanomedicines with small size and high catalytic activity has become a goal for scientists. Selenium (Se), an essential non-metallic trace element in humans and animals, plays a vital role in maintaining health. It has high biological safety and compatibility. As a key component of the body’s antioxidant system, Se is involved in the synthesis of Se-containing proteins, among which glutathione peroxidase (GPX) and thioredoxin reductase (TrxR) are the most common; these proteins exhibit excellent antioxidant effects, regulating immune responses to inhibit harmful inflammatory reactions [[61]19–[62]21]. GPX and TrxR family proteins play crucial roles in inhibiting oxidative stress following central nervous system injury, suppressing oxidative reactions caused by free radicals, reducing body damage, and inhibiting inflammation induced by oxidative stress. These proteins are crucial for the proper development and ongoing functionality of neurons. Studies related to SCI have found that Se can inhibit astrocyte activation and inflammatory factor release after SCI, significantly down-regulating the expression levels of variety oxidative stress markers, such as superoxide dismutase (SOD) and malondialdehyde (MDA). Se also reduces neuronal apoptosis through pathways involving Bax, Bcl-2, and caspase-3 [[63]22]. Additionally, Se supplementation can cause macrophage polarizing to M2 type results in the attenuated expression of interleukin 6 (IL-6), inducible nitric oxide synthase (iNOS) and tumor necrosis factor α (TNF-α) to suppress inflammation, and inhibit local oxidative stress and inflammation levels following SCI [[64]23]. Clinical studies have showed that analyzing blood from SCI patients who experienced trauma with or without neurological recovery revealed significantly higher selenoprotein concentrations in the recovery group than in the non-recovery group [[65]24]. The result indicates that blood Se levels and selenoprotein status reflect the recovery of patients with SCI and that Se aids in their neurological recovery. Selenium nanoparticles (SeNPs), a novel Se species, possess excellent antioxidant, anti-aging, and antitumor activities with low toxicity, high drug-loading capacity, superior biocompatibility, and degradability. Previous studies have demonstrated their therapeutic effects on SCI [[66]25–[67]30]. However, the blood-spinal cord barrier (BSCB) is the main factor influencing nanodrug distribution and efficacy in the clinical treatment of SCI. Owing to the continuous tight junctions between capillary endothelial cells, an intact basement membrane, and the formation of a neuroglial membrane by astrocyte foot processes, the BSCB hinders the passage of SeNPs approximately 100 nm in size from the bloodstream to the injury site. Therefore, there is an urgent clinical need for improving the BSCB penetration of SeNPs that do not affect its effect and safety. Some literatures reported the modification of transmembrane peptides, the improvement of fat solubility and the controlled particles size both advance the barrier penetration of nanodrugs. Thus, ultra-small Se particles have been prepared to better penetrate the BSCB, accumulate effectively at the injury site, and significantly improve SCI treatment efficacy. Although the ultra-small size allows Se nanoparticles to have better BSCB penetration, it introduces new problems, such as a short half-life and poor stability. Lentinan polysaccharides (LNT) exhibit excellent biocompatibility and possess a molecular structure rich in hydroxyl and other functional groups. These groups interact with the surfaces of nanomedicines to form stable chemical bonds or physical adsorption layers. This interaction prevents the aggregation of nanomedicines in aqueous solutions or physiological environments, enhancing their dispersibility and stability. Herein, we prepared an ultra-small Se-based nanosystem with LNT modification (LNT-UsSeNPs) to achieve high BSCB penetration and suitable biostability. Interestingly, its nanoscale size offers advantages when crossing the BSCB, effectively minimizing the impact of the barrier on therapeutic efficacy. Moreover, zero-valence Se-based drugs with strong reducing ability can neutralize ROS and inhibit the inflammatory response triggered by secondary oxidative stress injury, thereby reducing neuronal apoptosis. Finally, they provide protective benefits to an animal’s immune system by regulating selenoproteins, thus avoiding the immunosuppressive effects caused by glucocorticoids. Therefore, the ultra-small Se-based nanosystem, which possesses antioxidant, anti-inflammatory, and immune-protective functions, represents a promising non-immunosuppressive therapeutic strategy for SCI. Results and discussion Design and synthesis of LNT-UsSeNPs Firstly, we successfully synthesized two types of Se nanoparticles: LNT-SeNPs and LNT-UsSeNPs. Uniformly dispersed LNT-SeNPs with an average size of ~ 100 nm were revealed by transmission electron microscopy (TEM) images. Conversely, good monodispersity and a spherical shape were showed by LNT-UsSeNPs, which were notably smaller, averaging 10 nm in diameter (Fig. [68]1A and B). Furthermore, the results of hydrated particle size and zeta potential analysis showed that LNT-UsSeNPs have an average particle diameter of 13.00 ± 1.00 nm, considerably smaller than LNT-SeNPs, which have an average size of 123.25 ± 1.06 nm. The zeta potential of LNT-SeNPs was − 10.21 ± 0.35 mV, whereas LNT-UsSeNPs exhibited a zeta potential of -4.26 ± 0.39 mV (Fig. [69]1C and D). Next, we assessed the stability of the nanoparticles by monitoring changes in their zeta potential in various solutions. The zeta potential of LNT-SeNPs remained stable between − 3 and − 5 mV for up to 96 h, after which an increase in negative potential indicated increase in stability. This pattern was particularly evident in a saline solution (pH 5.0). Meanwhile, the zeta potential of LNT-UsSeNPs remained stable between − 5 and − 10 mV for up to 96 h, although an increase in negative values was observed at 96 h in pH 5.0, 6.8, and Dulbecco’s modified Eagle medium (DMEM). However, in a pH 7.4 buffer saline solution, the zeta potential of LNT-UsSeNPs remained stable between − 6 and − 8 mV, indicating enhanced stability under physiological conditions and highlighting potential biomedical applications (Fig. [70]1H, S1, Supporting Information). Moreover, EDS analysis indicated that the main structural component of both LNT-SeNPs and LNT-UsSeNPs was Se (Fig. [71]1E). The chemical structures of LNT-SeNPs and LNT-UsSeNPs were characterized using Fourier-transform infrared spectroscopy (FT-IR) and ultraviolet-visible (UV-Vis) absorption spectroscopy. UV-Vis absorption spectroscopy showed that both LNT-SeNPs and LNT-UsSeNPs had high absorption peaks at 250 nm and 350 nm, respectively. Additionally, the stretching vibrations were observed at 3420 cm⁻¹ for O-H, 2920 cm⁻¹ for C-H, and 1380 cm⁻¹ for carboxyl groups, which appeared in the spectra of both LNT-SeNPs and LNT-UsSeNPs, confirmed that LNT was successfully modified on the surface of UsSeNPs and SeNPs (Fig. [72]1F and G). Moreover, LNT-UsSeNPs were degraded after a 3 h incubation in a 1% tert-butyl hydroperoxide (TBHP) environment, suggesting that LNT-UsSeNPs can react with TBHP. These results indicated that LNT-UsSeNPs, owing to their smaller particle size and nanoscale dimensions, exhibited superior antioxidant properties and could quickly remove TBHP (Fig. [73]1I). Fig. 1. [74]Fig. 1 [75]Open in a new tab Characterization of LNT-SeNPs and LNT-UsSeNPs. (A, B) TEM images of LNT-SeNPs and LNT-UsSeNPs. (C, D) Size distribution and zeta potential of LNT-SeNPs and LNT-UsSeNPs in aqueous solution. (E) EDS mapping of LNT-UsSeNPs. (F) FT-IR spectra and UV-Vis spectra (G) of LNT-SeNPs and LNT-UsSeNPs. (H) Surface zeta potential of LNT-UsSe NPs over time in different solutions. (I) TEM images of LNT-SeNPs and LNT-UsSeNPs after incubating with 1% TBHP solution for 3 h. (J): The mechanism and process by which LNT-UsSeNPs scavenge free radicals in vitro Assessment of the ROS scavenging capacity of LNT-UsSeNPs Selenium-based drugs and materials are commonly recognized as effective antioxidants that aid in reversing SCI caused by harmful ROS. Thus, an ABTS scavenging assay was used to assess the extracellular ROS scavenging capacity of LNT-SeNPs and LNT-UsSeNPs. The results indicated that LNT-UsSeNPs exhibited a higher ABTS scavenging rate than LNT-SeNPs, with a clear concentration-dependent effect (Fig. [76]2A). With a concentration of 80 µg/mL and an incubation period of 5 min, LNT-UsSeNPs achieved an ABTS scavenging rate exceeding 80%, significantly higher than the 14% observed with LNT-SeNPs. Fig. 2. [77]Fig. 2 [78]Open in a new tab LNT-UsSeNPs reversed the damage of TBHP on PC-12 cell. (A) The in vitro antioxidant activity of LNT-SeNPs and LNT-UsSeNPs was determined using the ABTS radical scavenging assay. (B) EPR spectral analysis of LNT-SeNPs and LNT-UsSeNPs for scavenging •O[2]^−. •O[2]^− was produced via the Fenton reaction using the Fe^2+/TBHP system and was detected using DMPO. (C) UV-Vis spectra of salicylic acid after reacting with •OH radicals generated by the Fenton reaction using the Fe²⁺/TBHP system for 10 min. (D) The changes in total ROS levels in PC-12 cells were detected using DCFH-DA staining. (E) Uptake of LNT-SeNPs and LNT-UsSeNPs (30 µM) by PC-12 cells at different times as detected via ICP-MS. (F) PC-12 cells were incubated with different concentrations of LNT-SeNPs and LNT-UsSeNPs for 24 h, and then incubated with TBHP (10 µM) for another 24 h. (G) The ΔΨm of PC-12 cells was detected via JC-1 staining. (H) PC-12 cells in JC-1 fluorescence image. (I, J) Flow cytometry was performed to analyze the effect of LNT-SeNPs and LNT-UsSeNPs on the proportion distribution of the cell cycle in PC-12 cells treated with TBHP. (K, L) Annexin V-FITC/PI double staining was employed to assess the impact of LNT-SeNPs and LNT-UsSeNPs on the apoptosis of PC-12 cells following TBHP treatment. (M) Mechanism underlying the inhibitory activity of LNT-UsSeNPs against oxidative damage in PC-12 cells. Dates were expressed as the mean ± SD from three independent experiments. Bars with distinct features represent statistically significant differences at the P < 0.05 level The hydroxyl radical (•OH)-scavenging effects of LNT-SeNPs and LNT-UsSeNPs were further investigated using electron paramagnetic resonance (EPR) spectroscopy (Fig. [79]2B). The Fe^2+/TBHP system generated •OH via the Fenton reaction, which was detected using 5,5′ -dimethyl-1-pyrroline N-oxide (DMPO). The EPR spectrum revealed the characteristic signal of the DMPO-OH adduct, indicating the successful generation of •OH. Upon the addition of LNT-SeNPs and LNT-UsSeNPs to the Fe^2+/TBHP system, the signal intensity decreased sharply, particularly in the system with added LNT-UsSeNPs. The •OH scavenging activity of the nanoparticles was also measured using UV-Vis spectrophotometry. As showed in Fig. [80]2C, A characteristic peak at 520 nm in the UV-Vis spectrum was observed, attributed to the reaction of salicylic acid with •OH radicals produced through the Fenton reaction. As expected, LNT-UsSeNPs effectively scavenged •OH radicals, as indicated by their reduced absorbance at 520 nm. These findings highlight the strong extracellular free radical scavenging ability of LNT-UsSeNPs, suggesting that the nanosystem may effectively reduce oxidative stress and minimize oxidative damage in vivo by efficiently eliminating free radicals. ROS scavenging effects and protective effects of LNT-UsSeNPs on oxidative stress injury in PC-12 cells PC-12 cells, a rat-derived pheochromocytoma cell line, are widely used as models in studies on neurotoxicity and neurodegenerative diseases. To investigate the protective influences of LNT-SeNPs and LNT-UsSeNPs on PC-12 cells, we established an in vitro SCI model by using TBHP, a well-known inducer of oxidative stress. SeNPs have garnered significant attention because of their low cytotoxicity [[81]31]. First, the ROS-scavenging effects of LNT-SeNP and LNT-UsSeNPs on PC-12 cells were examined. Intracellular ROS levels were evaluated using the ROS-sensitive fluorescent probe 2′, 7′ -dichlorodihydrofluorescein diacetate (DCFH-DA) [[82]32]. As illustrated in Fig. [83]2D, incubation with TBHP for 10 min increased ROS levels in PC-12 cells by 347.18% and remained elevated at 236.00% after 120 min compared with the control group, which was set at 100%. Notably, LNT-UsSeNPs significantly inhibited the increase in intracellular ROS, reducing it to 106.48% at 120 min, which was lower than the 142.71% observed with LNT-SeNPs. This demonstrated that LNT-UsSeNPs have a superior ability to scavenge intracellular ROS. Next, we investigated the cell-protective effects of LNT-UsSeNPs after ROS scavenging. Initially, the safe dosage ranges of LNT-SeNPs and LNT-UsSeNPs in PC-12 cells were determined. The results indicated a lack of significant cytotoxicity within the concentration range of 0 to 5 µM (Figure S2, Supporting Information). We also analyzed the Se content in PC-12 cells at different time points post-administration using inductively coupled plasma mass spectrometry (ICP-MS). As illustrated in Fig. [84]2E, the intracellular Se concentration of LNT-SeNPs and LNT-UsSeNPs groups reached a higher level after incubation for 2 h, indicating that LNT-SeNPs and LNT-UsSeNPs had better potential to protect PC-12 cells after pre-administration for 2 h. Thus, the CCK-8 assay demonstrated that exposure to 100 µM TBHP reduced PC-12 cell viability to 65–68%. Treatment with both LNT-SeNPs and LNT-UsSeNPs counteracted this effect. Specifically, at a Se concentration of 40 nM, LNT-UsSeNPs increased the PC-12 cell survival rate from 65 to 83%, which was significantly higher than the survival rate of 75% achieved using LNT-SeNPs (Fig. [85]2F). In summary, the size effect gives LNT-UsSeNPs better antioxidant capacity, improving their ability to mitigate oxidative stress injury. Cell cycle arrest and apoptosis were also assessed to explore the possible protective mechanisms of LNT-SeNPs and LNT-UsSeNPs on PC-12 cells, which are the primary mechanisms by which oxidative stress impairs cell growth and induces cell death [[86]33]. Mitochondrial membrane potential (ΔΨm), an early marker in the mitochondrial-mediated apoptotic pathway, was evaluated using JC-1 dye in flow cytometry. As showed in Fig. [87]2G, treatment with 4 µM LNT-UsSeNPs reduced the proportion of cells with mitochondrial depolarization from 34.98 to 10.17%, while LNT-SeNPs reduced it to 6.09%. Thus, LNT-UsSeNPs are more effective at reducing mitochondrial damage. Fluorescence images confirmed these findings, showing that LNT-UsSeNPs reversed the TBHP-induced reduction in the red-to-green fluorescence ratio (Fig. [88]2H). Thus, LNT-UsSeNPs effectively downregulated TBHP-induced cell damage. Additionally, cell cycle analysis (Fig. [89]2I and J) indicated that TBHP treatment significantly increased S-phase cell cycle arrest in PC-12 cells. The Sub-G1 phase of cells was increased from 2.35 to 9.78%, whereas the proportion of cells in the G2/M phase was rose from 21.04 to 33.52%. LNT-UsSeNPs treatment effectively mitigated the TBHP-induced increasing of Sub-G1 phase (from 9.78 to 0.91%) and normalized the G2/M phase cell percentage to approximately 23.03%. Apoptosis was evaluated using Annexin V-FITC and propidium iodide (PI) staining, followed by flow cytometry analysis. As showed in Fig. [90]2K and L, the early apoptotic cell percentage was significantly increased from 0.55 to 24.40% after TBHP incubating. Conversely, the early apoptotic cell count of LNT-UsSeNPs treatment was substantially decreased to 2.95%, that highlighted the strong antiapoptotic properties of LNT-UsSeNPs. These results strongly indicate that LNT-UsSeNPs exert protective effects on mitochondria and normal cell cycle progression by scavenging TBHP-induced ROS in PC-12 cells, thereby inhibiting apoptosis (Fig. [91]2M). Metabolic and biosafety evaluation of LNT-SeNPs and LNT-UsSeNPs To investigate the in vivo metabolism and biosafety of LNT-SeNPs and LNT-UsSeNPs, we used ICP-MS to analyze their pharmacokinetics in rats. The half-life of LNT-UsSeNPs in the bloodstream of rats was determined to be 65.3 h, longer than the 45.6 h observed for LNT-SeNPs (Fig. [92]3A and B). This indicated that LNT-UsSeNPs maintained high plasma drug concentrations for prolonged periods, potentially enhancing drug delivery to SCI sites. Furthermore, ICP-MS analysis revealed that the kidneys showed the highest accumulation of these nanoparticles, suggesting renal-based metabolism for both LNT-SeNPs and LNT-UsSeNPs. Notably, the Se concentration of LNT-UsSeNPs group was up-regulated in the spinal cord, suggesting improved absorption by spinal tissues, potentially beneficial for treating SCI. ICP-MS analysis of various organs reveals that selenium concentrations in the LNT-SeNPs group, with the exception of the spinal cord, are higher compared to those in the LNT-UsSeNPs group. This observation suggests that, when administered at equivalent concentrations, LNT-UsSeNPs exhibit reduced absorption by non-SCI-related organs, thus maintaining elevated levels in systemic circulation over prolonged durations. Such extended circulation increases the likelihood of LNT-UsSeNPs crossing BSCB and reaching the targeted site of SCI. (Figure [93]3C and H). Fig. 3. [94]Fig. 3 [95]Open in a new tab In vivo analysis of biosafety and pharmacokinetics of LNT-SeNPs and LNT-UsSeNPs. (A) Pharmacokinetic analysis of LNT-SeNPs and LNT-UsSeNPs (B) in SD rats. (C–H) Selenium concentrations in various organs of the rats at 24 and 48 h post-injection of LNT-SeNPs and LNT-UsSeNPs. (I) H&E staining analysis of the major organs of rats after treatment with LNT-SeNPs and LNT-UsSeNPs. (J) In vivo fluorescence imaging of LNT-SeNPs and LNT-UsSeNPs at the final time point To assess the in vivo toxicity of the nanoparticles, we conducted histological examinations using hematoxylin and eosin (H&E) staining of sections of major organs post-injection. The results in Fig. [96]3I indicated no significant tissue damage to the main organs, confirming the adequate biosafety of LNT-SeNPs and LNT-UsSeNPs within the administered dose range. Furthermore, we explored the distribution efficiency and processing of LNT-UsSeNPs in vivo. After the injection of fluorescently labeled Se nanoparticles, the fluorescence intensity at the SCI site in the LNT-UsSeNPs group was significantly higher at various time points than in the LNT-SeNPs group, as observed in the fluorescence images of the mouse viscera. Furthermore, the images of LNT-UsSeNP group exhibited significantly higher fluorescence intensity at the SCI site than those of LNT-SeNPs group, with relatively high intensities also observed in the liver, spleen, and kidneys (Fig. [97]3J). These results indicate that LNT-UsSeNPs can more effectively cross the BSCB through blood circulation to reach the SCI site in mice, achieving a therapeutically effective local drug concentration that may facilitate subsequent treatment of SCI. Enhancement of motor function and neuronal survival in mice with SCI upon treatment with LNT-SeNPs and LNT-UsSeNPs To further evaluate the neuroprotective effects of LNT-SeNPs and LNT-UsSeNPs in vivo, we evaluated the recovery of hindlimb motor function in mice following the surgical procedure. Using the Basso Mouse Scale (BMS) and inclined plane tests, we recorded performance on days 1, 3, 5, 7, 10, 14, 21, and 28. The initial results, displayed in Fig. [98]4A, presented hind limb paralysis in all groups except for the sham surgery group, with a BMS score of 0 on day one. Over time, we observed gradual improvements in the BMS scores across all treatment groups, with the LNT-UsSeNPs group demonstrating significant enhancement by day 10. This group also showed improved climbing angles compared to the sham group (Fig. [99]4B). Gait analysis indicated that the SCI group displayed uncoordinated movement and significant resistance to hindlimb motion. Notably, the LNT-UsSeNPs group exhibited a substantial recovery of hind limb gait and motor coordination, as illustrated in Fig. [100]4C. Fig. 4. [101]Fig. 4 [102]Open in a new tab Improvement effect of LNT-UsSeNPs on motor recovery and neuronal survival in SCI mice. (A) BMS scores and slope test (B) results of mice at different times. (C) Footprint analysis was conducted for each group of mice to assess gait and locomotor function. (D) A snapshot from the recorded video shows the sequence of hind limb movements for each group of mice during walking. Points and lines are used to represent the iliac crest, knee joint, and ankle joint. The direction of hind limb movement is indicated by an arrow. (E) H&E staining and (F) Nissl staining was performed on spinal cord tissues from each group, with black arrows indicating the presence of Nissl bodies. (G) Immunofluorescence staining of the spinal cord tissue in each group (Neun, β-tubulin, myelin basic, and GFAP). Dates were expressed as the mean ± SD from three independent experiments. Bars with distinct features represent statistically significant differences at the P < 0.05 level Further investigation of the motion of the mouse hind limbs during walking broke down the movements into a sequence of phases–stance, push-off, lift, swing, and stance–completing one cycle, as depicted in Fig. [103]4D. The SCI group consistently demonstrated hind limb paralysis and dragging. In contrast, mice treated with LNT-UsSeNPs showed notable improvements, such as restored standing posture (stance ability), enhanced weight support, indicated by an increase in iliac crest height, and an expanded range of joint motion, facilitating effective push and swing movements. Effects of LNT-SeNPs and LNT-UsSeNPs on spinal cord tissue pathology and neuronal survival To evaluate the effect of LNT-SeNPs and LNT-UsSeNPs treatments on spinal cord tissue pathology, the experiments of H&E, Nissl, and dual immunofluorescence staining were conducted. H&E staining showed that the spinal cord structures in the sham surgery group remained intact, displaying well-organized neurons with clearly defined nucleoli. In contrast, the SCI group displayed disorganized gray matter, necrotic cavities, irregular neuronal shapes, nuclear pyknosis, and motor neuron loss. Treatment with LNT-UsSeNPs significantly alleviated these pathological changes, indicating a protective effect (Fig. [104]4E). Nissl staining, which is used to assess neuronal integrity [[105]34], revealed a marked decrease in Nissl bodies in the SCI group, whereas LNT-UsSeNPs treatment facilitated the restoration of Nissl bodies (Fig. [106]4F). (Neuron count: Sham: 9; SCI: 4; LNT-SeNPs: 7; LNT-UsSeNPs: 8) Dual immunofluorescence staining assessed the expression of neuronal nuclei protein (Neun), which is important for synaptic generation and neural circuit balance, and β-tubulin [[107]35, [108]36], which is essential for axon guidance and maturation. We observed increased Neun expression and a reduced number of staining cavities in the LNT-UsSeNPs group, suggesting decreased neuronal apoptosis and improved survival (Fig. [109]4G). Myelin integrity in the spinal cord was assessed using myelin basic protein staining [[110]37]. In the SCI group, extensive demyelination and axonal breaks were prominent. However, the LNT-UsSeNPs group exhibited significantly smaller cavities and less axonal damage, indicating preserved axonal integrity and myelination. Additionally, we evaluated glial fibrillary acidic protein (GFAP) expression to analyze astrocytic responses post-injury [[111]38]. SCI typically induces astrocytes to produce GFAP, leading to glial scar formation that impedes neuronal regeneration. GFAP staining revealed that GFAP expression was substantially lower in the LNT-UsSeNPs group, suggesting reduced oxidative stress, inflammation, and less glial scar formation. Collectively, these results confirm that LNT-UsSeNPs provide neuroprotective effects, inhibit axonal demyelination, and reduce glial scar formation in vivo, thereby offering significant therapeutic benefits for SCI treatment. LNT-UsSeNPs regulate selenoenzymes to mitigate oxidative stress and enhance immune protection following SCI MDA, a byproduct of lipid peroxidation, serves as a biomarker for oxidative stress and is prevalent in the lipid-rich spinal cord, where lipid peroxidation is likely to occur post-injury [[112]39]. Thus, we evaluated the MDA content in the spinal cord, finding it notably decreased in the LNT-UsSeNPs group compared to the SCI group (Fig. [113]5A). These results demonstrate that LNT-UsSeNPs exhibit superior antioxidant capacity compared to LNT-SeNPs, consistent with previous in vitro antioxidant studies. Fig. 5. [114]Fig. 5 [115]Open in a new tab Immunoprotective effect of LNT-UsSeNPs by regulating selenoenzyme expression. (A) MDA levels were detected to determine the levels of lipid peroxidation (mM) in the spinal cord tissue after injury in the mice of different groups (A: Sham, B: SCI, C: SCI + LNT-SeNPs, D: SCI + LNT-UsSeNPs). (B) A GSH-Px assay was used to detect the activity of GSH-Px in different groups of spinal cord tissue after injury. (C) qPCR detection results of anti-inflammatory and antioxidant-related Se-containing proteins among the different groups. (A: Sham, B: SCI, C: SCI + LNT-SeNPs, D: SCI + LNT-UsSeNPs) (D) LNT-UsSeNPs upregulate the expression of selenase in neurons. (E–J) Flow cytometry analysis of the proportion of splenic immune cells in the different groups of SCI mice (1: Sham, 2: SCI, 3: SCI + MPSS, 4: SCI + LNT-UsSeNPs). (K–R) Classification and statistics of white blood cells in SCI mice (1: Sham, 2: SCI, 3: SCI + MPSS, 4: SCI + LNT-UsSeNPs). (S) Effect of body immunity on repair of SCI. Dates were expressed as the mean ± SD from three independent experiments. Bars with distinct features represent statistically significant differences at the P < 0.05 level The human body contains at least 25 Se-containing proteins, several of which function as antioxidant enzymes that contribute to mitigating oxidative stress-induced damage [[116]40, [117]41]. Thus, we assessed the mRNA expression of the antioxidative selenoprotein as as glutathione peroxidase (GSH-Px), TrxR, selenoprotein K (SelK), and selenoprotein T (SelT). As illustrated in Fig. [118]5B, treatment with both LNT-SeNPs and LNT-UsSeNPs significantly enhanced GSH-Px activity, with LNT-UsSeNPs showing notably higher activity than LNT-SeNPs. Furthermore, we investigated the effect of LNT-UsSeNPs on the expression of antioxidant selenoproteins using qPCR and western blot analyses. The qPCR results (Fig. [119]5C) showed that treatment with LNT-UsSeNPs significantly increased the mRNA expression of GPX1, GPX2, SelW, and TrxR1 compared to the SCI group. Subsequent western blot analysis validated that LNT-UsSeNPs upregulated the expression of GPX1, GPX2, and TrxR1 in spinal cord tissues. These findings suggest that LNT-UsSeNPs mitigate oxidative stress-induced SCI by regulating antioxidant selenoproteins, thereby exerting neuroprotective effects in mice with SCI. The western blot results further verified the qPCR results (Fig. [120]5D). The common clinical treatment for SCI involves high-dose corticosteroid therapy, specifically with methylprednisolone sodium succinate (MPSS), which suppresses immune function despite its anti-inflammatory and antioxidative benefits. This suppression can lead to complications such as bedsores and urinary and respiratory infections, significantly hindering patient recovery and increasing both treatment duration and cost [[121]18, [122]42]. Therefore, we investigated whether LNT-UsSeNPs, which also exhibit anti-inflammatory and antioxidant properties, affect immune function using MPSS as a clinical comparator. Flow cytometric analysis of splenic cells revealed significant changes in immune cell populations. After MPSS treatment, there was a notable decrease in the proportion of B lymphocytes (CD19+) compared to the sham groups, while the LNT-UsSeNPs group showed an increase in the proportion of B lymphocytes. No significant differences existed among the groups in the NK cell proportion (Fig. [123]5E, S4). T cell profiling showed that the proportion of CD4 + cells decreased following MPSS treatment, whereas that of CD8 + cells increased in the MPSS group. Conversely, the LNT-UsSeNPs group showed an increased proportion of CD4 + cells, with no significant changes in CD8 + cells. The CD4+/CD8 + ratio, a key indicator of immune function, generally decreases when cellular immunity is suppressed and immune function is compromised [[124]43, [125]44]. The ratio was considerably lower for the MPSS group compared to the sham group, suggesting the presence of immunosuppression. However, the CD4+/CD8 + ratio in the LNT-UsSeNPs group increased, suggesting that LNT-UsSeNPs do not suppress immune function but exert antioxidative and anti-inflammatory effects (Fig. [126]5F and J, S5). Complete blood count results further supported these findings (Fig. [127]5K and R, Table [128]S1). The MPSS group showed a decrease in the proportion of lymphocytes among white blood cells compared to the sham and control groups, whereas the LNT-UsSeNPs group showed an increase in the lymphocyte proportion, surpassing that of the sham group. In addition, we found that the number and proportion of neutrophils in the MPSS group were significantly higher than those in the other groups, suggesting that mice injected with a high dose of MPSS experienced a certain degree of bacterial infection (Fig. [129]5K and O). This reflects a disruption in their immune system, leading to the occurrence of bacterial infection. However, no such phenomenon was observed in the LNT-UsSeNPs group. Therefore, safeguarding the immune system and minimizing immune-related complications during SCI treatment are crucial for patient recovery (Fig. [130]5S). LNT-UsSeNPs exert antioxidant and anti-inflammatory effects to inhibit neuronal apoptosis via the PI3K-AKT-mTOR-eIF4EBP1 and Ras-Raf-MEK-WIPI2 pathways To explore whether LNT-UsSeNPs regulate signaling pathways to inhibit neuronal apoptosis, we employed Tandem mass tag (TMT) labeling for quantitative proteomic analysis to identify differences in protein expression among experimental groups. Comprehensive enrichment analysis of the TMT proteomic data identified differentially expressed proteins between the LNT-UsSeNPs and SCI groups that had significant biological relevance (Fig. [131]6A). Subcellular localization analysis indicated that the majority of these differentially expressed proteins were primarily found in the cytoplasm, extracellular matrix and nucleus, suggesting that they play critical roles in different cellular compartments (Fig. [132]6B). Volcano plot analysis identified a number of proteins with significant differential expression (Fig. [133]6C), shedding light on the potential molecular mechanisms underlying the effects of LNT-UsSeNPs. Utilizing the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, we annotated the functions of the proteins that exhibited variable expression and conducted a comparative analysis to distinguish between those that were upregulated and those that were downregulated (Fig. [134]6D and F). The results demonstrated that LNT-UsSeNPs significantly upregulated key proteins in the PI3K-AKT-mTOR and Ras-Raf-MEK-ERK signaling pathways, promoting cell growth, proliferation, and differentiation. After SCI, oxidative stress and inflammation contribute significantly to secondary damage, neuronal injury, and functional loss. The PI3K-AKT-mTOR and Ras-Raf-MEK-ERK pathways are crucial in mitigating oxidative stress and inflammation. The PI3K-AKT-mTOR pathway promotes neural repair by regulating cell survival and inhibiting oxidative stress and inflammation. AKT activation reduces free radicals by upregulating antioxidant enzymes like SOD, while also suppressing the release of inflammatory factors such as TNF-α and IL-6. The mTOR pathway further supports neuroprotection by promoting protein synthesis and cell regeneration, reducing tissue destruction. The Ras-Raf-MEK-ERK pathway, essential for cell proliferation and survival, aids in tissue repair by enhancing neuronal survival and glial proliferation. ERK activation reduces oxidative stress by increasing antioxidant gene expression and limiting free radical accumulation, while also inhibiting pro-inflammatory factor production, thereby preventing additional neuronal damage and promoting neural repair. Therefore, we hypothesized that LNT-UsSeNPs could effectively enhance the survival of neurons following SCI by activating these signaling pathways, thereby protecting them from apoptosis induced by secondary oxidative stress and inflammation. This ultimately leads to an improved neuronal survival rate and partial recovery of motor function. Further analysis of cellular markers indicated that neuron-related proteins dominated the differential proteins (Fig. [135]6G), which was consistent with our initial hypothesis and further validated the neuron-specific effects of LNT-UsSeNPs. Additionally, KEGG pathway analysis suggested that LNT-UsSeNPs may further potentiate their neuroprotective effects through modulation of proteins expression associated with antioxidant, anti-inflammatory, and apoptotic processes linked to the PI3K-AKT-mTOR and Ras-Raf-MEK-ERK signaling pathways (Fig. [136]6I). These findings offer new perspectives on the potential mechanisms of action of LNT-UsSeNPs in the treatment of SCI and lay the foundation for further in-depth research. Fig. 6. [137]Fig. 6 [138]Open in a new tab LNT-UsSeNPs inhibit neuronal apoptosis by exerting antioxidant and anti-inflammatory effects. (A) Principal component analysis score interaction diagram of the SCI and LNT-UsSeNPs groups (3D). (B) Subcellular localization of differentially expressed proteins between the SCI and LNT-UsSeNPs groups. (C–F) Volcano plot illustrating the 27 differentially expressed genes between the SCI and LNT-UsSeNPs groups along with the KEGG pathway enrichment analysis of overlapping genes (C: Volcano map; D: Enrichment analysis of the differential genes; E: enrichment analysis of the upregulated genes; F: Enrichment analysis of downregulated genes). (G) Analysis of cellular markers of the differentially expressed genes. (H) The molecular mechanism LNT-UsSeNPs inhibit neuronal apoptosis. (I) Western blot analysis of the differentially expressed proteins identified using TMT proteomics(1: Sham, 2: SCI, 3: SCI + MPSS, 4: SCI + LNT-UsSeNPs) Western blotting was then performed to validate the proteomic data (Fig. [139]6H). The results revealed that LNT-UsSeNPs increased the expression of proteins within the PI3K-AKT-mTOR signaling pathway and simultaneously suppressed the expression of the translation initiation repressor, eukaryotic translation initiation factor 4E binding protein 1 (eIF4EBP1). This dual action enhanced mRNA translation and protein synthesis in neurons. Our data suggest that the upregulation of eIF4EBP1/2 activates eukaryotic Initiation Factor 4E (eIF4E), initiating cap-dependent mRNA translation. This process is essential for neuronal development and the growth of synaptic connections [[140]45]. Additionally, LNT-UsSeNPs downregulated the expression of the apoptosis-related protein WD repeat domain phosphoinositide-interacting protein 2 (WIPI2), enhancing neuronal cell survival and reducing apoptosis. Simultaneously, the upregulation of the Ras-Raf-MEK-ERK pathway inhibits the expression of Cullin 2, a component of the ubiquitin complex, suppressing neuronal apoptosis. Furthermore, LNT-UsSeNPs have been showed to reduce the expression levels of pro-inflammatory proteins, such as S100A9, iNOS, and IL-6, and concurrently enhance the expression of the anti-inflammatory protein IL-10. Regarding antioxidant properties, LNT-UsSeNPs promoted the expression of apoptosis protease activating factor-1 interacting protein (APIP), mitigating oxidative stress-induced neuronal apoptosis. Furthermore, LNT-UsSeNPs enhanced the levels of NADH dehydrogenase iron-sulfur protein 3 (NDUFS3), maintaining normal aerobic metabolism in neurons and upregulated acyl-CoA oxidase 1 (ACOX1) expression, stabilizing intracellular lipid metabolism. LNT-UsSeNPs treatment also increased the expression of neural cell adhesion molecule 1 (NCAM1), facilitating neural regeneration and transmembrane signal transmission, which are beneficial for neural function repair post-SCI. Collectively, these results robustly demonstrate that LNT-UsSeNPs mitigate neuronal apoptosis by upregulating the PI3K-AKT-mTOR and Ras-Raf-MEK-ERK pathways and modulating the expression of related antioxidant and anti-inflammatory proteins. These effects reduce local oxidative stress and inflammation while enhancing the expression of proteins involved in neural repair and promoting functional recovery. In general, LNT-UsSeNPs exerted anti-inflammatory and antioxidant effects without inhibiting the immune system, as demonstrated by immune-related and tissue protein-related experiments. This has a significant advantage over glucocorticoid therapy, as it avoids related infectious side effects. Conclusions In this study, we synthesized ultra-small LNT-UsSeNPs, which exhibited obvious therapeutic effects against SCI owing to their improved structural design and surface modifications. LNT was used as a modifying agent, producing LNT-UsSeNPs to improve stability and cellular uptake. LNT-UsSeNPs possess a larger specific surface area than LNT-SeNPs, resulting in relatively enhanced antioxidant capacity, which effectively reduces ROS-induced cellular damage. In addition, LNT-UsSeNPs effectively counteracted TBHP-induced cell cycle G2/M arrest, mitochondrial disruption, and apoptosis. In vivo, LNT-UsSeNPs significantly enhanced the hind limb motor function of SCI-affected mice. Histological analyses revealed the neuroprotective effects of LNT-UsSeNPs in mice with SCI. Proteomic and western blot analyses confirmed that these nanoparticles activated the PI3K and MAPK pathways, upregulated selenoprotein expression, mitigated oxidative damage and inflammation, and protected neurons from further injury. Furthermore, they exhibit antioxidant and anti-inflammatory properties without impairing the immune system. Overall, these ultra-small LNT-UsSeNPs offer several advantages. The smaller particles size of LNT-UsSeNPs provides an advantage in crossing the BSCB, effectively reducing the impact of the barrier on drug accumulation and enhancing therapeutic efficacy. Additionally, the strong reductive properties of zero-valent selenium in the nanoparticles can neutralize ROS and regulate selenoprotein levels, thereby inhibiting secondary oxidative stress and associated inflammatory responses, ultimately reducing neuron apoptosis caused by secondary injury. Moreover, LNT-UsSeNPs exhibited protective effects on the immune system, avoiding the immunosuppressive effects commonly induced by anti-inflammatory drugs such as glucocorticoids. In summary, LNT-UsSeNPs have promising potential as non-immunosuppressive nanomedicines for treating SCI and may be a novel therapeutic strategy for managing oxidative stress and inflammation via selenoprotein regulation. Materials and methods Preparation of LNT-UsSeNPs To synthesize ultra-small selenium nanoparticles (UsSeNPs), we began by mixing 20 mg of selenium powder with 10 mL of polyethylene glycol 400 (PEG 400) at ambient room temperature (20–25 °C). The resulting mixture was subjected to heating at 290 °C under continuous stirring at 360 rpm for 2 h to ensure thorough dispersion and nanoparticle formation. Upon completion of the reaction, the resulting UsSeNPs suspension was allowed to cool and subsequently stored at room temperature for later use. To create LNT-UsSeNPs, we first dissolved 20 mg of LNT in 8.4 mL of ultrapure water to produce a homogeneous LNT solution. Following, the UsSeNPs suspension (1.6 mL) was gradually introduced to the LNT solution dropwise, maintaining a controlled rate of 1–2 s per drop. Stirring was sustained at 300–500 rpm at room temperature (20–25 °C) for 12 h to promote stable nanoparticle-ligand interaction and uniform coating formation. To confirm successful nanoparticle formation and quantify the selenium content within the LNT-UsSeNPs, ICP-MS analysis was performed. Preparation of LNT-SeNPs The synthesis of LNT-SeNPs was initiated by dissolving 20 mg of LNT in 9 mL of ultrapure water, ensuring complete solubilization at room temperature (20–25 °C). Following, 0.5 mL of a 100 mM sodium selenite (Na₂SeO₃) solution was gradually added to the LNT solution. Next, 0.5 mL of a 400 mM ascorbic acid (vitamin C) solution was added into the mixture. The addition of ascorbic acid was performed gradually to ensure a controlled reduction process, promoting the uniform formation of selenium nanoparticles within the LNT solution. The mixture was stirred continuously at 300–500 rpm for 12 h, allowing sufficient time for complete reduction and particle stabilization. To remove any unreacted components and by-products, the resulting solution was dialyzed against ultrapure water for 48 h.The Se concentration in the final product, LNT-SeNPs, was determined using ICP-MS. Characterization of LNT-SeNPs and LNT-UsSeNPs The microstructural properties of LNT-SeNPs and LNT-UsSeNPs were examined using TEM, providing a detailed visualization of the nanoparticle morphology. The particle size, zeta potential and stability of LNT-SeNPs and LNT-UsSeNP were analyzed by Malvern Zetasizer Nano ZS. In order to evaluate the stability of LNT-SeNPs and LNT-UsSeNP under different physiological conditions, stability experiment was carried out in different time periods at pH 5.8, 6.1 and 7.4, phosphate buffer (PBS) and DMEM (0–168 h). The chemical structures of the nanoparticles were characterized by UV-Vis and FT-IR spectroscopy. These tests are critical for assessing the potential of these nanoparticles for biomedical applications, particularly in terms of their stability and integrity in diverse biological environments. Evaluation of the ROS scavenging activity The antioxidant capabilities of LNT-SeNPs and LNT-UsSeNPs were assessed on the basis of their ability to scavenge ROS using the ABTS (2,2’-azino-bis[3-ethylbenzothiazoline-6-sulfonic acid]) assay. ABTS solution (5 mM) was then activated by adding 10 mg of manganese dioxide (MnO[2]) and filtering through filter paper and a membrane filter. The activated ABTS solution was stored in the dark at -20 °C for 12 h prior to use. In the experimental setup, various concentrations of the nanoparticles (2.5–80 µg/mL) were prepared in PBS. An equal volume of these nanoparticle solutions (100 µL) was mixed with ABTS (100 µL) solution in a microplate format. The mixtures were subsequently incubated at 20–25 °C under dark conditions for 25–30 min. Absorbance readings at 734 nm were taken at one-minute intervals over a 15-minute period. The ROS scavenging rate was determined utilizing the following formula: graphic file with name M1.gif Evaluation of •OH and •O[2] Firstly, a 1% TBHP was prepared by dissolving 70% TBHP in ultrapure water. The TBHP in the solution undergoes decomposition, leading to the generation of •O[2]^−. In the TBHP reaction system, the oxygen generated from the decomposition of TBHP can combine with electrons to form •O[2]^−. Subsequently, •OH were produced via the Fenton reaction using a Fe²⁺/TBHP system, with 1.6 mM FeSO₄ and 1% TBHP for 15 min. The scavenging capacities of LNT-SeNPs and LNT-UsSeNPs against •OH and •O[2]^− were then analyzed using EPR spectroscopy. Briefly, LNT-SeNPs and LNT-UsSeNPs (15 µg/mL) were added to solutions containing the generated •OH or •O[2]^− radicals and incubated for 30 min. Then, DMPO was added and EPR was used to detect the ability of LNT-SeNPs and LNT-UsSeNPs to scavenge •OH and •O[2]^− radicals. The •OH scavenging capacities of LNT-SeNPs and LNT-UsSeNPs were indirectly measured by detecting the formation of 2,3-dihydroxybenzoic acid from salicylic acid (2 mM) and •OH radicals, presenting a characteristic absorption at 510 nm in UV-Vis spectroscopy. A DHE fluorescent probe was used to detect the oxygen-scavenging capacity of LNT-SeNPs and LNT-UsSeNPs. PC-12 cell culture and assessment of cell viability PC-12 cells were obtained from the ATCC (Manassas, VA, USA) and cultured in DMEM supplemented with fetal bovine serum (10%) and penicillin-streptomycin (1%) at 37 °C in an environment containing 5% CO₂. 4 × 10^3 PC-12 cells were plated in 96-well plates. After drug incubating, 20 µL of the CCK-8 was added to evaluated the cell viability. To investigate the cellular uptake dynamics of LNT-SeNPs and LNT-UsSeNPs, PC-12 cells were incubated with these nanoparticles under controlled conditions. Briefly, PC-12 (1 × 10^5 cells/mL) were seeded in 6-cm culture dishes. Both types of nanoparticles were administered to the cells, and incubation periods were set at 2, 4, 8, and 24 h to capture the time-dependent uptake. After incubation with LNT-SeNPs and LNT-UsSeNPs respectively, the cells were harvested, and the cell suspensions were subjected to nitric acid digestion to prepare them for elemental analysis. The Se concentrations in the cells at each time point were quantitatively measured using ICP-MS. This method allowed the precise determination of the amount of Se taken up by the cells over specified durations, providing insight into the kinetics of nanoparticle absorption by neural-like cells. This approach is crucial to understanding the bioavailability and potential therapeutic efficacy of Se-based nanoparticles in a neurological context. To evaluate the safe dose range of LNT-SeNPs and LNT-UsSeNPs for PC-12 cells, the cells were exposed to different concentrations of LNT-SeNPs and LNT-UsSeNPs for 24 h. Subsequently, we assessed the viability of PC-12 cells after 24 h using the CCK-8 assay. To determine the impact of LNT-SeNPs and LNT-UsSeNPs on reversing TBHP-induced oxidative damage to cell viability, PC-12 cells were treated with LNT-SeNPs and LNT-UsSeNPs for 24 h, followed by drug removal and subsequent exposure to TBHP for another 24 h. Next, the viability of PC-12 cells was evaluated 24 h post-treatment using the CCK-8 assay. Flow cytometry analysis of the cell cycle, apoptosis, and ΔΨm 1.6 × 10^5 PC-12 cells were cultured in 6-well plates. Initially, the cells were treated with LNT-SeNPs and LNT-UsSeNPs for a period of 24 h, after which they were further treated with TBHP for an additional 24 h. Afterward, probe staining and flow cytometry were conducted to analyze the cells. In conducting cell cycle analysis, PC-12 cells were first fixed using 70% ethanol and stored at -20 °C for 48 h. Subsequently, the cells were stained with PI at room temperature, ranging from 20 to 25 °C, for a duration of 30 min. For the assessment of apoptosis, the cells were stained with Annexin V and PI, adhering to the protocol specified in the Annexin V-FITC/PI Apoptosis Detection Kit. Additionally, the JC-1 probe at a concentration of 10 mg/L was used to stain PC-12 cells, which were then incubated at 37 °C for 30 min. Fluorescence microscopy was subsequently utilized to capture JC-1 fluorescence images of the cells, facilitating the analysis of the ΔΨm. Detection of total intracellular ROS levels PC-12 cells were initially seeded into a 96-well plate (10 × 10⁴ cells/mL). Following a 24 h period, the cells underwent pre-treatment with 4 µM concentrations of both LNT-SeNPs and LNT-UsSeNPs for 6 h. Subsequently, the cells were stained by incubation with DCF-DA (37 °C, 30 min), prior to a 5-min exposure to 100 µM TBHP. The resulting fluorescence intensity was measured at the wavelengths of 488/525 nm using the BioTek Cytation5 imaging reader. In vivo metabolism and toxicity studies 6–8 weeks old Sprague–Dawley rats were procured from the Guangdong Provincial Laboratory Animal Center and randomly assigned to two groups. Each group was intravenously injected with 1 mL of either LNT-SeNPs or LNT-UsSeNPs at a concentration of 3 µM. Orbital blood samples were collected before injection and at 0.5, 1, 2, 4, 6, 8, 12, 24, and 48 h post-injection. Blood Se concentrations were measured using ICP-MS. At 24 and 48 h post-injection, two rats from each group were euthanized, and their spleens, kidneys, lungs, livers, and hearts were collected in 4% paraformaldehyde, followed by paraffin embedding and sectioning. The H&E staining of tissue sections for microscopic examination to assess the acute visceral toxicity of the nanoparticles. SCI Animal model establishment Female C57/BL6J mice with 20–22 g weight (6–8 weeks) were procured from the Guangdong Jicui Yaokang Animal Center. The animal experiment protocol has been reviewed and approved by Laboratory Animal Welfare and Ethics Committee of Jinan University (Ethics Approval Number: IACUC-20230919-06). Mice were anesthetized with tribromoethanol and underwent laminectomy at the T10 level under sterile conditions. A Model III-NYU impactor (W.M. Keck/USA) was used to induce SCI with parameters set at a 0.6 mm impact depth, 1.2 m/s impact speed, and 80 ms dwell time. Sham-operated animals underwent laminectomy without inducing spinal cord damage. The wounds were then sutured in layers and were daily injected with ceftriaxone (0.3 g per mouse) for seven consecutive days. Mice were randomly divided into four groups (Sham, SCI, LNT-SeNPs, LNT-UsSeNPs). 2 mg/kg of LNT-SeNPs and LNT-UsSeNPs were administered intravenously. The sham and SCI groups were administered equivalent volumes of saline. All treatments were given once daily for seven consecutive days, starting on the day of SCI modeling. Behavioral assessments Basso Mouse Scale (BMS) and inclined plane tests were conducted regularly. On day 28, gait analysis was performed, followed by euthanasia of the mice and extraction of the spinal cord. Behavioral assessments Behavioral assessments were conducted by trained, blinded observers using the BMS, inclined plane test, footprint analysis, and hind limb locomotor cycle analysis. The BMS is a 9-point scale representing the recovery of hind limb function, ranging from 0 (no observed hind limb movement) to 9 (normal hind limb movement). The inclined plane test measured the maximum angle of the surface at which a mouse could maintain its position for five seconds. For footprint analysis, the mice were coated with the black dye on their paws and were allowed to walk along a 0.5-meter-long narrow corridor lined with white paper to record their footprints. Additionally, video recordings of mice walking along the corridor were used to analyze the locomotor cycle of the hind limbs, which included the phases of contact, propulsion, lift-off, swing, and back-to-contact. Histological staining After the necessary pretreatment, the tissue was were stained with H&E, Nissl, and dual immunofluorescence (Neun/DAPI, β-tubulin/DAPI, Myelin basic/DAPI, or GFAP/DAPI). All antibodies were purchased from Abcam, USA. Analysis of LNT-SeNPs and LNT-UsSeNPs metabolism in mice Briefly, 2.4 mL of a 2.5 mM LNT-SeNPs solution and 2.4 mL of 2.5 mM LNT-UsSeNPs solution were each mixed with 6 mg of Indocyanine Green-Thiol (ICG-SH) and stirred for 24 h to obtain ICG-SH-LNT-SeNPs and ICG-SH-LNT-UsSeNPs solutions, respectively. Twelve 6–8 weeks-old C57 mice were then selected, anesthetized, and subjected to SCI modeling. Six mice were randomly chosen and injected with 300 µL of ICG-SH-LNT-SeNPs and ICG-SH-LNT-UsSeNPs (300 µl) by intravenous injection. Then, the fluorescence imaging was performed on mice before and at 0.1, 0.5, 1, 2, 4, 6, and 8 h after drug injection to analyze and record the fluorescence intensity emitted by the drugs within the mice. After 8 h, the mice injected with ICG-SH-LNT-SeNPs and ICG-SH-LNT-UsSeNPs were euthanized, and the fluorescence imaging of major organ, including liver, heart, lungs, spleen, kidneys, and spine were measured to analyze and record the fluorescence intensity of the drugs. Finally, the liver, heart, lungs, spleen, kidneys, and spine were digested with aqua regia, quantified, and analyzed for Se concentrations using ICP-MS. In vivo antioxidant capacity analysis The antioxidant properties of LNTSeNPs and LNT–UsSeNPs in vivo were assessed through the quantification of GSH-Px activity and MDA levels in spinal cord homogenates. Firstly, samples were mixed with RIPA lysis buffer, which was fortified with protease inhibitors. Then, the BCA Protein Assay Kit was used to measure the protein content within these homogenates. GSH-Px activity and The MDA content were determined using specific assay kits the respective assay kits (Beyotime, China) following standard protocols. qPCR analysis of selenoprotein expression Trizol reagent (Takara Biotechnology, Japan) was utilized to extract total RNA from the spinal cord. Subsequently, the reverse transcription process was carried out using the PrimeScript RT Master Mix (Takara Biotechnology, Japan). The cDNA was then amplified via qPCR with SYBR Premix Ex Taq II, another product of Takara Biotechnology, Japan, on a CFX Connect Real-Time PCR Detection System, which is manufactured by Bio-Rad, USA. After thermal cycling conditions, the relative gene expression analysis used β-actin as internal control. Proteomic analysis Three mice were randomly selected from each group three days after SCI modeling. The animals were euthanized via cervical dislocation, and spinal cord tissues were harvested and stored in cryovials, which were then frozen in liquid nitrogen. TMT proteomic analysis was then performed on the samples. The experimental steps are as follows: TMT labeling Peptides were redissolved in 100 mM TEAB and labeled with TMT reagents (peptide = 5:1). The labeling reaction was performed at 20–25 °C for 1–2 h. Reversed-phase high-pressure liquid chromatography (HPLC) fractionation Peptides from each sample were combined and injected into a C18 column for separation via gradient elution (5–90%) at 0.3 mL/min (elution peaks: 214 nm), with ten fractions collected, merged based on chromatograms, and freeze-dried. Quantitative detection by nano-LC-MS/MS The dried peptides were reconstituted in 0.1% FA, centrifuged at 20,000×g (10 min), and loaded onto a self-packing C18 column. Separation was performed on a Thermo EASY-nLC™ 1200 system with a flow rate of 300 nL/min, using a gradient elution from 8 to 90% solvent B over 62 min. Peptides were ionized via nano-ESI and analyzed on an Orbitrap Exploris™ 480 mass spectrometer in DDA mode (MS range: 350-1,500 m/z, resolution: 120,000). Secondary fragmentation was carried out using HCD at 36% energy with dynamic exclusion (60 s) and a resolution of 45,000. Statistical analysis The differential expression thresholds were set at a 1.2-fold change relative to baseline (P < 0.05). Subcellular localization predictions were conducted using the WoLFPSORT database, while the associated protein functions were identified through UniProt and a review of relevant literature. The KEGG database was used to perform enrichment analysis on the differentially expressed proteins. Western blot analysis BCA assay was conducted to measure the concentration of protein, and total protein (30 µg) was separated on SDS-PAGE gel (10%). The proteins were transferred to the membrane of PVDF (Millipore, USA) using a microtransblot device (Bio-Rad, USA). The membrane after BSA solution (5%) blocking was incubated with the primary antibody overnight at 4 °C. It was then treated with an enzyme-labeled secondary antibody (4 °C). GAPDH and β-actin served as internal controls. Primary reactants: GPX1, GPX2, TrxR1, PI3K, P-AKT, mTOR, eIF4EBP1, Ras, Raf, MEK, ERK1/2, WIPI2, ACOX1, S100A9, iNOS, IL-6, IL-10, NCAM1, APIP, β-actin, and GAPDH (Abcam, USA). Secondary antibody: Anti-mouse IgG (Cell Signaling Technology, USA). In vivo immunological analysis in mice after LNT-UsSeNPs treatment The C57/BL6J mice were used for immunological analysis. LNT-UsSeNPs were administered intravenously at a Se concentration of 2 mg/kg daily for three consecutive days, starting from the day of SCI induction. MPSS was administered as a high-dose pulse therapy: 0.5 h after post-SCI, 30 mg/kg of MPSS was injected intravenously, followed by a second injection after 2 h (15 mg/kg), and a third injection of 15 mg/kg after 4 h. On the following day, 35 mg/kg MPSS was administered every 4 h for a total of three doses. Both sham and SCI groups received saline. All the mice were treated once daily. Day three, blood was collected via orbital puncture for a complete blood count with differential count, and the mice were euthanized via cervical dislocation. The spleens were then harvested, homogenized, and lysed. Splenic immune cells were harvested via centrifugation, labeled with specific antibodies, and assessed using flow cytometry. Electronic supplementary material Below is the link to the electronic supplementary material. [141]Supplementary Material 1^ (247.2KB, docx) Acknowledgements