4.1 Morphology and antigen identification of BMMSCs
Upon examination under an inverted microscope, P3 BMMSCs exhibited robust adhesion to the substrate, displaying a consistent fibroblast-like morphology characterized by uniform distribution and swirling growth patterns (Figure 1C). Subsequently, the flow cytometry analysis revealed that P3 BMMSCs exhibited positive expression of CD44 and CD90, indicating the presence of these markers on the cell surface. Conversely, the cells showed negative expression of CD45 and CD34, suggesting the absence of these markers. This surface antigen profile aligns with the characteristic phenotype of BMMSCs (Figure 1D).
4.2 BMMSCs facilitate the functional motor abilities recovery in rats with SCI.
The evaluation of mobility abilities utilizing the BBB score revealed that on the initial postoperative day, rats with SCI displayed total paralysis with a score of 0, indicating the absence of hindlimb activity (Figure 2A). By the third day following the injury, a gradual restoration of hindlimb functionality became evident, with no notable distinction observed between the group treated with BMMSCs and the SCI group. Nevertheless, by day 14 after the injury, a notable enhancement in hindlimb function became evident in the BMMSCs group compared to the SCI group (p<0.0001; Figure 2B). Remarkably, 28 days following the occurrence of the injury, the BMMSCs group demonstrated a markedly higher mean BBB score of 10, significantly exceeding the SCI group's performance (p<0.0001; Figure 2C). During the footprint analysis, SCI rats exhibited noticeable dragging in comparison to the distinct footprints observed in the Sham group. Conversely, rats treated with BMMSCs showed an increase in hind paw prints and overlapping fore and hind paw prints compared to the SCI group (Figure 2D). Quantitative assessments further revealed that the BMMSCs group exhibited a notable decrease in bases of support and stride lengths, both of which were statistically significant (p<0.05; p<0.0001; Figure 2E, F). Altogether, the findings underscore the therapeutic potential of BMMSCs in enhancing motor function restoration in rats with SCI.
4.3 BMMSCs promote histological repair of the damaged spinal cord.
Histological alterations in spinal cord morphology were assessed via HE staining. Comparative analysis unveiled conspicuous deformities and cavitations accompanied by substantial infiltration of inflammatory cells within the spinal cords of the group with SCI compared to the Sham group, suggesting extensive tissue damage. Following BMMSCs treatment, spinal cord integrity was notably restored, with attenuated infiltration of inflammatory cells and reduced cystic cavity size in contrast to the SCI group (p<0.01; p<0.01; Figure 2G-I). These observations underscore the potential of BMMSCs therapy in mitigating spinal cord tissue injury and facilitating repair processes within the damaged spinal cord.
4.4 BMMSCs inhibit neuronal apoptosis after SCI.
Studies have revealed that attenuating neuronal apoptosis is crucial for improving functional recuperation following SCI[28]. TUNEL staining unveiled a heightened incidence of neuronal loss in the SCI group contrasted with the Sham group, yet therapy utilizing BMMSCs mitigated this phenomenon (Figure 3A). Furthermore, assessment of neuronal preservation via Nissl staining revealed a greater density of intact neurons in the Sham group, in contrast to the presence of atrophied neuronal cell bodies and Nysted granule lysis in the SCI group. Notably, BMMSCs treatment significantly augmented neuronal numbers and restored neuronal morphology (Figure 3B), a finding corroborated by immunofluorescence staining (p<0.05; Figure 3C-D). Western blotting further substantiated the BMMSCs treatment partially reinstated the protein expression of NeuN (p<0.001, Figure 3E-F). Additionally, the BMMSCs group demonstrated decreased levels of the proapoptotic protein BAX and heightened expression of the antiapoptotic protein Bcl-xL in contrast to the SCI group (p<0.001; p<0.01; Figure 3G-I). To summarize, our results indicate that BMMSCs possess the capacity to effectively suppress neuronal apoptosis within the injured spinal cord.
4.5 BMMSCs protect neurons and promote axonal regeneration.
Microtubule-associated protein 2 (MAP2) serves as a mature neuronal marker crucial for regulating microtubule architecture, stability, and organelle transport within axonal and dendritic compartments. Immunofluorescence staining, coupled with quantitative analysis, revealed a reduced population of MAP2-positive axons within the SCI group in comparison to the Sham group. Additionally, compared to the Sham group, the SCI group showed a notable increase in the distance between the epicentre of the injury and the nearest neuron (p <0.01; Figure 4A-C). Conversely, the BMMSCs group exhibited heightened density and intensity of MAP2-positive axons, facilitating axonal growth towards the injury site (p <0.05; p <0.05; Figure 4A-C). Growth-associated protein 43 (GAP43), pivotal in nerve regeneration and axon guidance, plays a crucial role in fostering new axonal connections[29]. Notably, the SCI group displayed a dearth of significant GAP43-positive axons within the fibrotic scar encircled by activated astrocytes, whereas BMMSCs treatment engendered a notable increase in GAP43-positive axons compared to the SCI group (p <0.05; Figure 4D, E). In line with the results observed in immunofluorescence analysis, Western blotting corroborated the upregulation of MAP2 and GAP43 expression following BMMSCs intervention (p <0.05; p <0.05; Figure 3F-H). Collectively, these outcomes underscore the neuroprotective effects of BMMSCs in safeguarding neurons and fostering axonal regeneration post-SCI.
4.6 BMMSCs enhance autophagy after SCI.
Autophagy has garnered attention as an exciting way to treat central nervous system (CNS) diseases, exerting a pivotal role in modulating programmed cell death triggered by neuroinflammation[30]. To evaluate autophagic responses in spinal cord lesions post-SCI, we scrutinized the levels of protein markers associated with autophagosomes LC3II/I, Beclin1, VPS34, CTSD, and the autophagic cargo receptor protein p62 using immunofluorescence and Western blotting. Western blot analysis demonstrated a reduction in p62 protein levels concurrent with LC3II/I, Beclin1, VPS34 and CTSD upregulated after treatment with BMMSCs (p <0.01; p <0.05; p <0.05; p <0.05; p <0.001; Figure 5A-F). Furthermore, immunofluorescence demonstrated diminished p62 density and heightened LC3II signal within the BMMSCs group relative to the SCI group (p <0.05; p <0.01; Figure 5 G-J). BMMSCs not only augmented levels of autophagosome and autophagolysosome-associated markers but also alleviated the accumulation of autophagic substrates, potentially attributable to the overall enhancement of autophagic activity instigated by BMMSCs post-SCI.
4.7 BMMSCs enhance mitophagy after SCI.
To further substantiate the influence of BMMSCs on autophagic processes in SCI rats, we performed TEM analysis on sections of spinal cord tissue. TEM assessment unveiled a notable escalation with an abundance of autophagosomes within the BMMSCs group relative to the SCI group, as denoted by the red arrows, indicative of BMMSCs-induced autophagy activation (Figure 6A). Additionally, we observed alterations in mitochondrial morphology in the BMMSCs group. Contrasting with the oval-shaped mitochondria with internal cristae observed in the Sham and SCI groups, the mitochondria exhibited a contracted and rounded appearance post-BMMSCs treatment, suggestive of mitochondrial stress. Prior investigations[31] have underscored the intricate interplay between autophagy and mitochondrial autophagy. Hence, we postulated that BMMSCs could potentiate mitochondrial autophagy, potentially through autophagy activation. To validate this conjecture, Parkin, BNIP3 and Nix expression was assessed in all groups via Western blot analysis and immunofluorescence. Immunofluorescence analyses revealed a higher abundance of Nix-positive neurons in the Sham group relative to the SCI group, with a significant augmentation in Nix-positive neurons following BMMSCs treatment (p <0.001; p <0.05; Figure 6 B-C). The Western blotting unveiled revealed heightened expressions of Parkin, BNIP3, and Nix proteins within the SCI group as opposed to the Sham group (p <0.001; p <0.05; p <0.0001, Figure 6 D-G). Notably, the BMMSCs group exhibited enhanced protein expression levels of the markers in comparison to the SCI group (p <0.01; p <0.01; p <0.01; Figure 6 D-G). Collectively, these findings suggest that BMMSCs augment mitochondrial autophagy, potentially through their pro-autophagic properties.
4.8 BMMSCs promote autophagy through the AMPK, MAPK and PI3K/AKT
/mTOR/TFEB signaling cascade to promote functional recuperation following SCI
Our study aimed to clarify the fundamental mechanisms through which BMMSCs modulate autophagy and mitochondrial autophagy. TFEB serves as a pivotal regulator within the autophagy-lysosomal signaling cascade. Its potential involvement in autophagy regulation was investigated. In the SCI group, Western blot analysis unveiled heightened expression of p-AMPK and augmented the relocation of TFEB to the nucleus, concomitant with reduced levels of p-mTOR compared to the Sham group (p <0.05; p <0.05; p <0.0001; Figure 7 A-B, G-J). Remarkably, BMMSCs treatment further augmented p-AMPK levels and nuclear translocation of TFEB while diminishing p-mTOR levels (p <0.01; p <0.01; p <0.01; Figure 7 A-B, G-J). Moreover, the investigation revealed that BMMSCs facilitated the stimulation of Erk, JNK, and p38 MAPK signaling cascade, as evidenced through elevated levels of p-Erk, p-JNK, and p-P38 relative to the SCI group (p <0.05; p <0.01; p <0.001; Figure 7 C-D). Furthermore, BMMSCs were observed to attenuate the levels of phosphorylated PI3K and AKT (p <0.01; p <0.01; Figure 7 E-F). Taken together, these findings suggest that the activation of the AMPK, Erk/JNK/P38, and PI3K/AKT pathways inhibits mTOR activation and facilitates TFEB dephosphorylation, thereby enhancing mitochondrial autophagy in the context of SCI (Figure 7 G-J).
4.9 MiR-202-3p mimics inhibit apoptosis and promote autophagy in vivo and in vitro.
Our previous study illustrated that in vitro overexpression of miR-202-3p led to a notable reduction of pro-inflammatory cytokines levels such as TNF-α, IL-6, and IL-1β, whereas downregulation of miR-202-3p expression resulted in an elevation in these cytokines. This observation provides evidence that miR-202-3p may be implicated in the reparative processes associated with SCI. Consequently, we hypothesized that BMMSCs may exert their effects through miR-202-3p-mediated autophagy. In vivo, our examination of spinal cord tissue disclosed markedly elevated levels of miR-202-3p in the BMMSCs treatment group relative to the SCI group (p <0.0001; Figure 8A). To better elucidate the impact of miR-202-3p on neuronal cells, PC-12 cells underwent transfection with miR-202-3p mimics, miR-202-3p inhibitor, and miR-202-3p NC. The transfection experiments yielded compelling evidence indicating a substantial upregulation of miR-202-3p expression within the group transfected with miR-202-3p mimics. Notably, the inhibitor's mechanism of action involves sequestering endogenous free miRNA rather than degrading it, resulting in an insignificant decrease in miR-202-3p levels in the inhibitor group (Figure 8 B-C). Subsequently, we explored how miR-202-3p may modulate autophagy and apoptosis after SCI, given its crucial function in preserving cellular balance and enhancing cell survival under challenging circumstances. Western blot analysis revealed that transfection with miR-202-3p mimics upregulated LC3-II/I protein expression and downregulated p62 and BAX protein expression. Conversely, transfection with the miR-202-3p inhibitor led to the downregulation of LC3-II/I protein expression and the upregulation of p62 and BAX protein expression (Figure 8 D-F). Notably, the pro-autophagic and anti-apoptotic effects of miR-202-3p were more pronounced with increasing concentrations of miR-202-3p mimics (Figure 8 G-I). In summary, our findings indicate that miR-202-3p can suppress apoptosis and enhance autophagy both in vivo and in vitro.