Keywords: cellular communication, competing endogenous RNA, exosomes, Foxp1, necroptosis, neural stem cells, Slc16a3, spinal cord injury, transcriptome sequencing, Tsc2
Abstract
We previously demonstrated that inhibiting neural stem cells necroptosis enhances functional recovery after spinal cord injury. While exosomes are recognized as playing a pivotal role in neural stem cells exocrine function, their precise function in spinal cord injury remains unclear. To investigate the role of exosomes generated following neural stem cells necroptosis after spinal cord injury, we conducted single-cell RNA sequencing and validated that neural stem cells originate from ependymal cells and undergo necroptosis in response to spinal cord injury. Subsequently, we established an in vitro necroptosis model using neural stem cells isolated from embryonic mice aged 16–17 days and extracted exosomes. The results showed that necroptosis did not significantly impact the fundamental characteristics or number of exosomes. Transcriptome sequencing of exosomes in necroptosis group identified 108 differentially expressed messenger RNAs, 104 long non-coding RNAs, 720 circular RNAs, and 14 microRNAs compared with the control group. Construction of a competing endogenous RNA network identified the following hub genes: tuberous sclerosis 2 (Tsc2), solute carrier family 16 member 3 (Slc16a3), and forkhead box protein P1 (Foxp1). Notably, a significant elevation in TSC2 expression was observed in spinal cord tissues following spinal cord injury. TSC2-positive cells were localized around SRY-box transcription factor 2–positive cells within the injury zone. Furthermore, in vitro analysis revealed increased TSC2 expression in exosomal receptor cells compared with other cells. Further assessment of cellular communication following spinal cord injury showed that Tsc2 was involved in ependymal cellular communication at 1 and 3 days post-injury through the epidermal growth factor and midkine signaling pathways. In addition, Slc16a3 participated in cellular communication in ependymal cells at 7 days post-injury via the vascular endothelial growth factor and macrophage migration inhibitory factor signaling pathways. Collectively, these findings confirm that exosomes derived from neural stem cells undergoing necroptosis play an important role in cellular communication after spinal cord injury and induce TSC2 upregulation in recipient cells.
Introduction
Spinal cord injuries (SCI), primarily resulting from falls and traffic accidents, constitute a substantial portion of the global injury burden. The incidence of SCI is projected to rise due to escalating population density, an aging demographic, and increased motor vehicle usage. The need for expensive, complex medical care and the decreased productivity of individuals with SCI significantly strain the healthcare system. In addition, the irreversibility of primary injury, the complexities of secondary injury mechanisms, and the limited regenerative potential of the spinal cord significantly contribute to the unfavorable prognosis of SCI (Fan et al., 2018; Hellenbrand et al., 2024).
Neural stem cells (NSCs), situated in the periventricular zone encircling the central canal of the spinal cord (Fauser et al., 2021), are capable of self-renewal and differentiation into a diverse array of cell types, as verified by in vitro “neurosphere” assay (Watanabe et al., 2021). Definitive NSCs (dNSCs) predominate in the adult spinal cord, originating along the neuraxis at the embryonic day (E) 16.5 and persisting in the periventricular zone into adulthood (Marqués-Torrejón et al., 2021). dNSCs are characterized by the expression of specific markers, such as the intermediate filament protein nestin (Shu et al., 2022) and vimentin (Morrow et al., 2020), as well as the transcription factor SRY-box transcription factor 2 (SOX2) and the cilia marker forkhead box J1 (FoxJ1) (Gilbert et al., 2022). There is controversy over whether ependymal cells serve as the primary source of dNSCs (Furube et al., 2020).
Necroptosis is a type of programmed cell death distinct from apoptosis, characterized by necrotic morphological features that occur independent of caspase-8 (Yan et al., 2023). Necroptosis is mediated by the necrosome, which consists of mixed lineage kinase domain-like protein (MLKL) (Zhan et al., 2021), receptor-interacting protein kinase 1 (RIPK1) (Yuan et al., 2019), and RIPK3 (Morgan and Kim, 2022; Ye et al., 2023). We previously showed that necroptosis occurs in nestin-positive cells following SCI in mice. Moreover, inhibiting necroptosis is associated with enhanced recovery from SCI (Tong et al., 2023). However, our earlier studies focused on NSC depletion post-necroptosis and did not explore the impact of necroptosis on NSC secretory function.
Exosomes, cellular vesicles that range in size from 30 to 200 nm, are integral to the secretome. Carrying diverse components such as nucleic acids, proteins, and lipids, exosomes play a crucial role in intercellular communication (Harmati et al., 2019; Walbrecq et al., 2020). Recent studies indicate that exosomes from mouse embryonic fibroblasts undergoing necroptosis carry the necroptosis-associated proteins RIPK3 and MLKL. Additionally, these necroptosis-associated exosomes are linked to calcium ion depletion and lysosomal rupture (Gupta et al., 2022). Notably, the impact of necroptosis on exosomal transcriptome has not been reported.
The aims of this study were to examine alterations in the exosome transcriptome following necroptosis in NSCs, to identify hub genes associated with necroptosis in NSCs, and to determine whether these hub genes are involved in cellular communication after SCI.
Methods
Animals
Pregnant female mice (E 16–17) and 7-week-old male C57BL/6J mice weighing 20–25 g were procured from Liaoning Changsheng Biotechnology Company (Benxi, Liaoning, China, license No. SCXK (Liao) 2020-0001). All experiments involving animals were approved by the Institutional Animal Care and Use Committee of Sun Yat-sen University (approval No. SYSU-IACUC-2021-000196) on April 19, 2021. The mice had ad libitum access to food and water and were housed in a controlled environment with a constant temperature of 24 ± 2°C, humidity maintained at 55% ± 5%, and a 12-hour light/dark cycle throughout the study.
Neural stem cell isolation and culture
Pregnant mice were initially anesthetized with 3% isoflurane (RWD, Shenzhen, Guangdong, China) via inhalation and maintained under anesthesia with 1.8% isoflurane. A cesarean section was then performed, and the uterus and placenta were meticulously dissected to extract the fetal mice. Both pregnant and fetal mice were euthanized by an overdose of anesthesia. Fetal mice aged E16 to E17 were immersed in 75% ethanol, a dorsal incision was made through the skin and cartilage, and the spinal cord was acquired. The excised spinal cord was then placed in pre-cooled Dulbecco’s modified Eagle’s medium/Nutrient Mixture F-12 (Gibco, Gardena, CA, USA), rinsed, cut into small pieces, transferred to a 100-μm cell sieve (Falcon, Corning, NY, USA), and rinsed with a generous amount of Dulbecco’s modified Eagle’s medium/Nutrient Mixture F-12. The resulting cell suspension was centrifuged at 300 × g for 5 minutes at 4°C. The pelleted cells were resuspended in Dulbecco’s modified Eagle’s medium/Nutrient Mixture F-12 supplemented with 2% B27 (Gibco), glutamine (0.5 mM, Gibco), penicillin (100 IU/mL), streptomycin (100 mg/mL, Gibco), EGF (20 ng/mL, Peprotech, Rocky Hill, NJ, USA), and bFGF (20 ng/mL, Peprotech) and incubated at 37°C with 5% CO2 for 48 hours to generate neurospheres. Subsequently, the suspension was centrifuged at 250 × g for 5 minutes, and the supernatant was removed to isolate the neurospheres. These neurospheres were then enzymatically digested into single-cell suspensions for passaging; all subsequent experiments used third-generation NSCs (Ceto et al., 2020). NSCs were characterized by light microscopic observation as well as immunofluorescence staining, as described in more detail below.
Neural stem cell necroptosis induction
NSCs were pre-treated with 20 μM zVAD-fmk (MCE, Monmouth Junction, NJ, USA) for 30 minutes, then treated with recombinant mouse tumor necrosis factor α (TNFα, 40 ng/mL, Peprotech) and 4 μM SMAC mimetic (MCE) for 6 hours to establish a TSZ (TNFα, SMAC mimetic, zVAD-fmk)-induced in vitro model of necroptosis. All of the drugs were dissolved in dimethyl sulfoxide (DMSO), and thus the Control group was treated with an equal volume of DMSO, ensuring that concentrations of DMSO did not exceed 0.1% (Gupta et al., 2022).
Isolation of exosomes
The culture medium was harvested and centrifuged sequentially: first at 300 × g at 37°C for 10 minutes, then at 2000 × g for 10 minutes at 4°C, and finally at 10,000 × g for 60 minutes at 4°C. Following centrifugation, the supernatant was passed through a 0.22-μm sterile filter (Falcon) to eliminate cellular debris. The filtrate was ultracentrifuged at 120,000 × g for 70 minutes, the supernatant was discarded, and the precipitate was resuspended in 200 μL of phosphate buffered saline (PBS, Gibco) to obtain the exosomes (Liu et al., 2021).
Characterization of exosomes
Morphological characterization of the exosomes was performed by directly examining their structure via transmission electron microscopy (HT-7700, Hitachi, Tokyo, Japan). Nanoflow technology (N30E, NanoFCM, Xiamen, Fujian, China) was used to measure the exosome sizes and concentration. The presence of markers CD9, CD63, and CD81 was also assessed by Nanoflow technology (Welsh et al., 2024).
Exosome labeling
To fluorescently label the exosomes, 1,1-dioctadecyl-3,3,3,3-tetramethylindodicarbocyanine (4 mg/mL, AAT Bioquest, Sunnyvale, CA, USA) was added to exosomes at a ratio of 1:200 and incubated according the manufacturer’s instructions. Excess dye exosomes were eliminated by ultracentrifuging three times at 100,000 × g for 1 hour at 4°C, resuspending the pellet in PBS between each ultracentrifugation step. The final pellet was then resuspended in PBS (Wiklander et al., 2015).
Whole-transcriptome sequencing of exosomes
Exosomes isolated from NSCs from both the control and TSZ groups (n = 3 for each group) were subjected to whole-transcriptome sequencing using an Illumina platform (San Diego, CA, USA), carried out by Novogene Co. Ltd. (Beijing, China). Total exosome-derived RNA was purified using an exoRNeasy Maxi Kit (Qiagen, Strasse 1, 40724 Hilden, Germany). RNA quality and purity were assessed on 1% agarose gels. The purity of the isolated RNA was further verified using a NanoPhotometer® spectrophotometer (IMPLEN, Schillerstrasse 3, Germering, Germany, 82110). The RNA concentration was determined using a Qubit® RNA Assay Kit and a Qubit® 2.0 Fluorometer (Life Technologies, Carlsbad, CA, USA). Additionally, RNA integrity was evaluated using a RNA Nano 6000 Assay Kit and an Agilent Bioanalyzer 2100 system (Agilent Technologies, Santa Clara, CA, USA). Next, long non-coding RNA (lncRNA) and microRNA (miRNA) sequencing libraries were constructed using the NEBNext Ultra Directional RNA Library Prep Kit (NEB, Ipswich, MA, USA) for Illumina and NEBNext Multiplex Small RNA Library Prep Set for Illumina, respectively, following the protocols provided by the supplier. To construct the lncRNA sequencing library, the RNA was fragmented, first- and second-strand complementary DNA (cDNA) synthesis was performed, and the double-stranded cDNA products were purified and end-repaired, followed by adaptor ligation. The resulting lncRNA library was then amplified by PCR. For miRNA library construction, the RNA fragments were ligated to 3′ and 5′ adapters, RT-PCR was performed to generate cDNA, the sequencing of the miRNA libraries were carried out using the Hiseq2100 Truseq SBS Kit v3-HS (Illumina) (Ulintz et al., 2019; Zhang et al., 2023). The details of the quality control and analysis of the raw data are shown in Additional file 1 (177.7KB, pdf) .
Other data sources and general information
The GSE149669 and GSE162610 datasets were obtained from the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/gds). The GSE149669 dataset was generated from female C57BL/6 mice subjected to acupuncture to induce moderate SCI at the T9–T10 segment, followed by precise extraction of ependymal cells via laser microscopy. The dataset includes the control group (n = 6) and the SCI group 3 day post-SCI (n = 6). The GSE162610 dataset was generated by inducing moderate SCI at the T8 segment in female C57BL/6 mice using Allen’s method. The dataset comprises the control group (n = 5) and three time points (1, 3, and 7 days post-SCI) from the SCI group (n = 5, 3, and 3, respectively).
Ependymal cell RNA sequencing
Initially, GEO2R (https://www.ncbi.nlm./geo/geo2r/) differential analysis was conducted on the GSE149669 dataset, and the gene expression data were sorted based on the order of the gene sets. Subsequently, the R package “fgsea” was used to perform gene set enrichment analysis (GSEA). GSEA enrichment scores and trends were identified by comparing the gene expression data to the programmed cell death-related gene sets (Additional Table 1) and necroptosis-associated gene set (Additional Table 2) from the Kyoto Encyclopedia of Genes and Genomes (KEGG, https://www.kegg.jp) database (Fan et al., 2022; Li et al., 2022).
Additional Table 1.
Four gene sets for programmed cell death from KEGG database
Necroptosis | Apoptosis | Autophagy | Ferroptosis |
---|---|---|---|
H2ab1 | Apaf1 | Atg5 | Alox15 |
H2al2b | Birc3 | Igbp1 | Atg5 |
H2al1a | Birc2 | Ppp2ca | Cp |
H2al1b | Xiap | Ppp2cb | Cybb |
H2al1c | Birc5 | Atg9b | Acsl1 |
H2al1d | Bak1 | Pik3c3 | Fth1 |
H2al1f | Bax | Atg4d | Ftl1 |
H2al1g | Bcl2 | Atg4c | Gclc |
H2al1h | Bcl2l1 | Atg9a | Gclm |
H2al1i | Bid | Ulk2 | Lpcat3 |
Gm38574 | Bcl2l11 | Atg2a | Gss |
Chmp7 | Birc6 | Igbp1b | Hmox1 |
Sharpin | Casp3 | Atg13 | Slc3a2 |
Ticam1 | Casp7 | Wipi1 | Slc11a2 |
Camk2d | Casp8 | Becn1 | Pcbp2 |
Pygb | Casp9 | Gabarap | Prnp |
Pygl | Cycs | Mlst8 | Sat1 |
Parp1 | Cyct | Mtor | Slc39a14 |
Vps4a | Fadd | Gabarapl1 | Acsl6 |
Alox15 | Bbc3 | Atg4b | Trf |
Slc25a4 | Ngfr | Atg4a | Tfrc |
Slc25a5 | Septin4 | Atg10 | Trp53 |
Birc3 | Tnfrsf1a | Atg12 | Vdac2 |
Birc2 | Mapk10 | Atg3 | Vdac3 |
Xiap | Mapk8 | Atg101 | Pcbp1 |
Bax | Mapk9 | Atg7 | Slc7a11 |
Bcl2 | Birc7 | Rptor | Ncoa4 |
Bid | Bok | Wipi2 | Acsl5 |
Camk2a | Becn1 | Pik3r4 | Acsl4 |
Camk2b | Pmaip1 | Atg2b | Slc40a1 |
Camk2g | Htra2 | Atg16l1 | Gpx4 |
Capn1 | Diablo | Gabarapl2 | Map1lc3a |
Capn2 | Map1lc3b | ||
Casp1 | Slc39a8 | ||
Casp8 | Ftmt | ||
Cflar | Steap3 | ||
Cybb | Sat2 | ||
Fadd | Inhca | ||
Fas | Acsl3 | ||
Fasl | Atg7 | ||
Tlr3 | |||
Fth1 | |||
Ftl1 | |||
Glul | |||
Glud1 | |||
H2ac18 | |||
H2ax | |||
Hmgb1 | |||
Hsp90ab1 | |||
Hsp90aa1 | |||
Ifna1 | |||
Ifna11 | |||
Ifna2 | |||
Ifna4 | |||
Ifna5 | |||
Ifna6 | |||
Ifna7 | |||
Ifna9 | |||
Ifnab | |||
Ifnar1 | |||
Ifnar2 | |||
Ifnb1 | |||
Ifng | |||
Ifngr1 | |||
Ifngr2 | |||
Il1a | |||
Il1b | |||
Irf9 | |||
Jak1 | |||
Jak2 | |||
Jak3 | |||
Sqstml | |||
Pla2g4a | |||
Eif2ak2 | |||
Pygm | |||
Ripkl | |||
Vps4b | |||
Smpd1 | |||
Chmp6 | |||
Stat1 | |||
Stat2 | |||
Stat3 | |||
Stat4 | |||
Stat5a | |||
Stat5b | |||
Stat6 | |||
Pla2g4b | |||
Nlrp3 | |||
Tlr4 | |||
Tnf | |||
Tnfaip3 | |||
Tnfrsf10b | |||
Tnfrsf1a | |||
Traf2 | |||
Traf5 | |||
Tnfsf10 | |||
Vdac1 | |||
Vdac2 | |||
Vdac3 | |||
Ticam2 | |||
Ifna13 | |||
Ifna16 | |||
H2aj | |||
Pla2g4c | |||
Chmp1a | |||
H2al1j | |||
Rbck1 | |||
Ifna15 | |||
Ifna12 | |||
Spata2 | |||
Mapk10 | |||
Mapk8 | |||
Mapk9 | |||
Ppia | |||
Rnf31 | |||
Macroh2a1 | |||
Aifm1 | |||
Pla2g4f | |||
Usp21 | |||
H2ac25 | |||
H2ac1 | |||
H2ac6 | |||
H2ac7 | |||
H2ac8 | |||
H2ac11 | |||
H2ac12 | |||
H2ac15 | |||
H2ac22 | |||
H2ac24 | |||
H2ac4 | |||
H2ac10 | |||
H2ac20 | |||
H2ac13 | |||
H2ac19 | |||
Pla2g4e | |||
H2al1o | |||
H2al3 | |||
H2alln | |||
Gm5396 | |||
Ifna14 | |||
Macroh2a2 | |||
H2az1 | |||
H2al1k | |||
H2al1e | |||
Tyk2 | |||
H2al2c | |||
Ripk3 | |||
Zbp1 | |||
Trpm7 | |||
H2ac21 | |||
H2ab2 | |||
H2ab3 | |||
Chmp4c | |||
H2ac23 | |||
Chmp3 | |||
Pycard | |||
Chmp1b2 | |||
Chmp1b | |||
Ppid | |||
H2al2a | |||
Chmp2b | |||
Chmp2a | |||
Tradd | |||
Pgam5 | |||
Slc25a31 | |||
Dnm1l | |||
Cyld | |||
Mlkl | |||
Chmp4b | |||
H2al1m | |||
Chmp5 | |||
Il33 | |||
H2az2 | |||
Pla2g4d | |||
Spata2l |
Additional Table 2.
Marker genes in the necroptosis gene set
Ripkl |
Ripk3 |
Mlkl |
Tnf |
Tnfrsfla |
TnfsflO |
TnfrsflOb |
Fasl |
Fas |
Ifnal |
Ifnarl |
Tlr3 |
Tlr4 |
Zbpl |
Single-cell RNA sequencing
The GSE162610 dataset was analyzed using a variety of R software packages. The “Seurat,” “ggplot2,” “cowplot,” “Matrix,” “dplyr,” “ggsci,” and “SingleR” packages were used for integration, batch correction, dimensionality reduction, clustering, annotation, and plotting of the single-cell RNA-sequencing data. Additionally, the “Seurat,” “dplyr,” “tidyverse,” “stringr,” “CellChat,” “patchwork,” “mindr,” and “svglite” packages were used to analyze and plot cellular communication via analysis of ligand-receptor pair expression among the single-cell RNA-sequencing data. To enhance the data quality, we only included cells with over 400 genes and 1000 transcripts and excluded genes that were expressed in fewer than 10 cells. Additionally, cells for which mitochondrial genes accounted for more than 5% of the RNA reads were excluded. Dimensionality reduction and clustering were achieved through uniform manifold approximation and projection and 16-point principal component analysis (Rosenberg et al., 2018; Sathyamurthy et al., 2018; Milich et al., 2021; Yang et al., 2022). Cell types were named based on the annotated results from the “SingleR” package analysis, the CellMarker database (http://bio-bigdata.hrbmu.edu.cn/CellMarker/), and relevant literature (Aran et al., 2019). Proportions of cellular subtypes were calculated, and hub genes and receptor-ligand pairs were subjected to interaction network analysis using the STRING database (http://string-db.org). Patterns of communication between the different cellular subtypes were then investigated (Szklarczyk et al., 2023). The Uniform Resource Locators of all R packages used are displayed in Additional Table 3.
Additional Table 3.
The R packages and Uniform Resource Locators (URLs) used in this study
Spinal cord injury model establishment
In previous studies, we established an SCI model using male mice, which resulted in better SCI model homogeneity. Consequently, only male mice were used to establish the SCI model used in this study. Mature mice (n = 12) were randomly divided into two groups: the sham group (n = 6), which underwent modified laminectomy only, and the SCI group (n = 6), which was subjected to SCI using Allen’s method, employing a weight-drop contusion mechanism, following modified laminectomy (Wu et al., 2022). Prior to the surgical procedure, all animals were anesthetized with 3% isoflurane inhalation, followed by 1.8% isoflurane inhalation for maintenance of the anesthesia. To fully expose the spinal cord, a laminectomy at the T10 vertebrae was performed via layer-by-layer incision, involving the skin, subcutaneous tissues, and fascia, with separation of the lateral muscular tissues, as previously described. Subsequently, a controlled contusion injury was inflicted using a 10-g hammer in free fall from a height of 4 cm to induce SCI. Mice in the sham group underwent laminectomy only. Post-surgery, urination assistance was provided daily until reestablishment of the micturition reflex.
At 3 days post-SCI, mice were anesthetized with 3% isoflurane and then transcardially perfused with 4% paraformaldehyde (Aladdin, Shanghai, China) or PBS. Subsequently, spinal cord specimens were collected for further analysis.
In vitro cell culture
The neuroblastoma × spinal cord hybridoma cell line (NSC34, RRID: CVCL_D356) was provided by the Xiayu group at the Seventh Affiliated Hospital of Sun Yat-sen University. Cells were cultured and maintained in complete Dulbecco’s modified Eagle’s medium (Gibco, C11995500BT) supplemented with 10% Fetalgro bovine growth serum (Gibco, 10091148) and 1% penicillin-streptomycin (Gibco, 15140-122) (Khosla et al., 2023).
Flow cytometry
A Vybrant FAM Caspase-8 Assay Kit (Thermo Fisher Scientific, Waltham, MA, USA) was used for cell death analysis. NSCs were rinsed with the provided rinse solution and subsequently treated with 150× FLICA (Caspase-8, Thermo Fisher Scientific) for 60 minutes at 37°C. Following another round of washing with the rinse solution, 5 μL of propidium iodide (PI, Thermo Fisher Scientific) was added, and the cells were incubated on ice for 10 minutes. Subsequently, flow cytometry data were acquired using a CytoFLEX LX instrument (Beckman Coulter, Brea, CA, USA). Flowjo software (FlowJo, Ashland, OR, USA, version 8.0.2) was used for data analysis (Fritsch et al., 2019; Gupta et al., 2022).
Immunofluorescence staining
Spinal cords harvested from mice 3 days post-SCI were fixed by immersing in 4% paraformaldehyde for 1 day. Next, each specimen was sequentially transferred to 10%, 20%, and 30% sucrose (Biofroxx, Shanghai, China) solutions for gradient dehydration, over 96 hours. The dehydrated tissues were embedded in optimal cutting temperature compound (SAKURA, Tokyo, Japan) and sectioned using a frozen sectioning machine (Leica, Wetzlar, Germany). Treated NSCs and NSC-34 cells and the frozen tissue sections were fixed in pre-cooled paraformaldehyde (4%, w/v) for 20 minutes, then permeabilized with 0.2% Triton X-100 (Aladdin) for an additional 20 minutes, followed by blocking with 10% normal goat serum. Finally, the cells were incubated overnight at 4°C with the following primary antibodies: anti-Nestin (1:500, mouse IgG, Abcam, Cambridge, UK, Cat# ab6142, RRID: AB_305313), anti-SOX2 (1:500, rabbit IgG, Cell Signaling, Boston, MA, USA, Cat# 23064, RRID: AB_2714146), anti-SOX2 (1:500, mouse IgG, Abcam, Cat# ab171380, RRID: AB_2732072), and anti-tuberous sclerosis 2 (TSC2, 1:500, rabbit IgG, Cell Signaling, Cat# 4308, RRID: AB_10547134). The next day, the cells and frozen tissue sections were incubated at 37°C for 1 hour with the following secondary antibodies: Alexa Fluor 488 (goat anti rabbit IgG, 5 µg/mL, Thermo Fisher Scientific, Cat# A-11034, RRID: AB_2576217) and Alexa Fluor 594 (goat anti mouse IgG, 5 µg/mL, Thermo Fisher Scientific, Cat# A-11005, RRID: AB_2534073). Finally, the nuclei were counterstained with 4′,6-diamidino-2-phenylindole (Abcam) for 10 minutes. The immunoreactivity was visualized using a confocal fluorescence microscope (Carl Zeiss, Oberkochen, Germany) and a pathology slide scanner (Olympus, Tokyo, Japan).
Western blotting
Total protein was extracted from lysed NSC-34 cells or homogenized spinal cord tissues harvested 3 days post-SCI using radioimmune precipitation assay (Thermo Fisher Scientific) lysis buffer containing a mixture of protease and phosphatase inhibitors. Subsequently, the proteins were separated by polyacrylamide gel electrophoresis and transferred to polyvinylidene fluoride membranes using an iBlot 3 Dry Blotting System (Thermo Fisher Scientific). The membranes were probed with primary antibodies, including anti-solute carrier family 16 member 3 (SLC16A3, 1:1000, rabbit IgG, Thermo Fisher Scientific, Cat# PA587977, RRID: AB_2804558), anti-Forkhead box protein P1 (FOXP1, 1:1000, rabbit IgG, Abcam, Cat# ab134055, RRID: AB_2107103), anti-TSC2 (1:1000, rabbit IgG, Cell Signaling, Cat# 4308, RRID: AB_10547134), and anti-glyceraldehyde-3-phosphate dehydrogenase (GAPDH, 1:2000, rabbit IgG, Cell Signaling, Cat# 2118S, RRID: AB_561053) overnight at 4°C. Next, the membranes were incubated with a horseradish peroxidase–conjugated rabbit-specific antibody (1:3000, Cell Signaling, Cat# 7074, RRID: AB_2099233) for 1 hour at 37°C. The resulting bands were visualized using an enhanced chemiluminescence reagent (Thermo Fisher Scientific) and a chemiluminescent imaging system (ChemiDoc, BIO-RAD, Hercules, CA, USA). Relative optical density was evaluated using ImageJ Software (version 1.53, National Institutes of Health, Bethesda, MD, USA; Schneider et al., 2012). TSC2, SLC16A3, and FOXP1 expression levels were normalized to GAPDH.
Statistical analysis
No statistical methods were used to predetermine sample sizes. However, our sample sizes are similar to those reported in a previous publication (Tong et al., 2023). No animals or data points were excluded from the analysis. Data collection and analysis were carried out by investigators blinded to the group assignments. Statistical analysis was performed using GraphPad Prism version 8.0.2 for Windows (GraphPad Software, Boston, MA, USA, www.graphpad.com). The data reporting method varies depending on the sample size and the type of analysis conducted. Western blotting and immunofluorescence data are presented as mean ± standard deviation (SD). Conversely, RNA-sequencing and single-cell RNA-sequencing data are reported as mean ± standard error of mean (SEM). Two-tailed unpaired Student’s t-test was employed when comparing two groups. For multiple groups, one-way analysis of variance was used, followed by Bonferroni’s post hoc test. Statistical significance was defined as P < 0.05.
Results
Identification of neural stem cells mainly deriving from ependymal cells and undergoing necroptosis
To ascertain if NSCs originate from ependymal cells and undergo necroptosis, we analyzed the GSE162610 and GSE149669 datasets. We constructed an SCI single-cell atlas from the GSE162610 single-cell RNA-sequencing data that showed 29 distinct cell clusters (Figure 1A). Subsequent annotation of these clusters resulted in the identification of 15 distinct cell types (Figure 1B and C). Ependymal cells exhibited high levels of Sox2 and Foxj1 expression. Additionally, Vimentin was expressed in a wide range of cells, excluding oligodendrocyte progenitor cells (OPCs) and microglia, and was expressed at particularly high levels in ependymal cells. Nestin expression was detected in both endothelial and ependymal cells. Notably, a subset of ependymal cells that expressed high levels of vimentin also expressed Nestin (Figure 1D). Examination of necroptosis-associated gene expression showed that ependymal cells expressed high levels of the key necroptosis genes Ripk3 and Mlkl at various time points following SCI. Additionally, the tumor necrosis factor receptor superfamily member 1A (Tnfrsf1a) and tumor necrosis factor receptor superfamily member 6 (Tnfsf6/Fas) ligand genes demonstrated elevated expression after SCI, with expression peaking 1 day after SCI (Figure 1E). Furthermore, GSEA revealed enrichment of three gene sets, including necroptosis, apoptosis, and ferroptosis, in isolated ependymal cells 3 days after SCI (Figure 1F). Similarly, expression of the ligand-encoding gene Tnfrsf1a was highest in isolated ependymal cells, consistent with the single-cell RNA-sequencing results (Figure 1G). On this basis, we inferred that ependymal cells primarily undergo necroptosis after SCI through the tumor necrosis factor (TNF) signaling pathway. Therefore, TSZ treatment (TNF receptor–induced initiation) was used to induce necroptosis in our in vitro system.
Figure 1.
Identification of NSCs mainly derived from ependymal cells and undergoing necroptosis.
(A) UMAP plot of single-cell type clusters in SCI. (B) Annotated UMAP plot of single-cell type clusters in SCI. (C) Bubble plot of cell type marker gene expression in SCI. (D) Expression of NSC-related marker genes as determined by single-cell RNA-sequencing. (E) The expression of key genes and ligand receptor–related genes associated with necroptosis in ependymal cells was examined in the sham group and 1, 3, 7 days following SCI. (F) GSEA of PCD in ependymal cells (3 days post-SCI vs. control). (G) Enrichment of key genes and ligand receptor–related genes associated with necroptosis as assessed by GSEA. GSEA: Gene set enrichment analysis; NSC: neural stem cell; PCD: programmed cell death; SCI: spinal cord injury; UMAP: uniform manifold approximation and projection.
Identification of neural stem cells undergoing necroptosis in vitro and characterization of exosomes
To characterize the exosomes released subsequent to necroptosis in NSCs, we isolated NSCs, induced necroptosis, and collected the exosomes released by the NSCs. The NSCs formed neurospheres in vitro that exhibited a typical spherical shape and refractive index (Figure 2A). Both the neurospheres and individual cells express the NSC markers SOX2 and Nestin (Figure 2B). TSZ treatment induced NSC necroptosis that was characterized by a notable rise in the count of PI-positive and caspase-8–negative cells (Figure 2C). Exosomes from both the control and TSZ groups exhibited a classical “Teato-like” (semi-concave on one side) bilayer membrane structure, as observed by transmission electron microscopy. Furthermore, nanoflow analysis showed that these exosomes demonstrated particle sizes ranging from 50 to 150 nm, with a concentration of 4.48 × 109 particles/mL in the control group and 5.1 × 109 particles/mL in the TSZ group (Figure 2D and E). We validated the expression of the three surface markers CD9, CD63, and CD81 on exosomes via nanoflow analysis and observed that CD9 expression was the highest among the three markers in both the control and TSZ groups, followed by CD81 (Figure 2F).
Figure 2.
Validation of the necroptosis model and characterization of NSCs and exosomes.
(A) Characterization of NSC neurospheres. The NSCs displayed a typical spherical neurosphere morphology. Scale bars: 500 μm (upper) and 100 μm (lower). (B) Representative immunofluorescence images of SOX2 (green, Alexa Fluor 488) and Nestin (red, Alexa Fluor 594) expression in NSCs. Both neurospheres and individual NSCs expressed SOX2 and Nestin. Cell nuclei were counterstained with DAPI (blue). Scale bar: 100 μm. (C) NSCs were treated with DMSO or TSZ for 8 hours, after which cell death was analyzed by flow cytometry. (D) Exosome morphology as assessed by transmission electron microscopy. The exosomes in both the NSC-control and NSC-TSZ groups exhibited a typical one-sided semi-concave bilayer membrane structure. Scale bars: 200 nm. (E, F) Nanoflow cytometry detection of particle size distribution, concentration, and exosome surface marker expression. DAPI: 4′,6-Diamidino-2-phenylindole; DMSO: dimethyl sulfoxide; NSC: neural stem cell; SOX2: SRY-box transcription factor 2; TSZ: tumor necrosis factor α, SMAC mimetic, zVAD-fmk.
Identification, KEGG pathway and GO enrichment analysis of differentially expressed mRNAs, lncRNAs, circRNAs, and miRNAs
To investigate the impact of necroptosis on the NSC-derived exosome transcriptome, we conducted RNA sequencing of exosomes obtained from both the control and TSZ groups, performed Venn diagram analysis (Additional Figure 1 (4.3MB, tif) A), and analyzed the differential expression of messenger RNAs (DEmRNAs), lncRNAs (DElncRNAs), circular RNAs (DEcircRNAs), and miRNAs (DEmiRNAs). We identified 108 DEmRNAs (60 down-regulated and 48 up-regulated), 104 DElncRNAs (55 down- regulated and 49 up-regulated), 720 DEcircRNAs (253 down- regulated and 647 up-regulated), and 14 DEmiRNAs (9 down- regulated and 5 up- regulated) (Additional Figure 2 (6.4MB, tif) A). Clustering maps of the DEmRNAs, DElncRNAs, DEcircRNAs, and DEmiRNAs indicated clear differentiation between the control and TSZ samples (Additional Figure 1 (4.3MB, tif) B).
The top 20 KEGG pathways enriched in DEmRNAs, as well as target genes of the DEmRNAs, DElncRNAs, and DEcircRNAs, are presented as bubble plots in Additional Figure 2 (6.4MB, tif) B–D. Among them, the “Ubiquitin-mediated proteolysis” and “Autophagy” pathways were significantly enriched in DEmRNAs, DElncRNAs, and DEcircRNAs. The “Thermogenesis” and “Spliceosome” pathways were significantly enriched in DEmRNAs and DElncRNAs. The “Neurotrophin signaling pathway” was significantly enriched in DEmRNAs and DEcircRNAs. Additionally, the “Chronic myeloid leukemia” pathway was significantly enriched in DEcircRNAs, while only the “MAPK signaling pathway” was significantly enriched in DEmiRNAs.
Additional Figure 3 (6.3MB, tif) displays bar charts of the top 20 results of the GO analysis for the three fundamental classifications. For biological processes, “cellular process” was significantly enriched in DEmRNAs, DElncRNAs, DEcircRNAs, and DEmiRNAs. For cellular components, “cell part” and “intracellular part” were significantly enriched in all four groups, which could be because the transcriptome originated from exosomes. For molecular functions, “protein binding” was the most enriched in all four groups, suggesting that DEmRNAs, DElncRNAs, DEcircRNAs, and DEmiRNAs primarily function through protein binding.
Identification of hub genes by whole-transcriptome sequencing analysis of DEmRNAs, DElncRNAs, DEcircRNAs, and DEmiRNAs
To comprehensively analyze the roles of DEmRNAs, DElncRNAs, DEcircRNAs, and DEmiRNAs, we performed whole-transcriptome analysis. According to the competing endogenous RNA (ceRNA) theory, we identified lncRNA/circRNA–target gene pairs sharing the same miRNA binding site. We then established the lncRNA-miRNA-mRNA/circRNA-miRNA-mRNA regulatory relationships with lncRNA/circRNA as the decoy, miRNA as the core, and mRNA as the target and visualized the ceRNA network using Cytoscape software.
Following target gene prediction pairing, we constructed a regulatory network comprising 52 lncRNAs/74 circRNAs, 6 miRNAs, and 14 mRNAs (Additional Tables 4 and Additional Table 5 (186.4KB, pdf) ). On the basis of the competing ceRNA theory and the whole-transcriptome sequencing network, we generated two ceRNA networks: “down (lncRNA/circRNA)-up (miRNA)-down (mRNA)” and “up (lncRNA/circRNA)-down (miRNA)-up (mRNA).” The “down-up-down” network consisted of 21 down-regulated lncRNAs/37 down-regulated circRNAs, 3 up-regulated miRNAs, and 2 down-regulated mRNAs. The up-down-up network included 14 up-regulated lncRNAs/15 up-regulated circRNAs, 1 down-regulated miRNA, and 1 up-regulated mRNA (Figure 3A and B).
Additional Table 4.
Whole transcriptome sequencing network of LncRNA-miRNA- mRNA
RNA id | RNA name | Type | Up down |
---|---|---|---|
TCONS 00006110 | ENSMUSG00000026469-AS3 | LncRNA | Up |
TCONS 00227925 | ENSMUSG00000031788-AS3 | LncRNA | Down |
TCONS 00105206 | Prr3-OT1 | LncRNA | Down |
TCONS 00168233 | E130114P18Rik-OT7 | LncRNA | Down |
TCONS 00172228 | Smim1-OT1 | LncRNA | Up |
TCONS_00168236 | E130114P18Rik-OT5 | LncRNA | Down |
TCONS_00168237 | ENSMUSG00000028565-AS1 | LncRNA | Up |
TCONS_00168235 | E130114P18Rik-OT4 | LncRNA | Down |
TCONS_00060387 | Cd180-OT4 | LncRNA | Down |
TCONS_00123925 | ENSMUSG00000058835-AS4 | LncRNA | Down |
TCONS_00060389 | Cd180-OT3 | LncRNA | Down |
TCONS 00063475 | LINC328 | LncRNA | Up |
TCONS 00063473 | LINC330 | LncRNA | Down |
ENSMUST00000225342 | Hdgfl2-208 | LncRNA | Up |
TCONS 00015967 | LINC60 | LncRNA | Up |
TCONS 00103582 | Fam234a-OT1 | LncRNA | Down |
TCONS 00100465 | Crb3-OT2 | LncRNA | Down |
TCONS 00015968 | LINC63 | LncRNA | Down |
TCONS 00133538 | Dnajc1-OT6 | LncRNA | Up |
TCONS 00079175 | Chrac1-OT2 | LncRNA | Down |
TCONS 00061523 | Tbce-OT2 | LncRNA | Up |
ENSMUSG00000045503 | Sys1 | mRNA | Down |
TCONS 00088884 | Ncbp2-OT5 | LncRNA | Up |
TCONS 00227132 | Trmt1-OT6 | LncRNA | Up |
ENSMUSG00000002496 | Tsc2 | mRNA | Up |
TCONS 00033267 | Abhd15-OT2 | LncRNA | Down |
TCONS 00144648 | ENSMUSG00000037111-AS1 | LncRNA | Up |
mmu-miR-27b-3p | mmu-miR-27b-3p | miRNA | Down |
TCONS 00236182 | ENSMUSG00000043051-AS1 | LncRNA | Down |
mmu-miR-378c | mmu-miR-378c | miRNA | Up |
TCONS 00059908 | Gm20379-OT2 | lncRNA | Up |
TCONS 00033979 | LINC166 | lncRNA | Up |
mmu-miR-466i-5p | mmu-miR-466i-5p | miRNA | Down |
TCONS 00157470 | Gm12353-OT2 | LncRNA | Down |
TCONS 00157471 | Gm12353-OT1 | LncRNA | Down |
TCONS 00128180 | Cstf3-OT1 | LncRNA | Down |
ENSMUST00000231067 | Lmf2-208 | LncRNA | Up |
ENSMUST00000167718 | Cct3-207 | LncRNA | Up |
TCONS 00181761 | E130116L18Rik-OT2 | LncRNA | Up |
TCONS 00072993 | Cacna1d-OT1 | LncRNA | Up |
mmu-miR-320-3p | mmu-miR-320-3p | miRNA | Up |
TCONS 00166885 | ENSMUSG00000038827-AS2 | LncRNA | Up |
ENSMUST00000171017 | Racgap1-212 | LncRNA | Down |
ENSMUSG00000027323 | Rad51 | mRNA | Down |
TCONS 00262406 | LINC1373 | LncRNA | Up |
TCONS 00026283 | ENSMUSG00000061589-AS1 | LncRNA | Down |
TCONS 00263509 | LINC1399 | LncRNA | Down |
TCONS 00102440 | Dynlt2a2-OT7 | LncRNA | Up |
ENSMUSG00000108348 | Pnma8c | mRNA | Down |
TCONS 00143114 | ENSMUSG00000027589-AS2 | LncRNA | Down |
TCONS 00057742 | LINC271 | LncRNA | Up |
ENSMUSG00000030067 | Foxp1 | mRNA | Down |
ENSMUSG00000038729 | Pakap | mRNA | Down |
ENSMUSG00000027339 | Rassf2 | mRNA | Up |
ENSMUST00000209186 | Rtn2-203 | LncRNA | Down |
ENSMUSG00000035367 | Rmi1 | mRNA | Down |
TCONS 00033271 | Abhd15-OT3 | LncRNA | Down |
TCONS 00033270 | Abhd15-OT4 | LncRNA | Up |
ENSMUSG00000029769 | Ccdc136 | mRNA | Up |
TCONS 00022924 | Zbtb2-OT1 | LncRNA | Up |
mmu-miR-1306-3p | mmu-miR-1306-3p | miRNA | Up |
TCONS 00110943 | Ldlrad4-OT2 | LncRNA | Down |
TCONS 00093162 | Slc51a-OT1 | LncRNA | Down |
TCONS_00123934 | ENSMUSG00000058835-AS2 | LncRNA | Down |
TCONS 00244613 | Kri1-OT4 | LncRNA | Down |
ENSMUST00000159935 | Ptger1-202 | LncRNA | Up |
TCONS 00085942 | ENSMUSG00000022623-AS1 | LncRNA | Down |
TCONS 00095752 | Dynlt1c-OT4 | LncRNA | Down |
TCONS 00115178 | Zfp236-OT4 | LncRNA | Down |
TCONS 00115176 | Zfp236-OT2 | LncRNA | Up |
ENSMUSG00000025161 | Slc16a3 | mRNA | Down |
TCONS 00092103 | ENSMUSG00000026181-AS2 | LncRNA | Up |
TCONS 00092104 | ENSMUSG00000026181-AS1 | LncRNA | Up |
TCONS 00123924 | ENSMUSG00000058835-AS3 | LncRNA | Down |
ENSMUSG00000061175 | Fnip2 | mRNA | Up |
TCONS 00127784 | 1700029115Rik-OT1 | LncRNA | Up |
TCONS 00085301 | Ndufa6-OT3 | LncRNA | Up |
ENSMUSG00000039000 | Ube3c | mRNA | Up |
ENSMUSG00000025402 | Nab2 | mRNA | Down |
TCONS 00033272 | Abhd15-OT6 | LncRNA | Down |
TCONS 00095749 | Dynlt1c-OT3 | LncRNA | Up |
TCONS 00010465 | ENSMUSG00000026782-AS2 | LncRNA | Down |
TCONS 00010464 | ENSMUSG00000026782-AS1 | LncRNA | Up |
TCONS 00077020 | Gpr183-OT3 | LncRNA | Down |
TCONS 00147606 | St7l-OT2 | LncRNA | Down |
ENSMUSG00000030512 | Snrpa1 | mRNA | Up |
ENSMUST00000130120 | Jmjd6-203 | LncRNA | Up |
TCONS 00151642 | Gm21954-OT3 | LncRNA | Up |
mmu-miR-378a-3p | mmu-miR-378a-3p | miRNA | Up |
TCONS 00087226 | Glis2-OT3 | LncRNA | Up |
TCONS 00181129 | LINC943 | LncRNA | Up |
TCONS 00032852 | Ankfy1-OT1 | LncRNA | Up |
TCONS 00022183 | ENSMUSG00000025795-AS2 | LncRNA | Up |
Figure 3.
WTS network construction and the identification and validation of hub genes.
(A, B) Construction of the WTS regulatory network, which was subsequently divided into two ceRNA networks (down-up-down/up-down-up). In these networks, circles represent miRNAs, squares represent mRNAs, and triangles represent lncRNAs and circRNAs. Red indicates up-regulation, while green indicates down-regulation. (C) Bidirectional hierarchical clustering maps of hub genes between the control and TSZ groups. (D) Expression of hub genes at different time points following SCI, as assessed by single-cell RNA-sequencing. ceRNA: Competing endogenous RNA; circRNA: circular RNA; lncRNA: long non-coding RNA; miRNA: microRNA; SCI: spinal cord injury; WTS: whole-transcriptome sequencing.
To further clarify the role of the ceRNA, hub genes were identified based on the target genes of ceRNA network, including the up-regulated tuberous sclerosis 2 (Tsc2) gene and the down-regulated solute carrier family 16 member 3 (Slc16a3) and Forkhead box protein P1 (Foxp1) genes. The bidirectional clustering heatmap shown in Figure 3C depicts the differential mRNA expression levels of the hub genes between the control and TSZ groups.
Validation of hub gene expression levels after spinal cord injury
To confirm whether the expression of hub genes post-SCI corresponds to their expression in exosomes, we generated bubble plots of hub gene expression after SCI from the single-cell RNA-sequencing data. Tsc2 expression increased significantly after SCI but decreased over time. In contrast, Slc16a3 and Foxp1 expression levels were highest in the control group and decreased with time after SCI (Figure 3D). The expression trends of the hub genes that were differentially expressed after SCI were generally consistent with the expression patterns seen in the ceRNA networks.
Western blot analysis showed a substantial increase in TSC2 expression in spinal cord tissues from the SCI group compared with the sham group (P = 0.0298). Conversely, FOXP1 and SLC16A3 expression levels did not change significantly (Figure 4A and B). Immunofluorescence analysis of spinal cord tissues showed a marked increase in the number of SOX2-positive cells in the SCI group compared with the sham group, and these cells were concentrated in close proximity to the injury site. We observed a concurrent increase in the number of TSC2-positive cells within the injury zone. These findings suggest that exosomes secreted by endogenous NSCs upregulated TSC2 expression in exosome receptor cells (Figure 4C).
Figure 4.
Western blot and immunofluorescence analysis of the expression of proteins encoded by hub genes in the spinal cord 3 days following injury.
(A, B) Western blot and semi-quantitative analyses of the expression of proteins encoded by hub genes in the spinal cord 3 days after SCI. Data are expressed as mean ± SD (n = 3), and were analyzed by two-tailed unpaired Student’s t-test. (C) Immunofluorescence staining for the NSC marker SOX2 (green, Alexa Fluor 488) and the hub gene product TSC2 (red, Alexa Fluor 594). Compared with the sham group, TSC2 and SOX2 expression levels were up-regulated in the SCI group, and the fluorescence signals from both proteins overlapped. Cell nuclei were counterstained with DAPI. Scale bars: 250 μm (left) and 50 μm (middle and right). DAPI: 4′,6-Diamidino-2-phenylindole; dpi: day post-injury; ns: not significant; NSC: neural stem cell; SCI: spinal cord injury; SOX2: SRY-box transcription factor 2; TSC2: tuberous sclerosis 2.
Validation of changes in TSC2 expression induced by exosomes derived from TSZ-induced neural stem cells in vitro
Next, we performed an in vitro uptake assay in which NSC-34 cells were exposed to exosomes secreted by NSCs undergoing necroptosis (1 × 109 particles/mL, equivalent to adding 196 μL of exosomes to 1 mL of exosome-free medium). We then measured TSC2 expression following direct cellular uptake of exosomes derived from TSZ-treated NSCs. Western blot analysis showed a significant increase in TSC2 expression during the first 9 hours post–exosomes uptake in vitro. Conversely, no notable change in TSC2 expression was observed from 9 to 24 hours after exosome uptake (Figure 5A and B). Furthermore, immunofluorescence analysis showed that the TSC2 fluorescence intensity was more pronounced 12 hours after exosome uptake in cells that had taken up a larger quantity of exosomes (red, 1,1-dioctadecyl-3,3,3,3-tetramethylindodicarbocyanine–labeled) (Figure 5C).
Figure 5.
Western blot and immunofluorescence analyses of TSC2 expression after uptake of exosomes derived from TSZ-treated NSCs in vitro.
(A, B) Western blot and semi-quantitative analyses of TSC2 expression at different time points after uptake of exosomes derived from TSZ-treated NSCs in vitro. Data are expressed as mean ± SD (n = 3), and were analyzed by one-way analysis of variance followed by Bonferroni’s post hoc test. (C) Immunofluorescence staining for TSC2 (green, Alexa Fluor 488) and DID-labeled exosomes derived from TSZ-treated NSCs (red) 12 hours after uptake in vitro. When the NSC-34 cells took up the exosomes, there was a significant increase in TSC2 expression. Cell nuclei were counterstained with DAPI. Scale bars: 50 μm (left) and 20 μm (middle and right). DAPI: 4′,6-Diamidino-2-phenylindole; DID: 1,1-dioctadecyl-3,3,3,3-tetramethylindodicarbocyanine; TSC2: tuberous sclerosis 2; TSZ: tumor necrosis factor α, SMAC mimetic, zVAD-fmk.
Ependymal cell communication involving hub genes after spinal cord injury
Next, we conducted ligand-receptor pair analysis to analyze cellular communication among all cells in the control group and at three time points after SCI (Additional Tables 6 (1.6MB, pdf) –9 (1.7MB, pdf) ). Cellular communication was observed between ependymal cells and all cell types (including other ependymal cells) at all time points (Additional Figure 4 (5.4MB, tif) ). Ependymal cell communication with astrocytes and OPCs was stronger in the control group than in the SCI groups. At 1 day post-injury (dpi), almost all cellular communication decreased to similar levels. At 3 dpi, cellular communication was stronger with astrocytes and microglial cells. At 7 dpi, cellular communication in the SCI groups was similar to that seen in the control group, and again stronger with astrocytes and OPCs. Additionally, we elucidated the receptor-ligand pairs involved in intercellular communication between ependymal cells and all cell types in the control group and at three time points after SCI (Figure 6A).
Figure 6.
Ependymal cell communication in SCI and cellular communication involving hub genes.
(A) Ependymal cell communication with other cell types after SCI. (B) Cellular communication through the TNF signaling pathway after SCI. (C) Interaction network of hub genes and receptor–ligand genes after SCI, constructed using data from the STRING database. (D, E) Cellular communication involving the interaction of hub genes with receptors and ligand genes. The line thickness signifies the strength of the intercellular communication; the line color corresponds to the color of the cell from which the ligand originates. dpi: Day(s) post-injury; SCI: spinal cord injury; TNF: tumor necrosis factor.
On the basis of our previous findings indicating that ependymal cells undergo programmed necroptosis after SCI, mainly through the TNF signaling pathway, we further analyzed cellular communication through this pathway. Ependymal cells in the control and 7 dpi groups did not receive any cellular communication related to the TNF signaling pathway. In the 1 and 3 dpi groups, ependymal cells primarily received cellular communication related to the TNF signaling pathway from microglia, along with additional TNF signaling from neutrophils at 1 dpi. From this, we infer that Tsc2 up-regulation is mainly involved in ependymal cell communication at 1 and 3 dpi, while Slc16a3 and Foxp1 down-regulation was mainly involved in the control and 7-dpi groups (Figure 6B).
To determine whether the hub genes are involved in cellular communication after SCI, we analyzed the interactions among hub genes, ligands, and receptors. Slc16a3 and Foxp1 exhibited textmining (extracting a list of protein interactomes from the literature) and co-expression relationships with the vascular endothelial growth factor A (Vegfa) and interferon gamma (Ifnγ) ligand-encoding genes, respectively, whereas Tsc2 exhibited a textmining relationship with the Tnf, midkine (Mdk), transforming growth factor beta-1 (Tgfβ1), and Tgfβ2 ligand-encoding genes. Regarding receptors, textmining and experimentally determined relationships were found with Slc16a3 and Cd44, whereas Foxp1 did not show a relationship with any receptor. In contrast, Tsc2 showed textmining and experimentally determined relationships with transforming growth factor beta receptor type-2 (Tgfβr2), epidermal growth factor receptor (Egfr) and insulin-like growth factor 1 receptor (Igf1r), textmining and experimentally determined co-expression relationships with Tnfrsf1a, and a textmining relationship with Tlr4 (Figure 6C).
We deduced the involvement of hub genes in ependymal cell communication based on the relationship between hub genes and related ligand-receptor pairs. Specifically, Tsc2 participated in ependymal cell communication with OPCs and fibroblasts cells through the EGF signaling pathway at 1 dpi. At 3 dpi, Tsc2 involvement was limited to cellular between ependymal cells with fibroblast cells. Moreover, Tsc2 played a role in ependymal cell communication with all other cells type through the MDK signaling pathway at both 1 and 3 dpi, with greater communication observed with OPCs, fibroblasts, and astrocytes at 3 dpi compared with 1 dpi (Figure 6D). Furthermore, in the control group Slc16a3 played a role in cellular communication between ventricular ependymal cells and endothelial and pericyte cells through the VEGF signaling pathway. Concurrently, Slc16a3 participated in ventricular ependymal cell communication with dendritic cells and monocytes in both the control group and the 7-dpi group via the Cd74–Cd44 receptor-ligand pair, which is part of the macrophage migration inhibitory factor (MIF) signaling pathway, and was involved with ventricular ependymal cell communication with division-like-myeloid cells at 7 dpi (Figure 6E). Additionally, Foxp1 participated in ventricular ependymal cell communication.
Discussion
Following SCI, NSCs originating from the spinal periventricular zone proliferate, migrate to the injury site, and differentiate into mature neural cells (Vieira et al., 2018). Despite the prevailing consensus that NSCs derive from ependymal cells, which is supported by evidence from lineage tracing experiments (Ripoll et al., 2023), there are still dissenting perspectives (Ren et al., 2017). In this study, we examined the expression of NSC markers through single-cell RNA-sequencing data analysis and observed that Sox2 and Foxj1 were predominantly expressed in ependymal cells. While nestin and vimentin were expressed in other cells, ependymal cells expressed high levels of both proteins. From this, we inferred that NSCs originate from this specific subset of ependymal cells.
In our previous study, in which we used Nestin as a marker for NSCs, we demonstrated that nestin-positive cells undergo necroptosis after SCI and that inhibiting programmed necroptosis promotes recovery from SCI (Tong et al., 2023). In this study, we analyzed genes related to necroptosis using single-cell RNA-sequencing data and RNA-sequencing of ependymal cells extracted after SCI. We found that necroptosis occurred in ependymal cells after SCI and that Tnfrsf1a expression increased significantly after SCI. From this, we inferred that NSCs undergo necroptosis after SCI through the classical TNF signaling pathway.
The classical necroptosis pathway is a form of programmed cell death characterized by caspase-8 inhibition (Rodriguez et al., 2022), wherein the death process involves the release of cellular contents, including damage-associated molecular patterns. Previous studies have regarded necroptosis as being detrimental to SCI recovery, mainly due to the reduction in the number of NSCs and the increase in the inflammatory response (Liu et al., 2018; Hu et al., 2022). However, recent research suggests that necroptosis can alter the mode of exosome secretion and the exosome proteome (Gupta et al., 2022). Additionally, exosomes play a crucial role in cellular communication (Walbrecq et al., 2020; Huber and Wang, 2024; Xu et al., 2024; Yu et al., 2024). These findings indicate that necroptosis in SCI not only may damage cells but also may be involved in exosome-mediated ependymal cell communication. dNSCs are present at E16.5 and can persist into adulthood (Redmond et al., 2019). Because extracting NSCs after SCI yields low numbers of both cells and exosomes, we opted to use the spinal cord of E16 to E17 fetal mice as the source of NSCs. Exosome morphology, particle size distribution, concentration, and surface markers remain unchanged after TSZ induction in vitro.
In this study, we performed comprehensive transcriptome sequencing of purified exosomes. Initially, we conducted distinct analyses for mRNAs and non-coding RNAs, resulting in the identification of 108 DEmRNAs, 104 DElncRNAs, 720 DEcircRNAs, and 14 DEmiRNAs. Notably, the “Ubiquitin-mediated proteolysis” and “Autophagy” signaling pathways were significantly enriched in DEmRNAs, DElncRNAs, and DEcircRNAs. The heightened presence of “Autophagy” may be attributable to the transition from exocytosis to a lysosomal-mediated mechanism induced by necroptosis (Gupta et al., 2022). Furthermore, a recent report highlighted the involvement of ubiquitination in necroptosis (Chen et al., 2022), suggesting that the enriched “Ubiquitin-mediated proteolysis” pathway presents a novel avenue for understanding epigenetic modifications in exosomes. However, none of the mRNA classes studied were enriched in “BF” or “CC” in the GO enrichment analysis, whereas only “binding protein” was enriched in the “MF” analysis. This implies that the exosomal transcriptome might exert its influence through protein-mediated proteolysis and protein binding.
Subsequently, we constructed a ceRNA network involving lncRNA/circRNA-miRNA-mRNA interactions, adhering to the ceRNA principle. Through analysis of target genes within this network, we identified hub genes associated with programmed necroptosis in NSCs, specifically noting up-regulation of Tsc2 and down-regulation of Slc16a3 and Foxp1. Furthermore, we validated the expression trends of hub genes, showing that their up- and down-regulation after SCI aligned with the observed changes in hub gene expression following necroptosis in NSCs.
In our in vivo experiments, we observed a substantial increase in TSC2 in spinal cord tissues after SCI. Conversely, there were no significant alterations in SLC16A3 or FOXP1 after SCI. This result was in contrast to our single-cell RNA-sequencing findings. We hypothesize that this discrepancy may arise from our choice to assess protein expression levels 3 days post-SCI. It is also possible that extracting total protein from spinal cord tissues includes parts beyond the targeted region. Building on the in vivo validation results, we proceeded to confirm in vitro that exosomes influence TSC2 expression because TSC2 expression was increased in cells that had taken up exosomes in vitro. Notably, within the first 9 hours after exosome uptake, TSC2 expression increased in a time-dependent manner. Subsequently, from 9 to 24 hours after uptake, TSC2 expression stabilized, suggesting that the mRNAs carried by the exosomes had undergone transcription and translation. TSC2 expression in recipient cells exposed to exosomes was significantly elevated. Thus, our in vivo and in vitro results show that NSCs undergoing necroptosis can elevate TSC2 expression in recipient cells by releasing exosomes. This underscores the involvement of necroptotic NSCs in communicating with other cells via exosomes. The increased expression of TSC2 in recipient cells represents the most immediate outcome of this cellular communication, while subsequent downstream changes remain to be explored.
TSC2 is a pivotal regulator of various biological processes that binds to TSC1 to form the TSC1–TSC2 complex, which is a central integrator of external stress and serves as a core regulator in the mTORC1 and mTORC2 signaling pathways (Karalis et al., 2024). The TSC1–TSC2 complex plays a crucial role in orchestrating an appropriate stress response by modulating the level of mTOR signaling. By regulating Rheb activity, the TSC1–TSC2 complex inhibits mTORC1 signaling, while also activating mTORC2 signaling through direct interaction. Additionally, it plays a vital role in the crosstalk between mTORC1 and mTORC2 signaling through PI3K/Akt feedback regulation. In this context, Akt directly phosphorylates TSC2, thereby inhibiting TSC2’s inhibitory effect on Rheb and mTORC1. This limitation on the inhibitory function of TSC2 allows for the fine-tuning of mTORC1 signaling and underscores the intricate regulatory network involving the TSC1–TSC2 complex in cellular responses (Lin et al., 2023). The inhibitory effect of TSC2 on mTORC1 can result in a reduction in cellular metabolism and biosynthesis, along with the stimulation of autophagy, ultimately culminating in diminished cell growth (Liu and Sabatini, 2020). Previous studies propose that lysosomal function could represent a crucial upstream cellular event in the regulation of various pro-death pathways during secondary SCI. These pathways include inhibition of autophagic flux, activation of endoplasmic reticulum stress-dependent apoptosis, and sensitization to necrosis. In other central nervous system trauma models, such as models of traumatic brain injury, inhibiting the autophagy/lysosomal pathway and activating necrosis are associated with cell loss and tissue damage. Integrating these findings with our results, it is plausible that exosomes secreted after NSC necroptosis may mitigate further damage to spinal cord tissues. This potential protective effect could be achieved by upregulating TSC2 expression, inhibiting mTORC1 expression, and consequently promoting autophagic flux expression. In our previous studies, we investigated the role of Rictor in the mTORC2 complex following SCI and found that Rictor overexpression contributes to SCI recovery by exerting anti-inflammatory effects, as well as supporting neuronal and oligodendrocyte survival (Chen et al., 2020). Integrating these results with the outcomes of the present study, it can be inferred that exosomes released following NSC necroptosis post-SCI may protect spinal cord tissues. This protection is likely achieved by increasing TSC2/mTORC2 signaling in recipient cells, ultimately helping preserve spinal cord tissues (Chen et al., 2024). Questions regarding the downstream effects of TSC2, including modifications in the mTORC1/2 signaling pathway, will be a primary focus of our subsequent studies. In the current study, we delved deeper into the role of hub genes in SCI using single-cell RNA-sequencing to discern their roles in NSC communication with other neural cell types.
Analysis of ependymal cell communication revealed consistent interactions with all cell types, irrespective of the presence of SCI or the time point after SCI. Interestingly, the overall strength of cellular communication diminished at 1 dpi. Furthermore, we observed that communication between ependymal cells and astrocytes was significantly stronger than that between ependymal cells and other cell types. This finding suggests that NSCs, beyond their role as astrocyte precursors (Yeon et al., 2021), may influence glial cells through complex cellular communication pathways. Additionally, we analyzed the role of TNF signaling in cellular communication following SCI and found that ependymal cells received TNF signals from microglia at 1 and 3 dpi, providing further support for our earlier observations that NSC necroptosis post-SCI is mediated by the classical TNF signaling pathway and shedding light on the origin of TNF signals. Notably, TNF-α, a well-known pro-inflammatory factor, has recently been recognized as being crucial for NSC-mediated spinal cord regeneration (Tsarouchas et al., 2018). This suggests that the NSC response to TNF-α, including necroptosis, could have a positive impact on NSC activation.
We delved further into the relationship between hub genes and the interactions among all receptor ligands, focusing on the cellular communication associated with the up-regulated gene Tsc2 in the 1-dpi and 3-dpi groups, as well as the down-regulated genes Slc16a3 and Foxp1 in the control and 7-dpi groups, in light of reception of TNF signaling from ependymal cells. Our analysis showed that Tsc2 was intricately involved in ependymal cell communication through the EGF and MDK signaling pathways. Similarly, Slc16a3 played a role in ependymal cell communication through the VEGF and MIF signaling pathways. Although Foxp1 was not directly involved in cellular communication, the decreased expression of Foxp1, a motor neuron marker (Chiba et al., 2021), in exosomes after NSC necroptosis suggested potential disruption of NSC differentiation into neurons.
This study had several limitations. First, we were unable to directly extract NSCs after SCI for exosomal transcriptome sequencing. Exosomal sequencing demands a large number of cells, and NSC extraction relies on subsequent expansion and sorting in culture medium. It is challenging to extract NSCs from adult mice after SCI, and they exhibit poor survival in vitro. To overcome this limitation, spinal tissue from fetal mice aged E 16 to E17 were used as a source of NSCs to establish the in vitro necroptosis model. Furthermore, we did not perform a detailed analysis of the ubiquitin-mediated proteolysis pathway, which was enriched in differentially expressed RNAs and could be a promising avenue for future research. Finally, we employed CellChat for the analysis of cellular communication, with a specific emphasis on investigating ligand-receptor pairs within various cell types. A more recent analytical tool, NicheNet, has a greater focus on functional alterations of receptor cells and not only confirms the presence of cellular communication but also predicts the subsequent changes induced by such communication (Browaeys et al., 2020). Using NicheNet in a future study could help predict downstream effects resulting from cellular communication.
In summary, we conducted transcriptome sequencing of exosomes derived from NSCs following necroptosis and used whole-transcriptome sequencing analysis to identify hub genes (Tsc2, Slc16a3, and Foxp1) within the ceRNA network. We validated the expression of the hub genes in spinal tissues from a mouse model of SCI and observed a significant elevation in TSC2 expression post-SCI. Additionally, our findings suggest that NSCs may enhance TSC2 expression in cells at the injury site by secreting exosomes to that specific area. Furthermore, our in vitro results showed a time-dependent increase in TSC2 expression within 9 hours of cells taking up exosomes produced by NSCs treated with TSZ. This supports the notion that exosomes produced by NSCs treated with TSZ can effectively upregulate TSC2 expression in recipient cells. We also found that Tsc2 is involved in ependymal cell communication at 1 and 3 dpi through the EGF and MDK signaling pathways. Furthermore, Slc16a3 was found to be involved in cellular communication in the control group and 7 dpi through the VEGF and MIF signaling pathways. These findings provide novel insight into the role of necroptosis in SCI and shed light on the impact of exosomes in the context of SCI.
Additional files:
Additional Figure 1 (4.3MB, tif) : Differential analysis on mRNAs, LncRNAs, CircRNAs, and miRNAs.
Differential analysis on mRNAs, LncRNAs, CircRNAs, and miRNAs.
(A) Venn diagram analysis of mRNAs, LncRNAs, CircRNAs and miRNAs. (B) Clustering maps of differentially expressed of mRNAs, LncRNAs, CircRNAs and miRNAs. CircRNA: Circular ribonucleic acid; LncRNA: long non-coding RNA; miRNA: microRNA.
Additional Figure 2 (6.4MB, tif) : Differential analysis and KEGG enrichment analysis on mRNAs, LncRNAs, CircRNAs, and miRNAs.
Differential analysis and KEGG enrichment analysis on mRNAs, LncRNAs, CircRNAs, and miRNAs.
(A) Volcano plots show differentially expressed mRNAs, LncRNAs, CircRNAs, and miRNAs in the control and TSZ groups. The criterion of |log2 (fold change)| = 1 and P-value < 0.05 was used in three groups, except miRNAs (|log2 (fold change)| = 0 and P-value < 0.05). (B-E) KEGG analysis of differentially expressed mRNAs, as well as LncRNAs, CircRNAs, and miRNAs (target), in the TSZ group compared to the control group was conducted using KOBAS software. Rich factor represents the ratio of the number of genes located in the entry of the pathway to the total number of annotated genes. CircRNA: Circular ribonucleic acid; KEGG: Kyoto Encyclopedia of Genes and Genomes; LncRNA: long non-coding RNA; miRNA: microRNA; TSZ: tumor necrosis factor α, SMAC mimetic, zVAD-fmk.
Additional Figure 3 (6.3MB, tif) : GO enrichment analysis of DEmRNAs, DELncRNAs, DECircRNAs, and DEmiRNAs.
GO enrichment analysis of DEmRNAs, DELncRNAs, DECircRNAs, and DEmiRNAs.
(A-D) Enrichment analysis for GO was conducted on differentially expressed mRNAs, as well as LncRNAs, CircRNAs, and miRNAs (target), using the “GOseq” R package. The three different classifications correspond to the fundamental categories of GO terms: biological process (BP, red), cellular component (CC, blue), and molecular function (MF, green). CircRNA: circular ribonucleic acid; GO: Gene Ontology; LncRNA: long non-coding RNA; miRNA: microRNA.
Additional Figure 4 (5.4MB, tif) : Receptor-ligand pairs for intercellular communication between ependymal cells and other cells at different time points after SCI.
Receptor-ligand pairs for intercellular communication between ependymal cells and other cells at different time points after SCI.
The size of the circles represents the magnitude of the P-values, and the color depth represents the strength of communication. SCI: Spinal cord injury.
Additional Table 1: PCD-related gene sets in GSEA.
Additional Table 2: Genes tagged in necroptosis gene set.
Additional Table 3: The R packages and Uniform Resource Locators (URLs) used in this study.
Additional Table 4: Whole transcriptome sequencing network of LncRNA-miRNA-mRNA.
Additional Table 5 (186.4KB, pdf) : Whole transcriptome sequencing network of CircRNA-miRNA-mRNA.
Whole transcriptome sequencing network ofCircRNA-miRNA-mRNA
Additional Table 6 (1.6MB, pdf) : The communication signal pathways and related ligand receptors in the control group.
The communication signal pathways and related ligand receptors in the control group
Additional Table 7 (2.4MB, pdf) : The communication signal pathways and related ligand receptors in the 1-day post-spinal cord injury group.
The communication signal pathways and related ligand receptors in the 1-day post-spinal cord injury group
Additional Table 8: (1.8MB, pdf) The communication signal pathways and related ligand receptors in the 3-day post-spinal cord injury group.
The communication signal pathways and related ligand receptors in the 3-day post-spinal cord injury group
Additional Table 9: (1.7MB, pdf) The communication signal pathways and related ligand receptors in the 7-day post-spinal cord injury group.
The communication signal pathways and related ligand receptors in the 7-day post-spinal cord injury group
Additional file 1 (177.7KB, pdf) : Methods for whole transcriptome analysis of exosomes.
Methods for whole transcriptome analysis of exosomes
Additional file 2: Open peer review report 1 (82.1KB, pdf) .
Funding Statement
Funding: This study was supported by the National Natural Science Foundation of China, No. 81801907 (to NC); Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, No. ZDSYS20230626091402006 (to NC); Sanming Project of Medicine in Shenzhen, No. SZSM201911002 (to SL); Foundation of Shenzhen Committee for Science and Technology Innovation, Nos. JCYJ20230807110310021 (to NC), JCYJ20230807110259002 (to JL); and Science and Technology Program of Guangzhou, No. 2024A04J4716 (to TL).
Footnotes
Conflicts of interest: The authors declare that they have no competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Author statement: A segment of the findings from this study has been previously released as a pre-printed version on Research Square, accessible via the relevant link (https://www.researchsquare.com/article/rs-3673053/v1). The current study extends upon the preprint version by incorporating fundamental experiments for in vivo and in vitro correlation validation. It encompasses additional details on the methods employed for the basic experiments, as well as the inclusion of Figures 4 and 5 in the results section. Furthermore, this iteration incorporates an expanded discussion section to provide a comprehensive analysis of the obtained results.
Open peer reviewer: Sujeong Jang, Chonnam National University, Korea.
P-Reviewer: Jang S; C-Editor: Zhao M; S-Editors: Yu J, Li CH; L-Editors: Crow E, Yu J, Song LP; T-Editor: Jia Y
Data availability statement:
part of datasets analyzed in this study are available in the GEO database, including the datasets GSE149669 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE149669), and GSE162610 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE162610). The data on the exosome transcriptome came from the researcher’s sequencing data. The data generated or analysed during this study are available from the corresponding author on reasonable request.
References
- Aran D, Looney AP, Liu L, Wu E, Fong V, Hsu A, Chak S, Naikawadi RP, Wolters PJ, Abate AR, Butte AJ, Bhattacharya M. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat Immunol. 2019;20:163–172. doi: 10.1038/s41590-018-0276-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Browaeys R, Saelens W, Saeys Y. NicheNet: modeling intercellular communication by linking ligands to target genes. Nat Methods. 2020;17:159–162. doi: 10.1038/s41592-019-0667-5. [DOI] [PubMed] [Google Scholar]
- Ceto S, Sekiguchi KJ, Takashima Y, Nimmerjahn A, Tuszynski MH. Neural stem cell grafts form extensive synaptic networks that integrate with host circuits after spinal cord injury. Cell Stem Cell. 2020;27:430–440.e5. doi: 10.1016/j.stem.2020.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen G, Tong K, Li S, Huang Z, Liu S, Zhu H, Zhong Y, Zhou Z, Jiao G, Wei F, Chen N. Extracellular vesicles released by transforming growth factor-beta 1-preconditional mesenchymal stem cells promote recovery in mice with spinal cord injury. Bioact Mater. 2024;35:135–149. doi: 10.1016/j.bioactmat.2024.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen N, Zhou P, Liu X, Li J, Wan Y, Liu S, Wei F. Overexpression of rictor in the injured spinal cord promotes functional recovery in a rat model of spinal cord injury. FASEB J. 2020;34:6984–6998. doi: 10.1096/fj.201903171R. [DOI] [PubMed] [Google Scholar]
- Chen Y, Ren W, Wang Q, He Y, Ma D, Cai Z. The regulation of necroptosis by ubiquitylation. Apoptosis. 2022;27:668–684. doi: 10.1007/s10495-022-01755-8. [DOI] [PubMed] [Google Scholar]
- Chiba A, Soma K, Watanabe K, Nagashima H, Sato N. Development of fin-innervating motor neurons after peripheral target removal in medaka fish. Dev Neurobiol. 2021;81:110–122. doi: 10.1002/dneu.22799. [DOI] [PubMed] [Google Scholar]
- Fan B, Wei Z, Yao X, Shi G, Cheng X, Zhou X, Zhou H, Ning G, Kong X, Feng S. Microenvironment imbalance of spinal cord injury. Cell Transplant. 2018;27:853–866. doi: 10.1177/0963689718755778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fan J, Shi S, Qiu Y, Liu M, Shu Q. Analysis of signature genes and association with immune cells infiltration in pediatric septic shock. Front Immunol. 2022;13:1056750. doi: 10.3389/fimmu.2022.1056750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fauser M, Loewenbrück KF, Rangnick J, Brandt MD, Hermann A, Storch A. Adult neural stem cells from midbrain periventricular regions show limited neurogenic potential after transplantation into the hippocampal neurogenic niche. Cells. 2021;10:3021. doi: 10.3390/cells10113021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fritsch M, Günther SD, Schwarzer R, Albert MC, Schorn F, Werthenbach JP, Schiffmann LM, Stair N, Stocks H, Seeger JM, Lamkanfi M, Krönke M, Pasparakis M, Kashkar H. Caspase-8 is the molecular switch for apoptosis, necroptosis and pyroptosis. Nature. 2019;575:683–687. doi: 10.1038/s41586-019-1770-6. [DOI] [PubMed] [Google Scholar]
- Furube E, Ishii H, Nambu Y, Kurganov E, Nagaoka S, Morita M, Miyata S. Neural stem cell phenotype of tanycyte-like ependymal cells in the circumventricular organs and central canal of adult mouse brain. Sci Rep. 2020;10:2826. doi: 10.1038/s41598-020-59629-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gilbert EAB, Lakshman N, Lau KSK, Morshead CM. Regulating endogenous neural stem cell activation to promote spinal cord injury repair. Cells. 2022;11:846. doi: 10.3390/cells11050846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gupta K, Brown KA, Hsieh ML, Hoover BM, Wang J, Khoury MK, Pilli VSS, Beyer RSH, Voruganti NR, Chaudhary S, Roberts DS, Murphy RM, Hong S, Ge Y, Liu B. Necroptosis is associated with Rab27-independent expulsion of extracellular vesicles containing RIPK3 and MLKL. J Extracell Vesicles. 2022;11:e12261. doi: 10.1002/jev2.12261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harmati M, Gyukity-Sebestyen E, Dobra G, Janovak L, Dekany I, Saydam O, Hunyadi-Gulyas E, Nagy I, Farkas A, Pankotai T, Ujfaludi Z, Horvath P, Piccinini F, Kovacs M, Biro T, Buzas K. Small extracellular vesicles convey the stress-induced adaptive responses of melanoma cells. Sci Rep. 2019;9:15329. doi: 10.1038/s41598-019-51778-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hellenbrand DJ, Quinn CM, Piper ZJ, Elder RT, Mishra RR, Marti TL, Omuro PM, Roddick RM, Lee JS, Murphy WL, Hanna AS. The secondary injury cascade after spinal cord injury: an analysis of local cytokine/chemokine regulation. Neural Regen Res. 2024;19:1308–1317. doi: 10.4103/1673-5374.385849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu X, Xu Y, Zhang H, Li Y, Wang X, Xu C, Ni W, Zhou K. Role of necroptosis in traumatic brain and spinal cord injuries. J Adv Res. 2022;40:125–134. doi: 10.1016/j.jare.2021.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huber CC, Wang H. Pathogenic and therapeutic role of exosomes in neurodegenerative disorders. Neural Regen Res. 2024;19:75–79. doi: 10.4103/1673-5374.375320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karalis V, Wood D, Teaney NA, Sahin M. The role of TSC1 and TSC2 proteins in neuronal axons. Mol Psychiatry. 2024 doi: 10.1038/s41380-023-02402-7. doi: 10.1038/s41380-023-02402-7. [DOI] [PubMed] [Google Scholar]
- Khosla R, Bhagat H, Lal P, Anand A. ALS plasma reduces the viability of NSC34 cells via altering mRNA expression of VEGF: A short report. Heliyon. 2023;9:e18287. doi: 10.1016/j.heliyon.2023.e18287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li L, Li L, Sun Q. High expression of cuproptosis-related SLC31A1 gene in relation to unfavorable outcome and deregulated immune cell infiltration in breast cancer: an analysis based on public databases. BMC Bioinformatics. 2022;23:350. doi: 10.1186/s12859-022-04894-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin CP, Traets JJH, Vredevoogd DW, Visser NL, Peeper DS. TSC2 regulates tumor susceptibility to TRAIL-mediated T-cell killing by orchestrating mTOR signaling. EMBO J. 2023;42:e111614. doi: 10.15252/embj.2022111614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu GY, Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol. 2020;21:183–203. doi: 10.1038/s41580-019-0199-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu S, Li Y, Choi HMC, Sarkar C, Koh EY, Wu J, Lipinski MM. Lysosomal damage after spinal cord injury causes accumulation of RIPK1 and RIPK3 proteins and potentiation of necroptosis. Cell Death Dis. 2018;9:476. doi: 10.1038/s41419-018-0469-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu WZ, Ma ZJ, Li JR, Kang XW. Mesenchymal stem cell-derived exosomes: therapeutic opportunities and challenges for spinal cord injury. Stem Cell Res Ther. 2021;12:102. doi: 10.1186/s13287-021-02153-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marqués-Torrejón M, Williams CAC, Southgate B, Alfazema N, Clements MP, Garcia-Diaz C, Blin C, Arranz-Emparan N, Fraser J, Gammoh N, Parrinello S, Pollard SM. LRIG1 is a gatekeeper to exit from quiescence in adult neural stem cells. Nat Commun. 2021;12:2594. doi: 10.1038/s41467-021-22813-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Milich LM, Choi JS, Ryan C, Cerqueira SR, Benavides S, Yahn SL, Tsoulfas P, Lee JK. Single-cell analysis of the cellular heterogeneity and interactions in the injured mouse spinal cord. J Exp Med. 2021;218:e20210040. doi: 10.1084/jem.20210040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morgan MJ, Kim YS. Roles of RIPK3 in necroptosis, cell signaling, and disease. Exp Mol Med. 2022;54:1695–1704. doi: 10.1038/s12276-022-00868-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morrow CS, Porter TJ, Xu N, Arndt ZP, Ako-Asare K, Heo HJ, Thompson EAN, Moore DL. Vimentin coordinates protein turnover at the aggresome during neural stem cell quiescence exit. Cell Stem Cell. 2020;26:558–568.e9. doi: 10.1016/j.stem.2020.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Redmond SA, Figueres-Oñate M, Obernier K, Nascimento MA, Parraguez JI, López-Mascaraque L, Fuentealba LC, Alvarez-Buylla A. Development of ependymal and postnatal neural stem cells and their origin from a common embryonic progenitor. Cell Rep. 2019;27:429–441.e3. doi: 10.1016/j.celrep.2019.01.088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ren Y, Ao Y, O’Shea TM, Burda JE, Bernstein AM, Brumm AJ, Muthusamy N, Ghashghaei HT, Carmichael ST, Cheng L, Sofroniew MV. Ependymal cell contribution to scar formation after spinal cord injury is minimal, local and dependent on direct ependymal injury. Sci Rep. 2017;7:41122. doi: 10.1038/srep41122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ripoll C, Poulen G, Chevreau R, Lonjon N, Vachiery-Lahaye F, Bauchet L, Hugnot JP. Persistence of FoxJ1(+) Pax6(+) Sox2(+) ependymal cells throughout life in the human spinal cord. Cell Mol Life Sci. 2023;80:181. doi: 10.1007/s00018-023-04811-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodriguez DA, Quarato G, Liedmann S, Tummers B, Zhang T, Guy C, Crawford JC, Palacios G, Pelletier S, Kalkavan H, Shaw JJP, Fitzgerald P, Chen MJ, Balachandran S, Green DR. Caspase-8 and FADD prevent spontaneous ZBP1 expression and necroptosis. Proc Natl Acad Sci U S A. 2022;119:e2207240119. doi: 10.1073/pnas.2207240119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosenberg AB, Roco CM, Muscat RA, Kuchina A, Sample P, Yao Z, Graybuck LT, Peeler DJ, Mukherjee S, Chen W, Pun SH, Sellers DL, Tasic B, Seelig G. Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding. Science. 2018;360:176–182. doi: 10.1126/science.aam8999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sathyamurthy A, Johnson KR, Matson KJE, Dobrott CI, Li L, Ryba AR, Bergman TB, Kelly MC, Kelley MW, Levine AJ. Massively parallel single nucleus transcriptional profiling defines spinal cord neurons and their activity during behavior. Cell Rep. 2018;22:2216–2225. doi: 10.1016/j.celrep.2018.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schneider CA, Rasband WS, Eliceiri KW. NIH image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9:671–675. doi: 10.1038/nmeth.2089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shu M, Xue X, Nie H, Wu X, Sun M, Qiao L, Li X, Xu B, Xiao Z, Zhao Y, Fan Y, Chen B, Zhang J, Shi Y, Yang Y, Lu F, Dai J. Single-cell RNA sequencing reveals Nestin(+) active neural stem cells outside the central canal after spinal cord injury. Sci China Life Sci. 2022;65:295–308. doi: 10.1007/s11427-020-1930-0. [DOI] [PubMed] [Google Scholar]
- Szklarczyk D, Kirsch R, Koutrouli M, Nastou K, Mehryary F, Hachilif R, Gable AL, Fang T, Doncheva NT, Pyysalo S, Bork P, Jensen LJ, von Mering C. The STRING database in 2023: protein-protein association networks and functional enrichment analyses for any sequenced genome of interest. Nucleic Acids Res. 2023;51:D638–D646. doi: 10.1093/nar/gkac1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tong K, Li S, Chen G, Ma C, Liu X, Liu S, Chen N. Inhibition of neural stem cell necroptosis mediated by RIPK1/MLKL promotes functional recovery after SCI. Mol Neurobiol. 2023;60:2135–2149. doi: 10.1007/s12035-022-03156-z. [DOI] [PubMed] [Google Scholar]
- Tsarouchas TM, Wehner D, Cavone L, Munir T, Keatinge M, Lambertus M, Underhill A, Barrett T, Kassapis E, Ogryzko N, Feng Y, van Ham TJ, Becker T, Becker CG. Dynamic control of proinflammatory cytokines Il-1β and Tnf-α by macrophages in zebrafish spinal cord regeneration. Nat Commun. 2018;9:4670. doi: 10.1038/s41467-018-07036-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ulintz PJ, Wu W, Gates CM. Bioinformatics analysis of whole exome sequencing data. Methods Mol Biol. 2019;1881:277–318. doi: 10.1007/978-1-4939-8876-1_21. [DOI] [PubMed] [Google Scholar]
- Vieira MS, Santos AK, Vasconcellos R, Goulart VAM, Parreira RC, Kihara AH, Ulrich H, Resende RR. Neural stem cell differentiation into mature neurons: mechanisms of regulation and biotechnological applications. Biotechnol Adv. 2018;36:1946–1970. doi: 10.1016/j.biotechadv.2018.08.002. [DOI] [PubMed] [Google Scholar]
- Walbrecq G, Margue C, Behrmann I, Kreis S. Distinct cargos of small extracellular vesicles derived from hypoxic cells and their effect on cancer cells. Int J Mol Sci. 2020;21:5071. doi: 10.3390/ijms21145071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Watanabe F, Schoeffler A, Fair SR, Hester ME, Fedorko J, Imitola J. Generation of neurosphere-derived organoid-like-aggregates (NEDAS) from neural stem cells. Curr Protoc. 2021;1:e15. doi: 10.1002/cpz1.15. [DOI] [PubMed] [Google Scholar]
- Welsh JA, et al. Minimal information for studies of extracellular vesicles (MISEV2023): from basic to advanced approaches. J Extracell Vesicles. 2024;13:e12404. doi: 10.1002/jev2.12404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wiklander OP, Nordin JZ, O’Loughlin A, Gustafsson Y, Corso G, Mäger I, Vader P, Lee Y, Sork H, Seow Y, Heldring N, Alvarez-Erviti L, Smith CI, Le Blanc K, Macchiarini P, Jungebluth P, Wood MJ, Andaloussi SE. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J Extracell Vesicles. 2015;4:26316. doi: 10.3402/jev.v4.26316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu H, Tong K, Liu X, Li J, Li X, Gao M, Tian W, Chen D, Zhou Z, Liu S. A comparison between two laminectomy procedures in mouse spinal cord injury on Allen’s animal model. J Neurosci Methods. 2022;368:109461. doi: 10.1016/j.jneumeth.2021.109461. [DOI] [PubMed] [Google Scholar]
- Xu H, Li H, Zhang P, Gao Y, Ma H, Gao T, Liu H, Hua W, Zhang L, Zhang X, Yang P, Liu J. The functions of exosomes targeting astrocytes and astrocyte-derived exosomes targeting other cell types. Neural Regen Res. 2024;19:1947–1953. doi: 10.4103/1673-5374.390961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yan WT, Zhao WJ, Hu XM, Ban XX, Ning WY, Wan H, Zhang Q, Xiong K. PANoptosis-like cell death in ischemia/reperfusion injury of retinal neurons. Neural Regen Res. 2023;18:357–363. doi: 10.4103/1673-5374.346545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang AC, et al. A human brain vascular atlas reveals diverse mediators of Alzheimer’s risk. Nature. 2022;603:885–892. doi: 10.1038/s41586-021-04369-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye K, Chen Z, Xu Y. The double-edged functions of necroptosis. Cell Death Dis. 2023;14:163. doi: 10.1038/s41419-023-05691-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yeon GB, Shin WH, Yoo SH, Kim D, Jeon BM, Park WU, Bae Y, Park JY, You S, Na D, Kim DS. NFIB induces functional astrocytes from human pluripotent stem cell-derived neural precursor cells mimicking in vivo astrogliogenesis. J Cell Physiol. 2021;236:7625–7641. doi: 10.1002/jcp.30405. [DOI] [PubMed] [Google Scholar]
- Yu Z, Teng Y, Yang J, Yang L. The role of exosomes in adult neurogenesis: implications for neurodegenerative diseases. Neural Regen Res. 2024;19:282–288. doi: 10.4103/1673-5374.379036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yuan J, Amin P, Ofengeim D. Necroptosis and RIPK1-mediated neuroinflammation in CNS diseases. Nat Rev Neurosci. 2019;20:19–33. doi: 10.1038/s41583-018-0093-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhan C, Huang M, Yang X, Hou J. MLKL: Functions beyond serving as the Executioner of Necroptosis. Theranostics. 2021;11:4759–4769. doi: 10.7150/thno.54072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang J, Liu R, Xu A. Whole transcriptome sequencing analysis of blood plasma-derived exosomes from immune-related hearing loss. Int Immunopharmacol. 2023;120:110361. doi: 10.1016/j.intimp.2023.110361. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Differential analysis on mRNAs, LncRNAs, CircRNAs, and miRNAs.
(A) Venn diagram analysis of mRNAs, LncRNAs, CircRNAs and miRNAs. (B) Clustering maps of differentially expressed of mRNAs, LncRNAs, CircRNAs and miRNAs. CircRNA: Circular ribonucleic acid; LncRNA: long non-coding RNA; miRNA: microRNA.
Differential analysis and KEGG enrichment analysis on mRNAs, LncRNAs, CircRNAs, and miRNAs.
(A) Volcano plots show differentially expressed mRNAs, LncRNAs, CircRNAs, and miRNAs in the control and TSZ groups. The criterion of |log2 (fold change)| = 1 and P-value < 0.05 was used in three groups, except miRNAs (|log2 (fold change)| = 0 and P-value < 0.05). (B-E) KEGG analysis of differentially expressed mRNAs, as well as LncRNAs, CircRNAs, and miRNAs (target), in the TSZ group compared to the control group was conducted using KOBAS software. Rich factor represents the ratio of the number of genes located in the entry of the pathway to the total number of annotated genes. CircRNA: Circular ribonucleic acid; KEGG: Kyoto Encyclopedia of Genes and Genomes; LncRNA: long non-coding RNA; miRNA: microRNA; TSZ: tumor necrosis factor α, SMAC mimetic, zVAD-fmk.
GO enrichment analysis of DEmRNAs, DELncRNAs, DECircRNAs, and DEmiRNAs.
(A-D) Enrichment analysis for GO was conducted on differentially expressed mRNAs, as well as LncRNAs, CircRNAs, and miRNAs (target), using the “GOseq” R package. The three different classifications correspond to the fundamental categories of GO terms: biological process (BP, red), cellular component (CC, blue), and molecular function (MF, green). CircRNA: circular ribonucleic acid; GO: Gene Ontology; LncRNA: long non-coding RNA; miRNA: microRNA.
Receptor-ligand pairs for intercellular communication between ependymal cells and other cells at different time points after SCI.
The size of the circles represents the magnitude of the P-values, and the color depth represents the strength of communication. SCI: Spinal cord injury.
Whole transcriptome sequencing network ofCircRNA-miRNA-mRNA
The communication signal pathways and related ligand receptors in the control group
The communication signal pathways and related ligand receptors in the 1-day post-spinal cord injury group
The communication signal pathways and related ligand receptors in the 3-day post-spinal cord injury group
The communication signal pathways and related ligand receptors in the 7-day post-spinal cord injury group
Methods for whole transcriptome analysis of exosomes
Data Availability Statement
part of datasets analyzed in this study are available in the GEO database, including the datasets GSE149669 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE149669), and GSE162610 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE162610). The data on the exosome transcriptome came from the researcher’s sequencing data. The data generated or analysed during this study are available from the corresponding author on reasonable request.