Open Access

Identification of key genes and pathways associated with spinal cord injury

  • Authors:
    • Yue‑Hui Zhang
    • Jia Song
    • Li‑Gang Wang
    • Jiang Shao
  • View Affiliations

  • Published online on: February 10, 2017     https://doi.org/10.3892/mmr.2017.6192
  • Pages: 1577-1584
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study was designed to identify key genes or significant signaling pathways associated with spinal cord injury (SCI), and to clarify the underlying molecular mechanisms of SCI. Data from the GSE45550 array were downloaded from the Gene Expression Omnibus database. A total of 6 control samples, 6 samples at 3 days post‑SCI (SCI3d), 6 samples at 8 days post‑SCI (SCI8d) and 6 samples at 14 days post‑SCI (SCI14d) were included. The microarray data was preprocessed by the robust multi‑array average algorithm. The differentially expressed genes (DEGs) were identified using the limma package. The overlapping DEGs among groups were analyzed using the Venny 2.0 online tool. Modules enriched by DEGs were selected by weighted gene co‑expression network analysis. Gene Ontology annotation and the Kyoto Encyclopedia of Genes and Genomes pathways were identified for DEGs using the Database for Annotation, Visualization and Integrated Discovery. A total of 693 genes were obtained by combining the DEGs of the SCI3d, SCI8d and SCI14d groups. The pink module and green module with smaller P‑values obtained from weighted gene co‑expression network analysis module analyses of DEGs demonstrated a higher correlation with SCI. In addition, the peroxisome proliferator‑activated receptor (PPAR) signaling pathway that the cluster of differentiation 36 (CD36) was significantly enriched in, was one of the significant pathways in the pink module. The p53 signaling pathway that Caspase‑3 (Casp3) was significantly enriched in was one of the significant pathways in the green module. In conclusion, the results of the present study demonstrated that the PPAR and p53 signaling pathway may serve important roles in the progression of SCI. In addition, CD36 and Casp3 may be involved in the progression of SCI via the PPAR and p53 signaling pathways, respectively.

Introduction

Spinal cord injury (SCI) is an injury to the spinal cord caused by trauma or disease, which may lead to alterations to the normal motor, sensory or autonomic function of the spinal cord (1). SCI is associated with high morbidity and mortality rates, and the SCI annual incidence rate ranges between 12.1–57.8 cases/million individuals (1,2). In addition, the epidemiology of SCI is variable in different countries, and there is currently no effective treatment (3,4). Therefore, an effective therapy for the treatment of SCI is required, and it has been suggested that genes associated with SCI may be able to provide novel strategies for such a treatment.

Despite the lack of effective treatments, there have been some notable findings associated with the molecular mechanism of SCI. It has been observed that SCI results in secondary degeneration involving apoptosis, with an increased expression of genes associated with apoptosis and decreased expression of anti-apoptotic genes (5). In addition, a reduction in excessive M1 macrophage polarization and an enhancement of M2 macrophage polarization produced by regulating the levels of cytokines, including tumor necrosis factor a and interleukin (IL)-1β in the SCI microenvironment, may be a desirable treatment method (6). Tachibana et al (7) observed that 3 genes, including heat shock 27 kDa protein, tissue inhibitor of metalloproteinase-1 and epidermal fatty acid-binding protein, were upregulated in SCI. A recent study indicated that deletion of the IL-1α gene protected oligodendrocytes from SCI by overexpressing TOX high mobility group box family member 3 (8). Furthermore, numerous genes associated with inflammation, such as Arginase 1, are differentially expressed in the ephrin type A-receptor 4 knockout mouse model of SCI (9). In addition, it has been demonstrated that a temporal blockade of the IL-6 signaling pathway may modify the inflammatory response following SCI, and thus promote regeneration of the spinal cord (10). However, there are various additional important genes and pathways associated with SCI that have yet to be explored completely. Thus, a greater understanding of these genes and pathways is required as they may provide novel targets for SCI therapy.

In the present study, GSE45550 microarray data was obtained from the Gene Expression Omnibus (GEO) and used to identify the differentially expressed genes (DEGs) associated with SCI. Functional enrichment analyses were performed for DEGs. Furthermore, functions of gene modules were analyzed. The aim of the present study was to identify critical genes or significant signaling pathways associated with SCI, and clarify the underlying molecular mechanisms involved.

Materials and methods

Affymetrix microarray data

The microarray data from GSE45550 was downloaded from the GEO database (http://www.ncbi.nlm.nih.gov/geo/) (11). The following 4 groups were applied: 6 control samples, 6 samples at 3 days post-SCI (SCI3d), 6 samples at 8 days post-SCI (SCI8d) and 6 samples at 14 days post-SCI (SCI14d). Data from the GPL1355 platform [(Rat230_2) Affymetrix Rat Genome 230 2.0 Array; Affymetrix Inc., Santa Clara, CA, USA] were used for subsequent analysis.

Data preprocessing

The microarray data was preprocessed using the robust multi-array average algorithm with the Affy package (12) in Bioconductor (version 1.46.1; http://www.bioconductor.org/). Background correction, normalization and calculation of expression were all included in the process of preprocessing. The probe of the microarray data was transformed to gene symbols with Bioconductor AnnotationData software packages. If several probes were mapped to one gene symbol, then the mean value was set as the final expression value of this gene. A total of 18,634 gene expression matrixes were obtained from the above process.

DEGs analysis

The DEGs in the following three comparison groups: SCI3d vs. Control, SCI8d vs. Control and SCI14d vs. Control were analyzed using the limma package (13) in Bioconductor. The DEG P-values were calculated using the unpaired Student's t-test (14) provided by the limma package, and the P-values were adjusted to false discovery rate (FDR) values using the Benjamini-Hochberg correction (15). Log2 fold-change (FC) ≥1 and FDR values <0.05 were used as cut-off criterion for DEGs. Hierarchical clustering analysis of the DEGs was then performed and visualized using g-plots (16) in the R package.

Venn diagram analysis of DEGs

Venny is an interactive tool used to compare lists with Venn diagrams (17). The Kyoto Encyclopedia of Genes and Genomes (KEGG; www.genome.jp/kegg/) database is used to put associated gene sets into their respective pathway (18). The Database for Annotation, Visualization and Integrated Discovery (DAVID; http://david.ncifcrf.gov), used for analyzing gene lists, is an integrated data-mining environment (19).

The intersections of upregulated and downregulated genes in different sample groups were respectively analyzed using Venny 2.0 (17) (http://bioinfogp.cnb.csic.es/tools/venny/index.html) online tool. KEGG pathway enrichment analysis was performed for the intersection of genes by DAVID. P≤0.05 and gene counts ≥2 were used as threshold values.

Analysis of the correlation between gene modules and phenotype

Weighted gene co-expression network analysis (WGCNA) (20) is a tool used to identify gene clusters or modules which are highly associated with the phenotype of samples in expression profile data, and generalize module characteristic genes among these gene clusters. Furthermore, WGCNA provides correlation coefficients and significant thresholds in every module.

In the present study DEGs in the SCI3d, SCI8d and SCI14d groups were combined, and the correlation between these DEGs and SCI3d, SCI8d and SCI14d were analyzed, and gene sets with higher correlation were dug. Modules enriched by DEGs were selected by WGCNA in the R package, and modules significantly associated with SCI were identified with cluster analysis. The higher the absolute value of the correlation coefficient, the closer the correlation was between gene expression levels in modules and SCI.

Enrichment analysis of module function

Gene Ontology (GO) is a tool used to generate gene annotations by collecting defined, structured and controlled vocabulary (21). GO annotation and KEGG pathway analyses were performed for DEGs using DAVID. P<0.05 and gene counts ≥2 were set as threshold values.

Results

Normalized analysis of sample data

The boxplots of sample data prior to and following normalization are depicted in Fig. 1. The median line of the boxplot was at the same level following normalization, indicating that all data were successfully normalized.

DEG analysis

The DEG count of the three SCI groups compared with the control group are summarized in Table I with |log2FC| values ≥1 and FDR values <0.05. The heat maps of the DEGs are depicted in Fig. 2.

Table I.

Differentially expressed genes count relative to the control group.

Table I.

Differentially expressed genes count relative to the control group.

GroupUpregulated genes countDownregulated genes countTotal
SCI3d232322554
SCI8d121142263
SCI14d  64  71135

[i] SCI3d, samples taken at 3 days post-SCI; SCI8d, samples taken at 8 days post-SCI; SCI14d, samples taken at 14 days post-SCI.

Analysis of overlapping DEGs among groups

Venn diagram analyses for the upregulated and downregulated genes in the SCI3d, SCI8d and SCI14d groups were performed, and overlapping DEGs among groups is depicted in Fig. 3. A total of 9 genes were upregulated at all 3 time points and 48 genes were downregulated at all 3 time points.

Where P<0.05, there was no significant enrichment observed among the 9 intersecting upregulated genes using KEGG pathway analysis. However, the 48 intersecting downregulated genes were markedly enriched in the peroxisome proliferator-activated receptor (PPAR) signaling pathway (P=8.01×10−4; enriched genes including lipoprotein lipase, fatty acid binding protein 3, aquaporin 7 and adiponectin, C1Q and collagen domain containing) and in the synthesis and degradation of ketone bodies signaling pathway (P=2.39×10−2; enriched genes including 3-oxoacid CoA transferase 1, 3-hydroxybutyrate dehydrogenase, type 1) (data not shown).

WGCNA module analyses of DEGs

A total of 693 genes were obtained by combining the DEGs of the SCI3d, SCI8d and SCI14d groups. Cluster analyses using WGCNA were performed using the expression data of these genes, and the cluster dendrogram is presented in Fig. 4. The DEGs were divided into 7 modules (pink module, green/yellow module, black module, blue module, green module, red module and tan module). The correlation coefficient and P-value between gene counts of every module and SCI are summarized in Table II. The data indicated that the pink module and green module with smaller P-values demonstrated a higher correlation with SCI. The functional enrichment results of pink and green modules are summarized in Table III. It was demonstrated that the PPAR signaling pathway that cluster of differentiation 36 (CD36) was significantly enriched in, was one of the significant pathways in the pink module. In addition, the p53 signaling pathway, that caspase-3 (Casp3) was significantly enriched in, was one of the significant pathways in the green module (Table III).

Table II.

Results of module analysis.

Table II.

Results of module analysis.

FactorPink moduleGreen/yellow moduleBlack moduleBlue moduleGreen moduleRed moduleTan module
Gene count3753125273694690
Correlation−7.30E-01−1.00E-01−5.80E-01−9.00E-027.50E-01−7.80E-022.80E-01
P-value   5.29E-05   6.39E-01   3.08E-03   6.77E-012.28E-05   7.18E-011.88E-01

[i] Seven modules (pink, green/yellow, black, blue, green, red and tan) represent modules obtained from WGCNA module analyses of DEGs. One color represents one module. The degree of co-expression for the genes in the same modules was high. WGCNA, Weighted gene co-expression network analysis.

Table III.

Functional enrichment results of pink and green modules.

Table III.

Functional enrichment results of pink and green modules.

A, Pink module

GO/KEGGTermDescriptionP-valueGene
GO-BPGO:0006631Fatty acid metabolic process8.00E-04Cd36, Mlycd, Acot2, Fabp4, Decr1
GO:0006936Muscle contraction9.00E-04Tnnt3, Tnnc2, Mylpf, Actn3
GO:0003012Muscle system process1.30E-03Tnnt3, Tnnc2, Mylpf, Actn3
GO:0034440Lipid oxidation3.70E-03Cd36, Mlycd, Decr1
GO:0019395Fatty acid oxidation3.70E-03Cd36, Mlycd, Decr1
GO-CCGO:0015629Actin cytoskeleton9.80E-03Tnnt3, Tnnc2, Mylpf, Myoz1
GO:0005861Troponin complex1.70E-02Tnnt3, Tnnc2
GO:0030016Myofibril1.83 E-02Tnnt3, Tnnc2, Mylpf
GO:0005865Striated muscle thin filament1.91E-02Tnnt3, Tnnc2
GO:0043292Contractile fiber2.12E-02Tnnt3, Tnnc2, Mylpf
GO-MFGO:0048037Cofactor binding1.70E-03Ldhb, Nox1, Actn3, Decr1, Nqo1
GO:0008092Cytoskeletal protein binding4.30E-03Tnnt3, Tnnc2, Mylpf, Myoz1, Actn3
GO:0050662Coenzyme binding6.80E-03Ldhb, Nox1, Decr1, Nqo1
GO:0003779Actin binding7.50E-03Tnnt3, Tnnc2, Mylpf, Actn3
KEGGrno04510Focal adhesion1.67E-02Col6A3, Mylpf, Actn3, Col5A3
rno03320PPAR signaling pathway1.70E-02Cd36, Fabp4, Aqp7
rno04512ECM-receptor interaction2.18 E-02Cd36, Col6A3, Col5A3
rno04670Leukocyte transendothelial migration4.17E-02Nox1, Mylpf, Actn3

B, Green module

GO/KEGGTermDescriptionP-valueGene

GO-BPGO:0030199Collagen fibril organization1.00E-04Col3A1, Lox, Col5A2, Col5A1
GO:0030198Extracellular matrix organization4.00E-04Col3A1, Ccdc80, Lox, Col5A2, Col5A1
GO:0042060Wound healing6.00E-04Ccnb1, Casp3, Col3A1, Lox, Tfpi2, Col5A1
GO:0030155Regulation of cell adhesion1.30E-03Tnc, Ccdc80, Jak2, Mmp14, Col8A1
GO:0007049Cell cycle2.10E-03Ccnb1, Ccnb2, Mki67, Cks2, Pttg1, Cdkn3, Ube2C, Racgap1
GO-CCGO:0031012Extracellular matrix6.06E-14Matn2, Aspn, Cthrc1, Tnc, Col3A1, Ccdc80, Mmp14, Col5A2, Col5A1, Col6A2, Col6A1, Lox, Loxl1, Thbs4, Adamts4, Spon1
GO:0005578Proteinaceous extracellular matrix4.22E-12Matn2, Aspn, Cthrc1, Tnc, Col3A1, Ccdc80, Lox, Mmp14, Col5A2, Loxl1, Col5A1, Adamts4, Spon1, Thbs4
GO:0044421Extracellular region part1.36E-09Matn2, Aspn, Cthrc1, Tnc, Col3A1, Ccdc80, Mmp14, Col5A2, Col5A1, Ctsk, Cpxm1, Col6A2, Col6A1, Lox, Loxl1, Thbs4, Adamts4, Spon1
GO:0005576Extracellular region1.18E-08Matn2, Aspn, Cthrc1, Aebp1, Tnc, Col3A1, Ccdc80, Mmp14, Col5A2, Col5A1, Ctsk, Penk, Cpxm1, Sfrp4, Col6A2, Col6A1, Lox, Loxl1, Tfpi2, Thbs4, Adamts4, Spon1
GO:0005581Collagen9.18E-05Col3A1, Lox, Col5A2, Col5A1
GO-MFGO:0016641Oxidoreductase activity, acting on the CH-NH2 group of donors, oxygen as acceptor1.50E-03Lox, Loxl2, Loxl1
GO:0016638Oxidoreductase activity, acting on the CH-NH2 group of donors2.20E-03Lox, Loxl2, Loxl1
GO:0005201Extracellular matrix structural constituent5.20E-03Col3A1, Col5A2, Col5A1
GO:0016702Oxidoreductase activity, acting on single donors with incorporation of molecular oxygen, incorporation of two atoms of oxygen6.40E-03Plod2, P4Ha3, Hpd
GO:0016701Oxidoreductase activity, acting on single donors with incorporation of molecular oxygen6.80E-03Plod2, P4Ha3, Hpd
KEGGrno04512ECM-receptor interaction6.37E-07Tnc, Col3A1, Col6A2, Col6A1, Col5A2, Col5A1, Thbs4
rno04510Focal adhesion1.03E-04Tnc, Col3A1, Col6A2, Col6A1, Col5A2, Col5A1, Thbs4
rno04115p53 signaling pathway2.96E-02Ccnb1, Casp3, Ccnb2

[i] GO, gene ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; BP, biological process; CC, cellular component; MF, molecular function.

Discussion

In the present study, a total of 693 genes were identified by combining the DEGs of the SCI3d, SCI8d and SCI14d groups. The results obtained demonstrated that the PPAR signaling pathway, in which CD36 was significantly enriched, was one of the significant pathways in the pink module, while the p53 signaling pathway that Casp3 was significantly enriched in, was one of the significant pathways in the green module.

PPAR, which includes 3 isoforms (PPAR-α PPAR-γ PPAR-β/δ), is involved in the inflammation process (22). A previous study demonstrated that PPAR-γ and PPAR-δ are involved in the protective effects of palmitoylethanolamide (PEA) in SCI, indicating that PPAR-γ and PPAR-δ may contribute to the anti-inflammatory effects of PEA in SCI (23). It has been reported that PPAR participates in the pathogenesis of diseases, such as diabetes and SCI (24,25). In addition, it has been observed that the decrease of PPAR-δ expression in the spinal cord of streptozotocin (STZ)-diabetic rats may explain the higher mortality rate observed following SCI in STZ-diabetic rats (26). Thus, activation of PPAR-δ may reduce the severity of SCI, and PPAR-δ may be a target for therapy in SCI patients (27). In addition, van Neerven and Mey (28) indicated that endogenous PPAR ligands may contribute by preventing the expansion of the initial damage via modulating inflammation post-SCI, and concluded that the PPAR signaling pathway may be a target for treatment of SCI. Therefore, the results of the present study are in agreement with the results of previous studies and, as such, the PPAR signaling pathway may be closely associated with the pathogenesis of SCI. In addition, the results demonstrated that CD36 was significantly enriched in the PPAR signaling pathway. It has been indicated that the upregulation of CD36, an integral membrane protein, may resolve inflammation via the 5-lipoxygenase-dependent and PPAR-γ signaling pathways (29). The wnt-1 proto-oncogene protein promotes CD36 expression via β-catenin and PPAR-γ signaling pathways (30). CD36 is involved in the PPAR-γ signaling pathway, and activation of PPAR-γ leads to upregulation of CD36 in the PPAR signaling pathway (31). Previous studies have indicated that CD36 is involved in the PPAR-γ signaling pathway (22,32). Although the roles of CD36 in SCI have not been extensively studied, when considering the association between the PPAR signaling pathway and SCI, it is possible that CD36 may be involved in the progression of SCI via the PPAR signaling pathway.

The p53 signaling pathway was observed to be one of the significant KEGG pathways in the green module in the present study. A previous study observed DNA damage-induced p53-mediated mitochondrial apoptosis in SCI (5). Nerve injury is a significant consequence of SCI, and p53 is involved in glial cell death and survival in SCI (33). p53, a key molecular regulator of apoptotic cell death, is known to promote apoptosis by increasing the transcription of target genes (34,35). In addition, a number of apoptosis-associated molecules are expressed via p53, and apoptosis induction via the p53 pathway is a complex process (33). Furthermore, minocycline has been shown to reduce apoptosis in models of SCI (36). Therefore, the results of the present study are consistent with previous studies, and therefore indicate that the p53 signaling pathway may be significant in the progression of SCI. In the present study, it was demonstrated that Casp3 was significantly enriched in the p53 signaling pathway. It has been observed that Casp3 and the p53 signaling pathway may regulate nitric oxide-induced extracellular signal-regulated protein kinase and p38 kinase (37). In addition, p53 prevents the α6β4 integrin-mediated activation of serine/threonine-specific protein kinase B by promoting Casp3 dependent cleavage (38). It has been suggested that Casp3 is a critical mediator of p53-induced apoptosis (39). Zhang et al (40) investigated the apoptosis process in SCI, and demonstrated that the Casp3 apoptotic pathway components are activated following SCI in rats. Ultimately, it is thought that Casp3 may be involved in the progression of SCI via the p53 signaling pathway.

In conclusion, the PPAR and p53 signaling pathways may be important pathways associated with SCI. In addition, CD36 and Casp3 may be involved in the progression of SCI via the PPAR and p53 signaling pathways, respectively. However, the results of the present study are limited by the small sample size, and therefore further studies are required to evaluate the molecular mechanisms underlying SCI progression.

Acknowledgements

The present study was supported by the National Natural Science Foundation of China (grant no. 81472120) and the Science and Technology Commission of Shanghai Municipality (grant no. 14140903900).

Glossary

Abbreviations

Abbreviations:

Casp3

Caspase-3

DEGs

differentially expressed genes

CD36

cluster of differentiation 36

KEGG

Kyoto Encyclopedia of Genes and Genomes

PPAR

peroxisome proliferator-activated receptor

SCI

spinal cord injury

References

1 

Shah RR and Tisherman SA: Spinal Cord Injury. Springer; London: 2014

2 

Me VDB, Castellote JM, Mahillo-Fernandez I and De P-CJ: Incidence of spinal cord injury worldwide: A systematic review. Neuroepidemiology. 34:184–192. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Sekhon LH and Fehlings MG: Epidemiology, demographics, and pathophysiology of acute spinal cord injury. Spine (Phila Pa 1976). 26(24 Suppl): S2–S12. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Wyndaele M and Wyndaele JJ: Incidence, prevalence and epidemiology of spinal cord injury: What learns a worldwide literature survey? Spinal Cord. 44:523–529. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Kotipatruni RR, Dasari VR, Veeravalli KK, Dinh DH, Fassett D and Rao JS: p53- and bax-mediated apoptosis in injured rat spinal cord. Neurochem Res. 36:2063–2074. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Wanner IB, Anderson MA, Song B, Levine J, Fernandez A, Gray-Thompson Z, Ao Y and Sofroniew MV: Glial scar borders are formed by newly proliferated, elongated astrocytes that interact to corral inflammatory and fibrotic cells via STAT3-dependent mechanisms after spinal cord injury. J Neurosci. 33:12870–12886. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Tachibana T, Noguchi K and Ruda MA: Analysis of gene expression following spinal cord injury in rat using complementary DNA microarray. Neurosci Lett. 327:133–137. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Bastien D, Landete V Bellver, Lessard M, Vallières N, Champagne M, Takashima A, Tremblay MÈ, Doyon Y and Lacroix S: IL-1α gene deletion protects oligodendrocytes after spinal cord injury through upregulation of the survival factor Tox3. J Neurosci. 35:10715–10730. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Munro KM, Perreau VM and Turnley AM: Differential gene expression in the EphA4 knockout spinal cord and analysis of the inflammatory response following spinal cord injury. PLoS One. 7:e376352011. View Article : Google Scholar

10 

Guerrero AR, Uchida K, Nakajima H, Watanabe S, Nakamura M, Johnson WE and Baba H: Blockade of interleukin-6 signaling inhibits the classic pathway and promotes an alternative pathway of macrophage activation after spinal cord injury in mice. J Neuroinflammation. 9:402012. View Article : Google Scholar : PubMed/NCBI

11 

Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M, et al: NCBI GEO: Archive for functional genomics data sets-update. Nucleic Acids Res. 41(Database issue): D991–D995. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Gautier L, Cope L, Bolstad BM and Irizarry RA: Affy-analysis of Affymetrix GeneChip data at the probe level. Bioinformatics. 20:307–315. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

14 

Smyth GK: Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 3:Article32004.PubMed/NCBI

15 

Ferreira JA: The Benjamini-Hochberg method in the case of discrete test statistics. Int J Biostat. 3:Article 112007.PubMed/NCBI

16 

Warnes GR, Bolker B, Bonebakker L, Gentleman R, Huber W, Liaw A, Lumley T, Maechler M, Magnusson A, Moeller S, et al: gplots: Various R programming tools for plotting data. R package version. 2:2005.

17 

Oliveros JC: VENNY. An interactive tool for comparing lists with Venn Diagrams. 2007.

18 

Altermann E and Klaenhammer TR: PathwayVoyager: Pathway mapping using the kyoto encyclopedia of genes and genomes (KEGG) database. BMC Genom. 6:203–213. 2005. View Article : Google Scholar

19 

Huang DW, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nature Protoc. 4:44–57. 2008. View Article : Google Scholar

20 

Langfelder P and Horvath S: WGCNA: An R package for weighted correlation network analysis. BMC Bioinformatics. 9:5592008. View Article : Google Scholar : PubMed/NCBI

21 

Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene Ontology: Tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 25:25–29. 2000. View Article : Google Scholar : PubMed/NCBI

22 

Yamanaka M, Ishikawa T, Griep A, Axt D, Kummer MP and Heneka MT: PPARγ/RXRα-induced and CD36-mediated microglial amyloid-β phagocytosis results in cognitive improvement in amyloid precursor protein/presenilin 1 mice. J Neurosci. 32:17321–17331. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Paterniti I, Impellizzeri D, Crupi R, Morabito R, Campolo M, Esposito E and Cuzzocrea S: Molecular evidence for the involvement of PPAR-δ and PPAR-γ in anti-inflammatory and neuroprotective activities of palmitoylethanolamide after spinal cord trauma. J Neuroinflammation. 10:202013. View Article : Google Scholar : PubMed/NCBI

24 

Murphy GJ and Holder JC: PPAR-γ agonists: Therapeutic role in diabetes, inflammation and cancer. Trends Pharmacol Sci. 21:469–474. 2000. View Article : Google Scholar : PubMed/NCBI

25 

McTigue DM: Potential therapeutic targets for PPAR gamma after Spinal Cord Injury. PPAR Res. 2008:5171622008. View Article : Google Scholar : PubMed/NCBI

26 

Tsai CC, Lee KS, Chen SH, Chen LJ, Liu KF and Cheng JT: Decrease of PPARδ in type-1-like diabetic rat for higher mortality after spinal cord injury. PPAR Res. 2014:4563862014. View Article : Google Scholar : PubMed/NCBI

27 

Esposito E and Cuzzocrea S: Targeting the peroxisome proliferator-activated receptors (PPARs) in spinal cord injury. Expert Opin Ther Targets. 15:943–959. 2011. View Article : Google Scholar : PubMed/NCBI

28 

van Neerven S and Mey J: RAR/RXR and PPAR/RXR signaling in spinal cord injury. PPAR Res. 2007:292752007. View Article : Google Scholar : PubMed/NCBI

29 

Ballesteros I, Cuartero MI, Pradillo JM, de la Parra J, Pérez-Ruiz A, Corbí A, Ricote M, Hamilton JA, Sobrado M, Vivancos J, et al: Rosiglitazone-induced CD36 up-regulation resolves inflammation by PPARγ and 5-LO-dependent pathways. J Leukoc Biol. 95:587–598. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Yan H, Wang S, Chen T and Zhu J: oxLDL decreases wnt1 which promotes CD36 through b-catenin and PPAR-r signaling pathway in macrophage. European heart journal Oxford Univ Press; Great Clarendon St, Oxford OX2 6DP, England: pp. 1123. 2014

31 

Nagaraj S, Raghavan AV, Rao SN and Manjappara UV: Obestatin and Nt8U influence glycerolipid metabolism and PPAR gamma signaling in mice. Int J Biochem Cell Biol. 53:414–422. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Lourenco MV and Ledo JH: Targeting Alzheimer's pathology through PPARγ signaling: Modulation of microglial function. J Neurosci. 33:5083–5084. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Saito N, Yamamoto T, Watanabe T, Abe Y and Kumagai T: Implications of p53 protein expression in experimental spinal cord injury. J Neurotrauma. 17:173–182. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Fridman JS and Lowe SW: Control of apoptosis by p53. Oncogene. 22:9030–9040. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Slee EA, O'Connor DJ and Lu X: To die or not to die: How does p53 decide? Oncogene. 23:2809–2818. 2004. View Article : Google Scholar : PubMed/NCBI

36 

Teng YD, Choi H, Onario RC, Zhu S, Desilets FC, Lan S, Woodard EJ, Snyder EY, Eichler ME and Friedlander RM: Minocycline inhibits contusion-triggered mitochondrial cytochrome c release and mitigates functional deficits after spinal cord injury. Proc Natl Acad Sci USA. 101:3071–3076. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Kim SJ, Ju JW, Oh CD, Yoon YM, Song WK, Kim JH, Yoo YJ, Bang OS, Kang SS and Chun JS: ERK-1/2 and p38 kinase oppositely regulate nitric oxide-induced apoptosis of chondrocytes in association with p53, caspase-3, and differentiation status. J Biol Chem. 277:1332–1339. 2002. View Article : Google Scholar : PubMed/NCBI

38 

Bachelder RE, Ribick MJ, Marchetti A, Falcioni R, Soddu S, Davis KR and Mercurio AM: p53 inhibits alpha 6 beta 4 integrin survival signaling by promoting the caspase 3-dependent cleavage of AKT/PKB. J Cell Biol. 147:1063–1072. 1999. View Article : Google Scholar : PubMed/NCBI

39 

Communal C, Sumandea M, De Tombe P, Narula J, Solaro RJ and Hajjar RJ: Functional consequences of caspase activation in cardiac myocytes. Proc Natl Acad Sci USA. 99:6252–6256. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Zhang N, Yin Y, Xu SJ, Wu YP and Chen WS: Inflammation & apoptosis in spinal cord injury. Indian J Med Res. 135:287–296. 2012.PubMed/NCBI

Related Articles

Journal Cover

April-2017
Volume 15 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang YH, Song J, Wang LG and Shao J: Identification of key genes and pathways associated with spinal cord injury. Mol Med Rep 15: 1577-1584, 2017
APA
Zhang, Y., Song, J., Wang, L., & Shao, J. (2017). Identification of key genes and pathways associated with spinal cord injury. Molecular Medicine Reports, 15, 1577-1584. https://doi.org/10.3892/mmr.2017.6192
MLA
Zhang, Y., Song, J., Wang, L., Shao, J."Identification of key genes and pathways associated with spinal cord injury". Molecular Medicine Reports 15.4 (2017): 1577-1584.
Chicago
Zhang, Y., Song, J., Wang, L., Shao, J."Identification of key genes and pathways associated with spinal cord injury". Molecular Medicine Reports 15, no. 4 (2017): 1577-1584. https://doi.org/10.3892/mmr.2017.6192