12
Research Article Analysis of Crucial Genes and Pathways Associated with Spared Nerve Injury-Induced Neuropathic Pain Dong Mao , 1 Huang Zhai, 2 Gang Zhao , 3 Jingyi Mi, 2 and Yongjun Rui 4 1 Orthopaedic Institute, Wuxi 9th Peoples Hospital Aliated to Soochow University, Wuxi 214062, China 2 Department of Sports Medicine, Wuxi 9th Peoples Hospital Aliated to Soochow University, Wuxi 214062, China 3 Department of Hand Surgery, Wuxi 9th Peoples Hospital Aliated to Soochow University, Wuxi 214062, China 4 Department of Orthopedics, Wuxi 9th Peoples Hospital Aliated to Soochow University, Wuxi 214062, China Correspondence should be addressed to Gang Zhao; [email protected] Received 25 September 2020; Revised 6 November 2020; Accepted 10 November 2020; Published 21 November 2020 Academic Editor: Preston E. Garraghty Copyright © 2020 Dong Mao et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Purpose. The study was aimed at elucidating the molecular mechanism underlying neuropathic pain induced by spared nerve injury (SNI). Methods. The microarray data of GSE30691 were downloaded from the Gene Expression Omnibus database, including sciatic nerve lesion samples at 3, 7, 21, and 40 days after SNI and sham control samples at 3, 7, and 21 days. Dierential analysis along with Mfuzz clustering analysis was performed to screen crucial clusters and cluster genes. Subsequently, comprehensive bioinformatic analyses were performed, including functional enrichment analysis, protein-protein interaction (PPI) network and module analysis, and transcription factor- (TF-) gene and miRNA-target interaction predictions. Moreover, the screened dierentially expressed genes (DEGs) were corroborated using two other microarray datasets. Results. Three clusters with dierent change trends over time after SNI were obtained. Protein kinase CAMP-activated catalytic subunit beta (Prkacb), complement C3 (C3), and activating transcription factor 3 (Atf3) were hub nodes in the PPI network, and broblast growth factor 9 (Fgf9) was found to interact with more TFs. Prkacb and Fgf9 were signicantly enriched in the MAPK signaling pathway. Moreover, rno-miR-3583-5p was targeted by Fgf9, and rno-miR-1912-3p was targeted by neuregulin 1 (Nrg1). Key genes like Nrg1 and Fgf9 in cluster 1, Timp1 in cluster 2, and Atf3 and C3 in cluster 3 were screened out after corroborating microarray data with other microarray data. Conclusions. Key pathways like the MAPK signaling pathway and crucial genes like Prkacb, Nrg1, Fgf9, Timp1, C3, and Atf3 may contribute to SNI-induced neuropathic pain development in rats. 1. Introduction Neuropathic pain refers to chronic pain originating from neurological pathology, and it aects approximately 710% of the global population [1, 2]. It is characterized by sponta- neous hyperalgesia, dysesthesia, and allodynia [3, 4]. Neuro- pathic pain can negatively aect quality of life, and most neuropathic pain patients may suer from negative moods like depression and anxiety disorders [3, 5, 6]. Despite great progress in understanding the pathogenesis of neuropathic pain and patient prognosis, little is known about the genetic basis and mechanism underlying this disease, and many patients respond poorly to current therapies. A better under- standing of the molecular mechanism of neuropathic pain will be important for further eective therapies. The pathogenesis of neuropathic pain is complex, and elucidation of specic molecular alterations helps understand the mechanisms involved in neuropathic pain development. Several biological alterations like ion channel or inamma- tory mediator expression, extracellular proteins, and epige- netic inuences have been implicated in neuropathic pain [7]. In recent years, microarray data have been widely used to globally assess gene expression signatures that provide new insights into disease pathophysiology [8]. Spared nerve injury (SNI) is a robust neuropathic pain model [9]. Since the development of bioinformatics, massive microarray data have been used to extensively investigate the candidate mol- ecules associated with SNI-induced neuropathic pain and help identify potential targets for disease diagnosis and treat- ment. For instance, crucial genes like C-X-C motif Hindawi Neural Plasticity Volume 2020, Article ID 8822001, 12 pages https://doi.org/10.1155/2020/8822001

Analysis of Crucial Genes and Pathways Associated with ...downloads.hindawi.com/journals/np/2020/8822001.pdfSep 25, 2020  · different change trends over time after SNI were obtained

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Analysis of Crucial Genes and Pathways Associated with ...downloads.hindawi.com/journals/np/2020/8822001.pdfSep 25, 2020  · different change trends over time after SNI were obtained

Research ArticleAnalysis of Crucial Genes and Pathways Associated with SparedNerve Injury-Induced Neuropathic Pain

Dong Mao ,1 Huang Zhai,2 Gang Zhao ,3 Jingyi Mi,2 and Yongjun Rui4

1Orthopaedic Institute, Wuxi 9th People’s Hospital Affiliated to Soochow University, Wuxi 214062, China2Department of Sports Medicine, Wuxi 9th People’s Hospital Affiliated to Soochow University, Wuxi 214062, China3Department of Hand Surgery, Wuxi 9th People’s Hospital Affiliated to Soochow University, Wuxi 214062, China4Department of Orthopedics, Wuxi 9th People’s Hospital Affiliated to Soochow University, Wuxi 214062, China

Correspondence should be addressed to Gang Zhao; [email protected]

Received 25 September 2020; Revised 6 November 2020; Accepted 10 November 2020; Published 21 November 2020

Academic Editor: Preston E. Garraghty

Copyright © 2020 Dong Mao et al. This is an open access article distributed under the Creative Commons Attribution License,which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Purpose. The study was aimed at elucidating the molecular mechanism underlying neuropathic pain induced by spared nerve injury(SNI). Methods. The microarray data of GSE30691 were downloaded from the Gene Expression Omnibus database, includingsciatic nerve lesion samples at 3, 7, 21, and 40 days after SNI and sham control samples at 3, 7, and 21 days. Differential analysisalong with Mfuzz clustering analysis was performed to screen crucial clusters and cluster genes. Subsequently, comprehensivebioinformatic analyses were performed, including functional enrichment analysis, protein-protein interaction (PPI) network andmodule analysis, and transcription factor- (TF-) gene and miRNA-target interaction predictions. Moreover, the screeneddifferentially expressed genes (DEGs) were corroborated using two other microarray datasets. Results. Three clusters withdifferent change trends over time after SNI were obtained. Protein kinase CAMP-activated catalytic subunit beta (Prkacb),complement C3 (C3), and activating transcription factor 3 (Atf3) were hub nodes in the PPI network, and fibroblast growthfactor 9 (Fgf9) was found to interact with more TFs. Prkacb and Fgf9 were significantly enriched in the MAPK signalingpathway. Moreover, rno-miR-3583-5p was targeted by Fgf9, and rno-miR-1912-3p was targeted by neuregulin 1 (Nrg1). Keygenes like Nrg1 and Fgf9 in cluster 1, Timp1 in cluster 2, and Atf3 and C3 in cluster 3 were screened out after corroboratingmicroarray data with other microarray data. Conclusions. Key pathways like the MAPK signaling pathway and crucial genes likePrkacb, Nrg1, Fgf9, Timp1, C3, and Atf3 may contribute to SNI-induced neuropathic pain development in rats.

1. Introduction

Neuropathic pain refers to chronic pain originating fromneurological pathology, and it affects approximately 7–10%of the global population [1, 2]. It is characterized by sponta-neous hyperalgesia, dysesthesia, and allodynia [3, 4]. Neuro-pathic pain can negatively affect quality of life, and mostneuropathic pain patients may suffer from negative moodslike depression and anxiety disorders [3, 5, 6]. Despite greatprogress in understanding the pathogenesis of neuropathicpain and patient prognosis, little is known about the geneticbasis and mechanism underlying this disease, and manypatients respond poorly to current therapies. A better under-standing of the molecular mechanism of neuropathic painwill be important for further effective therapies.

The pathogenesis of neuropathic pain is complex, andelucidation of specific molecular alterations helps understandthe mechanisms involved in neuropathic pain development.Several biological alterations like ion channel or inflamma-tory mediator expression, extracellular proteins, and epige-netic influences have been implicated in neuropathic pain[7]. In recent years, microarray data have been widely usedto globally assess gene expression signatures that providenew insights into disease pathophysiology [8]. Spared nerveinjury (SNI) is a robust neuropathic pain model [9]. Sincethe development of bioinformatics, massive microarray datahave been used to extensively investigate the candidate mol-ecules associated with SNI-induced neuropathic pain andhelp identify potential targets for disease diagnosis and treat-ment. For instance, crucial genes like C-X-C motif

HindawiNeural PlasticityVolume 2020, Article ID 8822001, 12 pageshttps://doi.org/10.1155/2020/8822001

Page 2: Analysis of Crucial Genes and Pathways Associated with ...downloads.hindawi.com/journals/np/2020/8822001.pdfSep 25, 2020  · different change trends over time after SNI were obtained

chemokine receptor 2 protein coding (CXCR2) and Gprotein-coupled receptor kinase 1 (GRK1) and miRNAs likemiR-208a-5p and miR-135a-2-3p are differentially expressedin SNI based on microarray data, suggesting a possible role inneuropathic pain [10]. In addition to genes and miRNAs,several pathways like focal adhesion are also revealed to beinvolved in neuropathic pain development [11]. However,the possible mechanism underlying neuropathic painremains incompletely understood, and reliable biomarkersfor diagnosis and treatment are lacking.

In previous studies, the microarray data GSE30691 devel-oped by Costigan et al. [12] has been used to identify novelpain-related genes by mining expression profiling data inthree rodent neuropathic pain models using an iterativelyreweighted least squares outlier-resistant regression methodand weighted gene coexpression network analysis, followedby analysis of the associations between polymorphisms inthe gene and pain phenotypes in human cohorts by a combi-nation of bioinformatic analysis of transcriptional changes inrodent models and human gene polymorphism associationstudies. As a result, a neuropathic potassium channel modu-latory subunit (also called Kv9.1) was downregulated in allthree neuropathic pain models, and a common amino acid-altering KCNS1 polymorphism is associated with the painphenotype in five of six independent cohorts [12]. Moreover,the microarray data GSE30691 are used to screen candidategenes associated with neuropathic pain using functionaland weighted coexpression modular analysis [13] or differen-tial analysis with a random walk with restart [14]. In contrastto these previous studies, we also downloaded the microarraydata GSE30691 from the NCBI Gene Expression Omnibus(GEO) [15] and reanalyzed by other bioinformatic methods,aiming to find more neuropathic pain-related genes andpathways. In detail, differential analysis along with Mfuzzclustering analysis was utilized to screen crucial clustersand cluster genes. Subsequently, comprehensive bioinfor-matic analyses were performed, including functional enrich-ment analysis, protein-protein interaction (PPI) network andmodule analysis, and transcription factor- (TF-) gene andmiRNA-target interaction predictions. Moreover, thescreened DEGs were corroborated using two other microar-ray datasets. Our findings will help to elucidate key molecularmechanisms associated with neuropathic pain and discovernew potential targets for disease therapies.

2. Materials and Methods

2.1. Data Sources. The microarray data GSE30691 depositedin the NCBI GEO (http://www.ncbi.nlm.nih.gov/geo/) data-base by Costigan et al. [12], which were generated on theGPL85 [RG_U34A] Affymetrix Rat Genome U34 Array plat-form, was downloaded. This dataset contained adult rat L4and L5 dorsal root ganglion (DRG) ipsilateral samples fromdifferent sciatic nerve lesions, including SNI, spinal nerveligation, and chronic constriction injury. Expression profilingof sciatic nerve lesion samples at 3, 7, 21, and 40 days afterSNI and 3, 7, and 21 days of sham control samples (n = 3per time point; total = 24) were extracted. The data weredownloaded in May 2020.

2.2. Differential Expression Analysis. The differential expres-sion of probes between SNI samples and sham controls wasanalyzed at time points of 3, 7, and 21 days post-SNI usingthe online tool GEO2R (http://www.ncbi.nlm.nih.gov/geo/geo2r/). The probes that did not map to gene symbols, orprobes mapping to different genes were removed. Thethreshold value was set to p value < 0.05 and ∣log ðfoldchangeÞ ∣ >0:585. The number of differentially expressedprobes and genes was counted. The unions of DEGs thathad the same expression change tendency at three timepoints were collected for further analysis.

2.3. Mfuzz Clustering Analysis. To study the expression pat-tern of DEGs with the time-course injury, the expressionvalues of DEGs in 0, 7, 21, and 40 days of SNI samples wereextracted before using clustering analysis with Mfuzz (ver-sion 2.42.0, http://bioconductor.org/packages/release/bioc/html/Mfuzz.html) [16] in the R environment. In the cluster-ing analysis, the optimal cluster number was calculated usingdefault parameters. Meanwhile, the min score (membership)threshold was set to 0.6. For a given cluster, if the member-ship of two genes was higher, their expression trend wasmore similar, representing the importance of the gene inthe cluster to a certain extent. Clusters with different changetrends of genes over time were used for subsequent analysis.Cluster genes in each cluster were obtained using the onlinetool Metascape (http://metascape.org) [17].

2.4. Functional Enrichment Analysis. To investigate the func-tion of cluster genes in each cluster, Gene Ontology (GO)[18] biological process (BP) and Kyoto Encyclopedia ofGenes and Genomes (KEGG) [19] pathway enrichment anal-yses were performed, which are widely used for the func-tional annotation of large-scale data. The species was set asRattus norvegicus, and the parameters were default, includingmin overlap: 3, p value cutoff: 0.01, and min enrichment: 1.5.After obtaining the terms conforming to the above parame-ters, further term clustering was performed based on the sim-ilarity of genes enriched in each term > 0:3, and the term withthe greatest statistical significance (p value minimum) wasselected to represent the term cluster. The top 20 term clus-ters with the most significance were displayed. In addition,the interaction network of terms was constructed to furthercapture the relationship between terms in term clusters.The selection criteria for terms in this network were as fol-lows: the terms with the best p value in each cluster of thetop 20 clusters were selected, there were ≤15 terms in eachcluster, ≤250 terms in total, and similarity of >0.3. Afterobtaining the interaction relations of terms, the interactivenetwork of terms was established and visualized using Cytos-cape (version 3.4.0, http://chianti.ucsd.edu/cytoscape-3.4.0/)[20].

2.5. PPI Network Construction and Module Analysis. TheSearch Tool for the Retrieval of Interacting Genes (STRING)database [21] was used to analyze interactions between thegene coding proteins. The PPI pairs between cluster geneswere analyzed using a medium confidence of 0.4. The specieswas rat. The PPI network was constructed using Cytoscape

2 Neural Plasticity

Page 3: Analysis of Crucial Genes and Pathways Associated with ...downloads.hindawi.com/journals/np/2020/8822001.pdfSep 25, 2020  · different change trends over time after SNI were obtained

software. The degree centrality of nodes was excavated usingthe CytoNCA plugin (version 2.1.6, http://apps.cytoscape.org/apps/cytonca) [22] without weighting, and the hubnodes were identified by ranking the node degree. In addi-tion, the modules of the PPI network with nodes > 3 werescreened out using the Molecular Complex Detection(MCODE) (version 1.5.1, http://apps.cytoscape.org/apps/mcode) [23] plugin. The parameters were set to default,including Degree Cutoff = 2, Node Score Cutoff = 0:2, k‐core = 2, and max depth = 100. KEGG pathway enrichmentanalysis for DEGs in modules were conducted using cluster-Profiler (version:3.8.1,http://bioconductor.org/packages/release/bioc/html/clusterProfiler.html) [24]. The p valuewas adjusted using the Benjamini and Hochberg (BH)approach [25], and p adjust < 0.05 was defined as significant.

2.6. TF-Gene Interaction Prediction. TFs that could interactwith cluster genes were predicted using OverrepresentationEnrichment Analysis (ORA) with the online tool WebGestalt(version WebGestalt 2019, http://www.webgestalt.org/) [26].The species was Rattus norvegicus. The threshold values wereset as follows: enriched genes > 5 and false discovery rate ðFDRÞ < 0:05. The TF-target network was established byCytoscape.

2.7. miRNA-Target Interaction Prediction. We used miR-Walk2.0 (http://zmf.umm.uniheidelberg.de/apps/zmf/mirwalk2/generetsys-self.html) [27] to predict the upstreammiRNAs that could target cluster genes. The species wasrat. Meanwhile, the miRNA-target pairs should be simulta-neously presented in miRWalk, miRanda, miRmap, RNAhy-brid, and TargetScan databases. The miRNA-targetregulatory network was visualized using Cytoscape.

2.8. DEG Corroboration with More Microarray Data. Themicroarray data GSE15041 deposited by Vega-Avelairaet al. [28] and GSE18803 deposited by Costigan et al. [29],which were developed on the [Rat230_2] Affymetrix RatGenome 230 2.0 Array and GPL341 [RAE230A] AffymetrixRat Expression 230A Array platforms, respectively, were alsodownloaded from NCBI GEO. The GSE15041 dataset con-tained 16 neonates (P10) or adult (8-12wk) rat L4 and L5

DRG ipsilateral or contralateral seven days post-SNI/shamcontrol samples. GSE18803 contained 24 neonates (P10) oradult (8-12wk) rat DRG ipsilateral seven days post-SNI/-sham control samples. The expression profiling data of threeadult rat L4/L5 DRG ipsilateral SNI samples and three shamcontrols in the GSE15041 dataset and the expression profil-ing data of six adult rat DRG ipsilateral SNI samples andsix sham controls in the GSE18803 dataset were selected toensure sample source and age consistency. The data weredownloaded in May 2020. The expression data of three adultrat L4/L5 DRG ipsilateral SNI samples and three sham con-trols in the GSE15041 dataset have been used to analyze thekey genes associated with neuropathic pain in several studies[28, 30]. Similarly, the expression profiling data of six adultrat DRG ipsilateral SNI samples and six sham controls inthe GSE18803 dataset have also been utilized to identify cru-cial genes involved in neuropathic pain development [29, 31–33]. We thus selected the two microarray data for DEG cor-roboration. In this study, the differential expression of probesbetween SNI samples and sham controls in the GSE15041and GSE18803 databases were also analyzed using thismethod. The intersection of cluster genes obtained from theGSE30691 dataset and DEGs obtained from the GSE15041and GSE18803 datasets were acquired to corroborate the dif-ferential expression of key cluster genes.

3. Results

3.1. DEG Analysis. GSE30691 dataset analysis using theGEO2R online tool screened out 180 (109 up- and 71 down-regulated genes), 230 (133 up- and 97 downregulated genes),and 153 (124 up- and 29 downregulated genes) DEGsbetween SNI samples and sham controls at the time pointsof 3, 7, and 21 days post-SNI. The unions of DEGs that hadthe same expression change tendency at three time pointswere analyzed, and a total of 306 DEGs were obtained forsubsequent analysis.

3.2. Mfuzz Clustering Analysis. The expression trends of 306DEGs at 0, 3, 7, 21, and 40 days post-SNI were analyzed usingMfuzz clustering analysis, and three clusters with differentchange trends over time were screened out (Figure 1). Cluster

Cluster 1

Time0 3 7 21 40

Expr

essio

n ch

ange

s

1.5

0.5

–0.5

–1.5

Time0 3 7 2121 40

Expr

essio

n ch

ange

s

1.51.00.5

–0.5

–1.5

Cluster 2

Time0 3 7 2121 40

Expr

essio

n ch

ange

s

1.5

0.5

–0.5

–1.5

Cluster 3

Figure 1: The results of Mfuzz clustering analysis. Three clusters with different change trends over time were screened out. The horizontalaxis represents the days after spared nerve injury (SNI), and the vertical axis represents the relative expression value of cluster genes. Thecolor from blue to red indicates that the membership value of this gene increases, which represents the gene’s importance in the cluster tosome extent.

3Neural Plasticity

Page 4: Analysis of Crucial Genes and Pathways Associated with ...downloads.hindawi.com/journals/np/2020/8822001.pdfSep 25, 2020  · different change trends over time after SNI were obtained

GO:0006813: potassium ion transportGO:0043267: negative regulation of potassium ion transportGO:0023061: signal releaseGO:0009612: response to mechanical stimulusGO:0001505: regulation of neurotransmitter levelsGO:0006898: receptor-mediated endocytosisGO:1900084: regulation of peptidyl-tyrosine autophosphorylationGO:0048545: response to steroid hormoneGO:1901652: response to peptideGO:0007610: behaviorGO:2001023: regulation of response to drugGO:0042391: regulation of membrane potentialGO:0014706: striated muscle tissue developmentGO:0050804: modulation of chemical synaptic transmissionGO:0010038: response to metal ionGO:0044057: regulation of system processGO:0043408: regulation of MAPK cascaderno04010: MAPK signaling pathwayGO:0006954: inflammatory responseGO:0009611: response to wounding

Clus

ter 1

Clus

ter 2

Clus

ter 3

0

234

6

10

20

–Log

10(P

)

(a)

Response to peptide

Response to metal ion

MAPK signaling pathway

Regulation of MAPK cascade

Striated muscle tissue development

Regulation of peptidyl-tyrosine

autophosphorylation

Regulation of system process

Regulation of membrane

potential

Negative regulation of potassium ion

transport Potassium ion transport

signal releaseModulation of

chemical synaptic transmission

Regulation of response to drug

Regulation of neurotransmitter

levels

Behavior

Response to mechanical

stimulus

Inflammatoryresponse

Receptor-mediatedendocytosis

Response to wounding

Response to syteroid

Cluster 2Cluster 1

Cluster 3

hormone

(b)

Figure 2: The significantly enriched cluster terms. (a) The top 20 enriched cluster terms by cluster genes in three clusters. The darker thecolor, the smaller the p value. (b) The term interaction network constructed by important terms in clusters. The color of different nodesrepresents different clusters, and the connection line represents the genetic similarity between terms.

4 Neural Plasticity

Page 5: Analysis of Crucial Genes and Pathways Associated with ...downloads.hindawi.com/journals/np/2020/8822001.pdfSep 25, 2020  · different change trends over time after SNI were obtained

Aif1Vgf

Tgm1

Syt1

Cd55Casr

Rapgef4

Pcsk2C1qbGal

Slc3a1

Rgs18Ucn

Adcyap1

PygbLyz2

Ctss

Pfkm

RT1-Bb

Klc1 Fcgr2bTubb2b

Stmn4

Aplp2

C3Car2

Serpina1

Htr1d

Myl1

Mylpf

Ckmt2

Tnnt3

Myh4

Kcnk1

Dpp6

Kcna1

Acsl6

Abcd3

LOC688459

Dhfr

Spr

Rrad

Pdlim1 Crisp1

Slc1a3Vsnl1

Scd

Plin2

Nfia

Syt9

Gpam

Snap25

Gabbr2

Vamp1

Nsf

Slc12a2

Syt11

Gabrg2

Gpc1Ppp1r2

Sdc1Gfra2

Cav1

Ppp2r2cHspb1Fgf9

Gadd45a

Ndufs1

JunPrkacb

CrkHtr3a

Mapk9

Ppp3r1

Camk2dFgf1

Vegfa

Ap1s1Serpinb2

Ssr3

Canx

Nrg1

Fstl1

Utrn

Dgcr6 Timp1

RT1-Db1

Mmp16

Rgs4 Hsph1

St13

Hsp90b1

Timp3Trim23

Scn1a

Kcnc2 Gabra5

Kcns3Sprr1a

Kcns1

Kcnab1

Cadm1

Ptpn5

Sox11

Tspo

Pak1

Gap43Atp1a1

RhocVipPrkce

Atf3Ppp1r1a

AcadsbCacna2d1

Ecel1 Etfdh

(a)

Syt11

Syt9Nsf

Syt1

Snap25

Vamp1

C3

Aplp2

Vgf

Timp1

Fstl1

Myh4

Tnnt3

Myl1

Mylpf

Mapk9

Ppp3r1

Prkacb

Jun

Atp1a1

Hspb1

Gap43

Ecel1

Aif1

Fcgr2b

C1qb

Sox11

Ctss

Sprr1a

Module 1 Module 2

Module 3 Module 4 Module 5

(b)

Figure 3: Continued.

5Neural Plasticity

Page 6: Analysis of Crucial Genes and Pathways Associated with ...downloads.hindawi.com/journals/np/2020/8822001.pdfSep 25, 2020  · different change trends over time after SNI were obtained

1 contained 77 DEGs and presented a trend of decreasingbefore increasing. The expression value reached the lowestlevel from three to seven days, and the overall expression ofthe SNI group was decreased compared to the control group(0 days after injury). Cluster 2 contained 13 DEGs and pre-sented a trend of increasing before decreasing. The expres-sion level rose to the highest level on the third day beforegradually declining, and the overall SNI group expressionwas higher than that of the control group. Cluster 3 con-tained 49 DEGs and presented a trend of increasing beforeleveling off. The expression level rose to the highest level onday three before gradually declining, and the overall SNIgroup expression was higher than that of the control group.

3.3. Functional Enrichment Analysis. We performed GO BPand KEGG pathway analyses for cluster genes in three clus-ters. In total, 344 GO BP and 33 KEGG pathway terms weresignificantly enriched by cluster genes in cluster 1, one GOBP and no KEGG pathway term was enriched by clustergenes in cluster 2, and 187 GO BP and 20 KEGG pathwayterms were enriched by cluster genes in cluster 3. The top20 enriched cluster terms are shown in Figure 2(a). More-

over, several terms were simultaneously enriched by clustergenes in different clusters like GO:0009611: response towounding, GO:0043408: regulation of MAPK cascade, andrno04010:MAPK signaling pathway. Subsequently, the terminteraction network was constructed using important termsin clusters (Figure 2(b)).

3.4. PPI Network Construction. The PPI network of clustergenes in three clusters included 117 nodes and 243 interac-tions (Figure 3(a)). Notably, the nodes coded by cluster 1genes were all downregulated, and the nodes coded by cluster2 and 3 genes were all upregulated in this PPI network. Basedon connectivity degree analysis, the nodes with greaterconnectivity degrees were protein kinase CAMP-activatedcatalytic subunit beta (Prkacb), complement C3 (C3),synaptosome-associated protein 25 (Snap25), activatingtranscription factor 3 (Atf3), and protein phosphatase 3regulatory subunit B, alpha (Ppp3r1), which were consideredas PPI network hub nodes. Notably, Prkacb, Ppp3r1, andMapk9 were revealed to be remarkably enriched in therno04010:MAPK signaling pathway. Further module analy-sis identified five modules with node > 3 (Figure 3(b)). The

Amphetamine addictioncAMP signaling pathway

Complement and coagulation cascadesFocal adhesion

GABAergic synapseHuman T-cell leukemia virus 1 infection

Insulin secretionLegionellosis

LeishmaniasisLeukocyte transendothelial migration

MAPK signaling pathwayPertussis

PhagosomePrion diseases

Regulation of actin cytoskeletonSalmonella infection

SNARE interactions in vesicular transportStaphylococcus aureus infection

Synaptic vesicle cycleTight junction

TuberculosisVasopressin-regulated water reabsorption

Wnt signaling pathway

Mod

ule 1

Mod

ule 2

Mod

ule 3

Mod

ule 4

Mod

ule 5

Module

KEG

G p

athw

ay

Count12

3

4

5

2

3

4

−Log10(P adjust)

(c)

Figure 3: Protein-protein interaction (PPI) network constructed by cluster genes in three clusters, five modules, and KEGG pathway analysisfor module genes. (a) The PPI network and (b) five modules. Red nodes represent cluster 1 genes, blue nodes represent cluster 2 genes, andgreen nodes represent cluster 3 genes. Circle nodes represent upregulated genes, and rhombus nodes represent downregulated genes. Nodesize represents the degree of connectivity. (c) KEGG pathway analysis for module genes. Bubble size indicates the number of enriched genes.The redder the bubble color, the more significant this pathway.

6 Neural Plasticity

Page 7: Analysis of Crucial Genes and Pathways Associated with ...downloads.hindawi.com/journals/np/2020/8822001.pdfSep 25, 2020  · different change trends over time after SNI were obtained

KEGG pathway showed that genes in modules 1-5 were dra-matically enriched in the synaptic vesicle cycle, complementand coagulation cascades, focal adhesion, wnt signaling path-way, and phagosome (Figure 3(c)).

3.5. TF-Gene Regulatory Network Analysis. TFs that couldinteract with cluster genes were predicted using WebGestalt.The results showed that 15 TFs that could interact with clus-ter 1 genes were obtained, whereas TFs that could interactwith cluster 2 and 3 genes were not predicted. The TF-generegulatory network containing 15 TFs and 54 target cluster1 genes was constructed (Figure 4). Most of cluster 1 genesin this network were downregulated. According to the num-ber of target TFs, the important cluster 1 genes that couldinteract with more TFs were protein phosphatase 2 regula-tory subunit B gamma (Ppp2r2c), visinin-like 1 (Vsnl1), and

fibroblast growth factor 9 (Fgf9). Fgf9 was also found to beenriched in the rno04010:MAPK signaling pathway.

3.6. miRNA-Target Regulatory Network Analysis. In total,4546 miRNA-target interactions, including 681 miRNAsand 103 target genes, were obtained using miRWalk2.0. Sincethere were so many miRNAs, the miRNAs with targetcluster genes ≥ 15 were screened out again. The miRNA-target regulatory network was established, containing 14miRNAs and 70 target cluster genes (Figure 5). Among them,rno-miR-3583-5p was found to be targeted by Fgf9 and rno-miR-1912-3p was targeted by neuregulin 1 (Nrg1).

3.7. DEG Corroboration with More Microarray Data. To ver-ify the important cluster genes, microarray data GSE15041and GSE18803 were also downloaded from NCBI GEO andused to identify DEGs between SNI samples and sham

St13

Myl1Myh4

Cd55

Kcnc2

Pygb

Rgs4

SF1_Q6

Cadm1

Kcns1

Dpp6

Acadsb

TEF_Q6

Cast

MEF2_Q6_01

Car2

CEBPA_01

Canx

Pcsk2Atp1a1

Fstl1

Fgf9

Ppp2r2c

Gyg1

Slc24a2

Kcns3

Prkce

Snap25

Mylpf

Slc3a1

Rapgef4

Nfia

Hsph1

S8_01

Scd

TGACCTY_ERR1_Q2

Ckmt2

RTAAACA_FREAC2_01

Syt9

CTTTAAR_UNKNOWN

Aplp2

Csde1

Hsp90b1

Ppp1r2

ZIC2_01

Nrg1

Vegfa

PrkacbAcsl6

Vsnl1

Scn1aYGCANTGCR_UNKNOWN

CrkTGCCAAR_NF1_Q6

Add3

Timp3

HMEF2_Q6

Utrn

Syt1

Vamp1

YY1_01

POU3F2_02Kcna1

IK3_01

Gabbr2Gabrg2

Kcnk1

EtfdhSlc12a2

Figure 4: The transcription factor- (TF-) gene regulatory network. Red nodes indicate the cluster 1 genes, and yellow octagon nodes indicatepredicted TFs. Rhomboid nodes represent downregulated genes and circular nodes represent upregulated genes. Arrow lines indicate TF-target relationship.

7Neural Plasticity

Page 8: Analysis of Crucial Genes and Pathways Associated with ...downloads.hindawi.com/journals/np/2020/8822001.pdfSep 25, 2020  · different change trends over time after SNI were obtained

controls at seven days post-SNI. The results showed that 830(498 up- and 332 downregulated genes) and 188 (184 up-and four downregulated genes) DEGs were screened outbetween SNI samples and sham controls based on theGSE15041 and GSE18803 datasets, respectively. Since mostof cluster 1 genes were downregulated according to the afore-mentioned analysis, we investigated the intersection of clus-ter 1 genes and downregulated genes obtained fromGSE15041 and GSE18803 databases. The results showed that14 intersected genes between cluster 1 genes and downregu-lated genes obtained from the GSE15041 database wereobtained, including Nrg1 and Fgf9 (Figure 6(a)). Since theoverall SNI group expression in cluster 2 and cluster 3 washigher than that in the control group, the intersection of clus-ter 2 genes or cluster 3 genes and upregulated genes obtainedfrom GSE15041 and GSE18803 databases were analyzed. Theresults showed that only one intersected gene, named TIMPMetallopeptidase Inhibitor 1 (Timp1), was obtained betweencluster 2 genes and upregulated genes obtained fromGSE15041 and GSE18803 databases (Figure 6(b)). Nineintersected genes, including Atf3 and C3, were screenedbetween cluster 3 genes and upregulated genes obtained fromGSE15041 and GSE18803 databases (Figure 6(c)).

4. Discussion

Neuropathic pain is chronic pain with an elusive mechanism.To discover the possible mechanism, this study utilized acomprehensive bioinformatic approach to screen candidategenes associated with SNI-induced neuropathic pain. Consis-tent with previous findings by Costigan et al. [12], we alsofound that KCNS1 was downregulated in the SNI-inducedneuropathic pain model, suggesting that our results were reli-able. In addition to this, our analysis further revealed threeclusters with different change trends over time after SNI.Prkacb, C3, and Atf3 were hub nodes in the PPI network,and Fgf9 was found to interact with more TFs. Prkacb andFgf9 were significantly enriched in the MAPK signaling path-way. Moreover, rno-miR-3583-5p was found to be targetedby Fgf9, and rno-miR-1912-3p was targeted by neuregulin 1(Nrg1). After corroborating microarray data with two othermicroarray datasets, key genes, like Nrg1 and Fgf9 in cluster1, Timp1 in cluster 2, and Atf3 and C3 in cluster 3, werescreened out to be implicated in SNI-induced neuropathicpain.

Increasing evidence has revealed that nerve injury leadsto p38 MAPK pathway activation in the spinal cord,

Slc12a2

Vegfa

RT1-Db1Serpina3n

Kcnab1

Pygbrno-let-7a-2-3p

rno-miR-743b-3p

Rab33b

St13

rno-miR-3584-5p

C1qb

Kcnc2

Vip

Litaf

rno-miR-186-5p

Vgf

Reg3b

Gabra5

Mmp16

Scn1a

Smad1

Ecel1

Hsph1

Ppp3r1

Cav1

Sv2b

Gadd45a

Vamp1 Gfra2Plin2

rno-miR-873-3p

Lyz2

Vsnl1

Myl1

Camk2d

Fgf9

rno-miR-3583-5p

rno-miR-322-5p

Csde1

rno-miR-672-3p

rno-miR-203a-3p

Abcd3

Mapk9Tmpo

Nsg1

Rhoc

Canx

Sox11

Htr3a

Atf3

Serpinb2

Pfkm

Gap43

Atp1a1rno-miR-3541

Casr

Ap1s1Gabbr2

Pdlim1

Cast

Crisp1

Rgs4

rno-miR-33-3p

Slc1a3

Ptpro

Sdc1

Ptpn5

Add3

rno-miR-1912-3p

Ppp1r1a

Cd55

Dgcr6

Nrg1

Stmn4

Timp3

Prkce

Gfra1

rno-miR-152-3p

Hsp90b1

Tspo Cacna2d1Kcns1

rno-miR-148b-3p

Figure 5: The miRNA-target regulatory network. Red nodes represent cluster 1 genes, blue nodes represent cluster 2 genes, and green nodesrepresent cluster 3 genes. Circle nodes represent upregulated genes, and rhombus nodes represent downregulated genes. The pink trianglenodes represent the predicted miRNAs with the number of target genes ≥ 15. The gray lines represent target gene regulation by miRNA.

8 Neural Plasticity

Page 9: Analysis of Crucial Genes and Pathways Associated with ...downloads.hindawi.com/journals/np/2020/8822001.pdfSep 25, 2020  · different change trends over time after SNI were obtained

63 279

4

0 0

0

14

Cluster 1 GSE15041

GSE18803

PrkceHtr1dKcns3Sv2bAtp1a1Kcnab1Nrg1

Acsl6Kcns1Htr3aKcnc2Car2Fgf9Ckmt2

(a)

6 382

121

1 47

1

5

Cluster 2 GSE15041

GSE18803

Timp 1

Uck2UcnPdlim1Rgs18Serpinb2

Plin2

(b)

3 350

122

0 39

9

37

Cluster 3 GSE15041

GSE18803

C1qbFcgr2bC3Aif1Reg3b

Atf3CtssTspoLyz2

(c)

Figure 6: The intersection of cluster genes with differentially expressed genes obtained from GSE15041 and GSE18803 databases. (a) Theintersection of cluster 1 genes and downregulated genes obtained from GSE15041 and GSE18803 databases. (b) The intersection of cluster2 genes and upregulated genes obtained from GSE15041 and GSE18803 databases. (c) The intersection of cluster 3 genes and upregulatedgenes obtained from GSE15041 and GSE18803 databases.

9Neural Plasticity

Page 10: Analysis of Crucial Genes and Pathways Associated with ...downloads.hindawi.com/journals/np/2020/8822001.pdfSep 25, 2020  · different change trends over time after SNI were obtained

consequently resulting in neuropathic pain development byregulating proinflammatory cytokine production [34–36].Moreover, the p38MAPK pathway is considered a promisingtherapeutic target for neuropathic pain [37]. Here, theMAPK signaling pathway was simultaneously enriched bycluster genes in different clusters, confirming the key role ofthis pathway in SNI-induced neuropathic pain. Moreover,Prkacb and Fgf9were significantly enriched in theMAPK sig-naling pathway. Prkacb is a subunit of the cAMP-dependentprotein kinase, which has a catalytic role in numerous cellu-lar processes like cell proliferation, gene transcription, anddifferentiation [38]. Fgf9 is a member of the highly conservedFGF family, which is reportedly important for glial cell devel-opment in the nervous system [39]. Fgf9 silencing recapitu-lates the inhibitory effect of miR-182 overexpression onSchwann cell proliferation at an early stage following SNI[40], suggesting the potential role of Fgf9 after SNI. Althoughthe key role of Prkacb and Fgf9 in neuropathic pain has notbeen fully disclosed, we speculate that Prkacb and Fgf9 maybe implicated in SNI-induced neuropathic pain in rats bybeing involved in the MAPK signaling pathway.

In addition to Fgf9, other important genes, like Nrg1 incluster 1, Timp1 in cluster 2, and Atf3 and C3 in cluster 3,were also found to be differentially expressed in SNI samplesafter corroborating microarray data with two other microar-ray datasets. Nrg1 reportedly plays a pivotal role in neuraldevelopment and plasticity [41]. Wang et al. demonstratedthat Nrg1 upregulation reversed the signs of SNI-inducedneuropathic pain in rats, and modulating Nrg1might exhibittherapeutic value for neuropathic pain treatment [42].Timp1 is well known as an inhibitor of matrix metallopro-teinases (MMPs) that are widely involved in pain develop-ment following a variety of injury and inflammatoryconditions [43, 44]. Knight et al. revealed that Timp1 playeda crucial role in pathological pain states associated withinflammation [45]. Moreover, Timp1 is upregulated inDRG and spinal cord tissues after tibial nerve transection,and it may be related to neuropathic pain following periph-eral nerve injury [46]. A recent microarray analysis alsoshowed that hub genes like C3 and Timp1 are closely relatedto SNI-induced neuropathic pain development, providingthe theoretical basis for treatment of this disease [14]. Inaddition, ATF3 has been shown to be implicated in theBortezomib-induced painful peripheral neuropathy [47].Atf3 expression was found to be significantly increased afterSNI operation, and it may be a key regulator in neuropathicpain development [48], which agrees with our bioinformaticresults. Based on our results, we speculate that these genesmay be involved in neuropathic pain development and couldserve as the therapeutic targets of this disease. Furthermore,rno-miR-3583-5p was found to be targeted by Fgf9, andrno-miR-1912-3p was targeted by Nrg1. However, there arefew reports about the roles of rno-miR-3583-5p and rno-miR-1912-3p in neuropathic pain. Given the potential roleof Fgf9 and Nrg1, we speculate that rno-miR-3583-5p andrno-miR-1912-3p may also contribute to neuropathic paindevelopment in rats.

However, there are some limitations in this study. First,the sample size was small which influences the sample power

stability. Second, we only used other microarray data to cor-roborate the identified key genes; no experimental valida-tions were conducted to confirm our findings. In the future,more studies with large samples and experimental valida-tions are needed to verify the role of critical genes and path-ways in neuropathic pain development.

5. Conclusions

Our results reveal that key pathways like the MAPK signalingpathway and crucial genes like Prkacb,Nrg1, Fgf9, Timp1, C3,and Atf3may be implicated in SNI-induced neuropathic paindevelopment in rats. Our findings will provide new insightfor designing effective neuropathic pain therapies.

Data Availability

The data used to support the findings of this study areincluded within the article.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Authors’ Contributions

Dong Mao and Huang Zhai participated in the data acquisi-tion and performed analysis and data interpretation. GangZhao and Jingyi Mi designed the research and drafted themanuscript. Yongjun Rui reviewed the manuscript. Allauthors approved the final version to be published.

Acknowledgments

This work was supported by grants from the ScientificResearch Project Funded by the Health Commission of WuxiCity (MS201825) and Shanghai Key Laboratory of PeripheralNerve and Microsurgery (17DZ2270500).

References

[1] I. Gilron, R. Baron, and T. Jensen, “Neuropathic pain: princi-ples of diagnosis and treatment,” Mayo Clinic Proceedings,vol. 90, no. 4, pp. 532–545, 2015.

[2] L. Colloca, T. Ludman, D. Bouhassira et al., “Neuropathicpain,” Nature reviews Disease primers, vol. 3, no. 1, pp. 1–19,2017.

[3] N. B. Finnerup, S. Haroutounian, P. Kamerman et al., “Neuro-pathic pain,” Pain, vol. 157, no. 8, pp. 1599–1606, 2016.

[4] D. Bouhassira and N. Attal, “Translational neuropathic painresearch: a clinical perspective,” Neuroscience, vol. 338,pp. 27–35, 2016.

[5] R. H. Dworkin, A. B. O’Connor, M. Backonja et al., “Pharma-cologic management of neuropathic pain: evidence-based rec-ommendations,” Pain, vol. 132, no. 3, pp. 237–251, 2007.

[6] N. Attal, M. Lanteri-Minet, B. Laurent, J. Fermanian, andD. Bouhassira, “The specific disease burden of neuropathicpain: results of a French nationwide survey,” Pain, vol. 152,no. 12, pp. 2836–2843, 2011.

[7] D. Schomberg, G. Miranpuri, T. Duellman, A. Crowell,R. Vemuganti, and D. Resnick, “Spinal cord injury induced

10 Neural Plasticity

Page 11: Analysis of Crucial Genes and Pathways Associated with ...downloads.hindawi.com/journals/np/2020/8822001.pdfSep 25, 2020  · different change trends over time after SNI were obtained

neuropathic pain: molecular targets and therapeuticapproaches,” Metabolic Brain Disease, vol. 30, no. 3, pp. 645–658, 2015.

[8] A. Sanchez-Palencia, M. Gomez-Morales, J. A. Gomez-Capillaet al., “Gene expression profiling reveals novel biomarkers innonsmall cell lung cancer,” International Journal of Cancer,vol. 129, no. 2, pp. 355–364, 2011.

[9] S. Boccella, F. Guida, E. Palazzo et al., “Spared nerve injury as along-lasting model of neuropathic pain,” in Neurotrophic Fac-torspp. 373–378, Springer2018.

[10] H. Li, H. Q. Wan, H. J. Zhao, S. X. Luan, and C. G. Zhang,“Identification of candidate genes and miRNAs associatedwith neuropathic pain induced by spared nerve injury,” Inter-national Journal of Molecular Medicine, vol. 44, no. 4,pp. 1205–1218, 2019.

[11] X. He, L. Fan, Z. Wu, J. He, and B. Cheng, “Gene expressionprofiles reveal key pathways and genes associated with neuro-pathic pain in patients with spinal cord injury,” MolecularMedicine Reports, vol. 15, no. 4, pp. 2120–2128, 2017.

[12] M. Costigan, I. Belfer, R. S. Griffin et al., “Multiple chronic painstates are associated with a common amino acid-changingallele in KCNS1,” Brain, vol. 133, no. 9, pp. 2519–2527, 2010.

[13] C. Zhang, H. Wan, K. Ma, S. Luan, and H. Li, “Identification ofbiomarkers related to neuropathic pain induced by peripheralnerve injury,” Journal of Molecular Neuroscience, vol. 69, no. 4,pp. 505–515, 2019.

[14] K. Wang, D. Yi, Z. Yu, B. Zhu, S. Li, and X. Liu, “Identificationof the hub genes related to nerve injury-induced neuropathicpain,” Frontiers in Neuroscience, vol. 14, p. 488, 2020.

[15] T. Barrett, S. E. Wilhite, P. Ledoux et al., “NCBI GEO: archivefor functional genomics data sets—update,” Nucleic AcidsResearch, vol. 41, no. D1, pp. D991–D995, 2012.

[16] L. Kumar and M. E. Futschik, “Mfuzz: a software package forsoft clustering of microarray data,” Bioinformation, vol. 2,no. 1, pp. 5–7, 2007.

[17] Y. Zhou, B. Zhou, L. Pache et al., “Metascape provides abiologist-oriented resource for the analysis of systems-leveldatasets,” Nature Communications, vol. 10, no. 1, 2019.

[18] M. Ashburner, C. A. Ball, J. A. Blake et al., “Gene ontology: toolfor the unification of biology,” Nature Genetics, vol. 25, no. 1,pp. 25–29, 2000.

[19] M. Kanehisa and S. Goto, “KEGG: Kyoto Encyclopedia ofGenes and Genomes,” Nucleic Acids Research, vol. 28, no. 1,pp. 27–30, 2000.

[20] P. Shannon, A. Markiel, O. Ozier et al., “Cytoscape: a softwareenvironment for integrated models of biomolecular interac-tion networks,” Genome Research, vol. 13, no. 11, pp. 2498–2504, 2003.

[21] C. von Mering, M. Huynen, D. Jaeggi, S. Schmidt, P. Bork, andB. Snel, “STRING: a database of predicted functional associa-tions between proteins,” Nucleic Acids Research, vol. 31,no. 1, pp. 258–261, 2003.

[22] Y. Tang, M. Li, J. Wang, Y. Pan, and F.-X. Wu, “CytoNCA: acytoscape plugin for centrality analysis and evaluation of pro-tein interaction networks,” BioSystems, vol. 127, pp. 67–72,2015.

[23] G. D. Bader and C. W. Hogue, “An automated method forfinding molecular complexes in large protein interaction net-works,” BMC Bioinformatics, vol. 4, no. 1, p. 2, 2003.

[24] G. Yu, L.-G. Wang, Y. Han, and Q.-Y. He, “clusterProfiler: anR package for comparing biological themes among gene clus-

ters,” Omics: a journal of integrative biology, vol. 16, no. 5,pp. 284–287, 2012.

[25] Y. Benjamini and Y. Hochberg, “Controlling the false discov-ery rate: a practical and powerful approach to multiple test-ing,” Journal of the Royal Statistical Society: Series B(Methodological), vol. 57, no. 1, pp. 289–300, 1995.

[26] J. Wang, D. Duncan, Z. Shi, and B. Zhang, “WEB-based geneset analysis toolkit (WebGestalt): update 2013,” Nucleic AcidsResearch, vol. 41, no. W1, pp. W77–W83, 2013.

[27] H. Dweep and N. Gretz, “miRWalk2.0: a comprehensive atlasof microRNA-target interactions,” Nature Methods, vol. 12,no. 8, pp. 697–697, 2015.

[28] D. Vega-Avelaira, S. M. Géranton, and M. Fitzgerald, “Differ-ential regulation of immune responses and macrophage/n-euron interactions in the dorsal root ganglion in young andadult rats following nerve injury,” Molecular Pain, vol. 5,pp. 1744-8069-5-70–1744-8069-8069, 2009.

[29] M. Costigan, A. Moss, A. Latremoliere et al., “T-cell infiltra-tion and signaling in the adult dorsal spinal cord is a majorcontributor to neuropathic pain-like hypersensitivity,” TheJournal of Neuroscience, vol. 29, no. 46, pp. 14415–14422,2009.

[30] H. Liu, T. Xia, F. Xu, Z. Ma, and X. Gu, “Identification of thekey genes associated with neuropathic pain,” Molecular Medi-cine Reports, vol. 17, no. 5, pp. 6371–6378, 2018.

[31] Y. Yang, X. Lu, Y.Wang, M. Yang, and D. Jiang, “Identificationcrucial genes in peripheral neuropathic pain induced by sparednerve injury,” European Review for Medical and Pharmacolog-ical Sciences, vol. 18, no. 15, pp. 2152–2159, 2014.

[32] J. Wang, S.-H. Ma, R. Tao, L.-J. Xia, L. Liu, and Y.-H. Jiang,“Gene expression profile changes in rat dorsal horn after sci-atic nerve injury,” Neurological Research, vol. 39, no. 2,pp. 176–182, 2017.

[33] R. Chen, C. Yin, Q. Hu et al., “Expression profiling of spinalcord dorsal horn in a rat model of complex regional pain syn-drome type-I uncovers potential mechanisms mediating painand neuroinflammation responses,” Journal of Neuroinflam-mation, vol. 17, no. 1, 2020.

[34] K. Obata, H. Yamanaka, K. Kobayashi et al., “Role of mitogen-activated protein kinase activation in injured and intact pri-mary afferent neurons for mechanical and heat hypersensitiv-ity after spinal nerve ligation,” Journal of Neuroscience, vol. 24,no. 45, pp. 10211–10222, 2004.

[35] L. Xu, Y. Huang, X. Yu, J. Yue, N. Yang, and P. Zuo, “The influ-ence of p38 mitogen-activated protein kinase inhibitor on syn-thesis of inflammatory cytokine tumor necrosis factor alpha inspinal cord of rats with chronic constriction injury,” Anesthe-sia & Analgesia, vol. 105, no. 6, pp. 1838–1844, 2007.

[36] J. Zhou, L. Wang, J. Wang et al., “Paeoniflorin and albiflorinattenuate neuropathic pain via MAPK pathway in chronicconstriction injury rats,” Evidence-Based Complementary andAlternative Medicine, vol. 2016, 11 pages, 2016.

[37] E. Rojewska, K. Popiolek-Barczyk, A. M. Jurga, W. Makuch,B. Przewlocka, and J. Mika, “Involvement of pro- and antino-ciceptive factors in minocycline analgesia in rat neuropathicpain model,” Journal of Neuroimmunology, vol. 277, no. 1-2,pp. 57–66, 2014.

[38] Y. Chen, Y. Gao, Y. Tian, and D. L. Tian, “PRKACB is down-regulated in non-small cell lung cancer and exogenousPRKACB inhibits proliferation and invasion of LTEP-A2cells,” Oncology Letters, vol. 5, no. 6, pp. 1803–1808, 2013.

11Neural Plasticity

Page 12: Analysis of Crucial Genes and Pathways Associated with ...downloads.hindawi.com/journals/np/2020/8822001.pdfSep 25, 2020  · different change trends over time after SNI were obtained

[39] M. Lum, A. Turbic, B. Mitrovic, and A.M. Turnley, “Fibroblastgrowth factor-9 inhibits astrocyte differentiation of adultmouse neural progenitor cells,” Journal of NeuroscienceResearch, vol. 87, no. 10, pp. 2201–2210, 2009.

[40] B. Yu, T. Qian, Y. Wang et al., “miR-182 inhibits Schwann cellproliferation and migration by targeting FGF9 and NTM,respectively at an early stage following sciatic nerve injury,”Nucleic Acids Research, vol. 40, no. 20, pp. 10356–10365, 2012.

[41] M. J. Kipanyula, W. H. Kimaro, F. N. Yepnjio et al., “Signalingpathways bridging fate determination of neural crest cells toglial lineages in the developing peripheral nervous system,”Cellular Signalling, vol. 26, no. 4, pp. 673–682, 2014.

[42] G. Wang, D. Dai, X. Chen et al., “Upregulation of neuregulin-1reverses signs of neuropathic pain in rats,” International Jour-nal of Clinical and Experimental Pathology, vol. 7, no. 9,pp. 5916–5921, 2014.

[43] Y. Kawasaki, Z.-Z. Xu, X. Wang et al., “Distinct roles of matrixmetalloproteases in the early- and late-phase development ofneuropathic pain,” Nature Medicine, vol. 14, no. 3, pp. 331–336, 2008.

[44] A. G. Remacle, S. K. Hullugundi, J. Dolkas et al., “Acute- andlate-phase matrix metalloproteinase (MMP)-9 activity is com-parable in female and male rats after peripheral nerve injury,”Journal of Neuroinflammation, vol. 15, no. 1, p. 89, 2018.

[45] B. E. Knight, N. Kozlowski, J. Havelin et al., “TIMP-1 attenu-ates the development of inflammatory pain through MMP-dependent and receptor-mediated cell signaling mechanisms,”Frontiers in Molecular Neuroscience, vol. 12, 2019.

[46] N. Cheng, Z. Zhang, Y. Guo et al., “Weighted gene co-expression network analysis reveals specific modules and hubgenes related to neuropathic pain in dorsal root ganglions,”Bioscience Reports, vol. 39, no. 11, 2019.

[47] C. Liu, S. Luan, H. Ouyang et al., “Upregulation of CCL2 viaATF3/c-Jun interaction mediated the Bortezomib-inducedperipheral neuropathy,” Brain, Behavior, and Immunity,vol. 53, pp. 96–104, 2016.

[48] S. Tang, H. Jing, Z. Huang et al., “Identification of key candi-date genes in neuropathic pain by integrated bioinformaticanalysis,” Journal of Cellular Biochemistry, vol. 121, no. 2,pp. 1635–1648, 2019.

12 Neural Plasticity