hdbmr 2073690 1.downloads.hindawi.com/journals/bmri/2020/2073690.pdf2.4. protein-protein interaction...

10
Research Article Identification of Potential Biomarkers Associated with Basal Cell Carcinoma Yong Liu , Hui Liu, and Queqiao Bian Department of Dermatology & STD, The Third Central Hospital of Tianjin, Tianjin Key Laboratory of Articial Cell, Articial Cell Engineering Technology Research Center of Public Health Ministry, Tianjin Institute of Hepatobiliary Disease, Tianjin 300170, China Correspondence should be addressed to Yong Liu; [email protected] Received 18 October 2019; Revised 2 February 2020; Accepted 26 March 2020; Published 20 April 2020 Academic Editor: Lei Yao Copyright © 2020 Yong Liu 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. This work is aimed at identifying several molecular markers correlated with the diagnosis and development of basal cell carcinoma (BCC). Methods. The available microarray datasets for BCC were obtained from the Gene Expression Omnibus (GEO) database, and dierentially expressed genes (DEGs) were identied between BCC and healthy controls. Afterward, the functional enrichment analysis and protein-protein interaction (PPI) network analysis of these screened DEGs were performed. An external validation for the DEG expression level was also carried out, and receiver operating characteristic curve analysis was used to evaluate the diagnostic values of DEGs. Result. In total, ve microarray datasets for BCC were downloaded and 804 DEGs (414 upregulated and 390 downregulated genes) were identied. Functional enrichment analysis showed that these genes including CYFIP2, HOXB5, EGFR, FOXN3, PTPN3, CDC20, MARCKSL1, FAS, and PTCH1 were closely correlated with the cell process and PTCH1 played central roles in the BCC signaling pathway. Moreover, EGFR was a hub gene in the PPI network. The expression changes of six genes (CYFIP2, HOXB5, FOXN3, PTPN3, MARCKSL1, and FAS) were validated by an external GSE74858 dataset analysis. Finally, ROC analysis revealed that CYFIP2, HOXB5, PTPN3, MARCKSL1, PTCH1, and CDC20 could distinguish BCC and healthy individuals. Conclusion. Nine gene signatures (CYFIP2, HOXB5, EGFR, FOXN3, PTPN3, CDC20, MARCKSL1, FAS, and PTCH1) may serve as promising targets for BCC detection and development. 1. Introduction Basal cell carcinoma (BCC) is one of common malignant epi- thelial neoplasms that derive from basal cells and accounts for nearly 75% of skin cancers [1]. It was reported that the BCC incidence rates have been increasing and roughly reached 2.75 million cases around the world [2]. BCC was generally categorized into several types such as supercial, nodular, and nevoid BCC on the basis of its histological and clinical characteristics. Notably, the majority of lesions for BCC occurred on the head and neck [3]. The convincing evidence has suggested that gender, age, sunlight exposure (especially ultraviolet radiation), smoking, chemicals, and fair skin were all BCC risk factors [4, 5]. Although BCC recurrence was extremely frequent due to local tissue destruction, it rarely metastasizes or leads to death [6]. The imiquimod application, surgical excision, and radiation ther- apy have been unsatisfactory for BCC treatment. Therefore, it is imperative to develop promising strategies for early diag- nosis and intervention of BCC. Fortunately, the next-generation high-throughput sequenc- ing provides fascinating opportunities for bioinformatics anal- ysis. A growing number of studies have demonstrated that gene expression proling could eectively screen molecular makers and predict pathogenesis for tumorigenesis in the last decades, thereby oering novel therapeutic strategies for can- cer treatment [7]. Fang et al. previously found that the expres- sion level of EGR1 (early growth response 1) was decreased in BCC based on a microarray analysis. Furthermore, they also noted that EGR1 might suppress epidermal proliferation via modulating CDC25A that is critical for cell cycle progression from G1 to S phase [8]. Heller et al. conducted a global gene expression analysis and identied BCC-associated dieren- tially expressed gene (DEG) targets, providing several new diagnostic signatures for the susceptibility of BCC [9]. Later, Yunoki et al. identied three BCC-correlated genes including Hindawi BioMed Research International Volume 2020, Article ID 2073690, 10 pages https://doi.org/10.1155/2020/2073690

Upload: others

Post on 10-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: HDBMR 2073690 1.downloads.hindawi.com/journals/bmri/2020/2073690.pdf2.4. Protein-Protein Interaction (PPI) Network. The Biologi-cal General Repository for Interaction Datasets (BioGRID)

Research ArticleIdentification of Potential Biomarkers Associated with BasalCell Carcinoma

Yong Liu , Hui Liu, and Queqiao Bian

Department of Dermatology & STD, The Third Central Hospital of Tianjin, Tianjin Key Laboratory of Artificial Cell, Artificial CellEngineering Technology Research Center of Public HealthMinistry, Tianjin Institute of Hepatobiliary Disease, Tianjin 300170, China

Correspondence should be addressed to Yong Liu; [email protected]

Received 18 October 2019; Revised 2 February 2020; Accepted 26 March 2020; Published 20 April 2020

Academic Editor: Lei Yao

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

Purpose. This work is aimed at identifying several molecular markers correlated with the diagnosis and development of basal cellcarcinoma (BCC). Methods. The available microarray datasets for BCC were obtained from the Gene Expression Omnibus(GEO) database, and differentially expressed genes (DEGs) were identified between BCC and healthy controls. Afterward, thefunctional enrichment analysis and protein-protein interaction (PPI) network analysis of these screened DEGs were performed.An external validation for the DEG expression level was also carried out, and receiver operating characteristic curve analysis wasused to evaluate the diagnostic values of DEGs. Result. In total, five microarray datasets for BCC were downloaded and 804DEGs (414 upregulated and 390 downregulated genes) were identified. Functional enrichment analysis showed that these genesincluding CYFIP2, HOXB5, EGFR, FOXN3, PTPN3, CDC20, MARCKSL1, FAS, and PTCH1 were closely correlated with the cellprocess and PTCH1 played central roles in the BCC signaling pathway. Moreover, EGFR was a hub gene in the PPI network.The expression changes of six genes (CYFIP2, HOXB5, FOXN3, PTPN3, MARCKSL1, and FAS) were validated by an externalGSE74858 dataset analysis. Finally, ROC analysis revealed that CYFIP2, HOXB5, PTPN3, MARCKSL1, PTCH1, and CDC20could distinguish BCC and healthy individuals. Conclusion. Nine gene signatures (CYFIP2, HOXB5, EGFR, FOXN3, PTPN3,CDC20, MARCKSL1, FAS, and PTCH1) may serve as promising targets for BCC detection and development.

1. Introduction

Basal cell carcinoma (BCC) is one of common malignant epi-thelial neoplasms that derive from basal cells and accountsfor nearly 75% of skin cancers [1]. It was reported that theBCC incidence rates have been increasing and roughlyreached 2.75 million cases around the world [2]. BCC wasgenerally categorized into several types such as superficial,nodular, and nevoid BCC on the basis of its histologicaland clinical characteristics. Notably, the majority of lesionsfor BCC occurred on the head and neck [3]. The convincingevidence has suggested that gender, age, sunlight exposure(especially ultraviolet radiation), smoking, chemicals, andfair skin were all BCC risk factors [4, 5]. Although BCCrecurrence was extremely frequent due to local tissuedestruction, it rarely metastasizes or leads to death [6]. Theimiquimod application, surgical excision, and radiation ther-apy have been unsatisfactory for BCC treatment. Therefore,

it is imperative to develop promising strategies for early diag-nosis and intervention of BCC.

Fortunately, the next-generation high-throughput sequenc-ing provides fascinating opportunities for bioinformatics anal-ysis. A growing number of studies have demonstrated thatgene expression profiling could effectively screen molecularmakers and predict pathogenesis for tumorigenesis in the lastdecades, thereby offering novel therapeutic strategies for can-cer treatment [7]. Fang et al. previously found that the expres-sion level of EGR1 (early growth response 1) was decreased inBCC based on a microarray analysis. Furthermore, they alsonoted that EGR1 might suppress epidermal proliferation viamodulating CDC25A that is critical for cell cycle progressionfrom G1 to S phase [8]. Heller et al. conducted a global geneexpression analysis and identified BCC-associated differen-tially expressed gene (DEG) targets, providing several newdiagnostic signatures for the susceptibility of BCC [9]. Later,Yunoki et al. identified three BCC-correlated genes including

HindawiBioMed Research InternationalVolume 2020, Article ID 2073690, 10 pageshttps://doi.org/10.1155/2020/2073690

Page 2: HDBMR 2073690 1.downloads.hindawi.com/journals/bmri/2020/2073690.pdf2.4. Protein-Protein Interaction (PPI) Network. The Biologi-cal General Repository for Interaction Datasets (BioGRID)

BCL2 (B-cell lymphoma 2), PTCH1 (Patched 1), and SOX9(SRY-box 9) and unique gene networks involved with tumor-igenesis through a microarray analysis [10]. Additionally, Jeeet al. classified BCC into squamous cell carcinoma- (SCC-)like BCC, normal-like BCC, and classical BCC subtypes by acomparative analysis of gene expression profiles from BCC,SCC, and normal skin tissues. Moreover, functional analysisrevealed that hedgehog signaling pathways were possiblyrelated to classical BCC development [11]. Although numer-ous investigations have extracted several BCC-related genemakers, a deeper insight into the underlying molecular mech-anisms of BCC has not been completely achieved.

In this study, the eligible mRNA microarray datasetsfrom BCC tissues were retrieved and downloaded from theNational Center for Biotechnology Information GeneExpression Omnibus (GEO) database. Then, the DEGsbetween BCC and normal tissues were screened followed byfunctional enrichment analyses. The protein-protein (PPI)network was also constructed to explore underlying interac-tions among key genes. Finally, the expression levels of DEGswere further examined by an external dataset, and diagnosticperformance of these genes were also evaluated. This workprovided novel gene signatures for BCC diagnosis and pro-moted a deeper understanding for BCC progression.

2. Materials and Methods

2.1. Data Acquisition. The publicly accessible microarraydatasets in BCC were searched and downloaded from theNCBI-GEO [12] database (https://www.ncbi.nlm.nih.gov/geo/). The keyword terms of carcinoma, basal cell (MeSHTerms) OR basal cell carcinoma (All Fields) AND “Homosapiens” (porgn) AND “gse” (Filter) were used for precisesearching. The inclusive criteria for datasets were as follows:(i) the datasets were gene expression profiles, (ii) the datawas obtained from tumor tissues of patients with BCC ornormal skin tissues from healthy individuals (normal con-trols), and (iii) the patients did not receive medication orother treatments. Finally, five datasets (GSE103439,GSE53462, GSE42109, GSE39612, and GSE7553) were cho-sen according to the abovementioned screening criteria asshown in Table 1. Of these, four datasets (GSE103439,GSE42109, GSE39612, and GSE7553) were generated by the

GPL570 [HG-U133_Plus_2] Affymetrix Human GenomeU133 Plus 2.0 Array platform, and the platform forGSE53462 was GPL10558 Illumina Human HT-12 V4.0expression beadchip. These datasets would be used for thefollowing integrated analyses, which consisted of 48 BCC tis-sues and 85 normal tissue samples.

2.2. Identification of Differentially Expressed Genes (DEGs). Ameta-analysis of five microarray datasets from different plat-forms was carried out according to the guidelines providedby Ramasamy et al. [13]. The preprocessing of raw dataacross different platforms, mainly including background cor-rection, normalization, log2 transformation, and gene expres-sion calculation, was performed. The overlapped genes in fivedatasets were selected. Then, the R metaMA package wasused to calculate effect sizes from unpaired data by eitherclassical or moderated t-tests (Limma) for each study andcombine these effect sizes [14]. The DEGs between BCCand normal controls were screened according to the thresh-old of the multiple comparison correction false discoveryrate ðFDRÞ < 0:05. The whole R sentences for data processingand identification of DEGs in this study were shown in Sup-plementary Materials (available here). Finally, the hierarchi-cal clustering analysis of identified DEGs was performed bythe pheatmap package (https://cran.r-project.org/package=pheatmap) in R language.

2.3. Gene Ontology (GO) and Pathway Enrichment Analyses.To explore the possible biological roles of the DEGs, thefunctional analyses were conducted. Firstly, the GO analysisinvolving three categories of molecular function (MF), cellu-lar component (CC), and biological process (BP) was con-ducted with BiNGO plugin in cytoscape software [15]. Theparameters were set as follows: (i) hypergeometric test wasused as the selected statistical test; (ii) Benjamini and Hoch-berg FDR correction was set as selected correction; (iii)selected significance level was 0.05; and (iv) for testingoption, we used whole annotation as the reference set. Inaddition, the Kyoto Encyclopedia of Genes and Genomes(KEGG) pathway analysis was undertaken using KOBAS(http://kobas.cbi.pku.edu.cn/index.php), which is a web-accessible tool and could identify enriched pathways for aninput set of genes by mapping to genes using known

Table 1: The microarray datasets for basal cell carcinoma.

GEO accession Control Case Platform Year Country Author

GSE103439 2 4GPL570 [HG-U133_Plus_2] AffymetrixHuman Genome U133 Plus 2.0 Array

2017 Japan Yoshiaki Tabuchi

GSE53462 5 16GPL10558 Illumina Human HT-12

V4.0 expression beadchip2014 South Korea Hyun Goo Woo

GSE42109 10 11GPL570 [HG-U133_Plus_2] Affymetrix Human

Genome U133 Plus 2.0 Array; GPL571 [HG-U133A_2]Affymetrix Human Genome U133A 2.0 Array

2013 USA Mayte Suarez-Farinas

GSE39612 64 2GPL570 [HG-U133_Plus_2] Affymetrix Human

Genome U133 Plus 2.0 Array2012 USA Paul Harms

GSE7553 4 15GPL570 [HG-U133_Plus_2] Affymetrix Human

Genome U133 Plus 2.0 Array2008 USA Sean Yoder

GEO: Gene Expression Omnibus.

2 BioMed Research International

Page 3: HDBMR 2073690 1.downloads.hindawi.com/journals/bmri/2020/2073690.pdf2.4. Protein-Protein Interaction (PPI) Network. The Biologi-cal General Repository for Interaction Datasets (BioGRID)

pathways in the KEGG database [16]. Moreover, the statisti-cally significantly enriched pathways were selected with thecutoff of P value < 0.05.

2.4. Protein-Protein Interaction (PPI) Network. The Biologi-cal General Repository for Interaction Datasets (BioGRID)database is available online and provides 1,728,498 biologicalPPI interactions by August 2019. Here, we performed a PPIanalysis for screened DEGs based on this database to identifysignificant protein pairs. The cytoscape software (http://apps.cytoscape.org/apps/cytonca) was utilized to construct thePPI network. Moreover, the CytoNCA [17] (http://apps.cytoscape.org/apps/cytonca), a cytoscape plugin, was usedto analyze topological characteristics of PPI nodes using thewithout weight parameter. Several metrics such as DegreeCentrality (DC), Betweenness centrality (BC), and Closenesscentrality (CC) were estimated. In this study, hub protein wasdetermined on the basis of high scores. In addition, theMCODE plugin (http://apps.cytoscape.org/apps/mcode) incytoscape software was employed to further identify severalPPI submodules using the default parameters of degreecutoff = 2, node score cutoff = 0:2, K‐core = 2, and max:depth = 100 [18]. Notably, the PPI score > 3 was consideredas the cutoff for screening significantly enriched functionalmodules from the PPI network.

2.5. Validation of DEG Expression Levels by a NoncodingRNA Expression Profile. The GSE74858 dataset, including3 BCC patients and 3 healthy individuals, was obtainedfrom the GEO database and used as a verification datasetto evaluate the expression levels of DEGs. Furthermore,the receiver operating characteristic (ROC) analysis wasalso carried out to assess the performance of DEGs in thisresearch using the “pROC” package (https://cran.r-project.org/web/packages/pROC/index.html) in R language. Thearea under the ROC curve (AUC) was then computed.Herein, if AUC values of the genes were greater than0.9, these genes were considered to discriminate patientsundergoing BCC from healthy controls with high specific-ity and sensitivity.

3. Results

3.1. DEG Screening. The meta-analysis of five gene expressionprofiles was conducted, and the DEGs were identified. Con-sequently, a total of 804 DEGs were extracted between BCCand normal controls according to screening criteriadescribed in Materials and Methods. Of these, there were414 upregulated genes and 390 downregulated genes inpatients with BCC. Moreover, the clustering analysis of theseDEGs indicated that they could clearly distinguish BCC andhealthy controls as exhibited in Figure 1, suggesting thatthe differential gene expression was possibly responsible forBCC occurrence.

3.2. Functional Enrichment Analyses of DEGs. As shown inTable 2, the GO functional annotation analysis showed thatidentified DEGs were enriched in 53 GO-BP terms includingcellular component organization, cell cycle, cell proliferation,and ectoderm development. Meanwhile, they were focused

on 54 GO-CC terms such as intracellular part, intracellu-lar, and cytoplasm. It was also observed that there werethree GO-MF terms (protein binding, binding, and ribo-nuclease H activity). Notably, numerous genes were closelyassociated with the cellular process, including CYFIP2(cytoplasmic FMR1 interacting protein 2; upregulated),HOXB5 (homeobox B5; downregulated), EGFR (epidermalgrowth factor receptor; downregulated), FOXN3 (forkheadbox N3; upregulated), PTPN3 (protein tyrosine phospha-tase nonreceptor type 3; downregulated), CDC20 (cell divi-sion cycle 20; upregulated), MARCKSL1 (myristoylatedalanine rich protein kinase C substrate like 1; upregulated),FAS (fas cell surface death receptor; downregulated), andupregulated PTCH1. More specifically, FOXN3, CDC20,and EGFR played essential roles in the cell cycle process,and PTCH1 and EGFR were involved in cell proliferation.Additionally, KEGG pathway enrichment analysis revealedthat these genes were primarily enriched in pathways incancer, p53 signaling pathway and BCC pathway. We alsonoted that PTCH1 exerted critical biological roles in theBCC pathway (Figure 2).

3.3. PPI Analysis. PPI network analysis was conducted toexplore the underlying interactions of DEGs. In total, 549gene nodes (296 upregulated genes and 254 downregulatedgenes) and 1462 protein pairs were obtained in the PPI net-work (Figure 3(a)). The top 5 gene nodes with a high degreewere EGLN3 (Egl-9 family hypoxia inducible factor 3;degree = 82), XPO1 (exportin 1; degree = 81), EGFR(degree = 77), CFTR (CF transmembrane conductance regu-lator; degree = 48), and MCM2 (minichromosome mainte-nance complex component 2; degree = 47), and they wereregarded as hub genes. Furthermore, the PPI submoduleanalysis suggested that 3 significantly enriched submodules

Label

LabelNormal skinBasal cell carcinoma

−3

−2

−1

0

1

2

3

Figure 1: The heatmap of differentially expressed genes in basal cellcarcinoma. The “label” represents sample type (basal cell carcinomaand normal skin samples). The light red color shows the basal cellcarcinoma samples, and light blue color shows normal skin samples.

3BioMed Research International

Page 4: HDBMR 2073690 1.downloads.hindawi.com/journals/bmri/2020/2073690.pdf2.4. Protein-Protein Interaction (PPI) Network. The Biologi-cal General Repository for Interaction Datasets (BioGRID)

were identified based on the PPI score > 3 (Figures 3(b)–3(d)). Interestingly, we found that the genes in submodule1 were all upregulated.

3.4. Verification of DEG Expression Levels and ROCAnalysis. A noncoding RNA profile GSE74858 was down-loaded from the GEO repository, and there were 145 over-lapped genes between this dataset and the integrateddatasets. We found that 25 overlapped genes exhibited sig-nificant differential expression, and their expression trendswere also consistent with our integration analysis. More nota-bly, six genes (CYFIP2, HOXB5, FOXN3, PTPN3,MARCKSL1,and FAS) were predominately correlated with the cellular bio-logical process, and their expression patterns were validatedby an external GSE74858 dataset (Figure 4). Besides, the ROCcurve analysis was conducted to assess the diagnostic values ofDEGs in BCC by fivemicroarray datasets. There were 439 genesaccording to AUC > 0:9, and 129 genes were overlapped withDEGs by integrated analysis and then extracted. Our findingsrevealed that CYFIP2 (AUC = 0:949), HOXB5 (AUC = 0:908),PTPN3 (AUC = 0:952), MARCKSL1 (AUC = 0:962), PTCH1(AUC = 0:981), and CDC20 (AUC = 0:956) could significantlydistinguish BCC samples and healthy controls, implying thatthese genes might serve as diagnostic makers for BCC detec-tion (Figure 5).

4. Discussion

In the present study, we performed an integrated bioinfor-matics analysis using five microarray datasets about BCC.In total, 804 DEGs (414 upregulated and 390 downregu-lated genes) were screened between BCC and normal con-trol tissues. The GO functional analysis showed thatseveral genes, including CYFIP2, HOXB5, EGFR, FOXN3,PTPN3, CDC20, MARCKSL1, FAS, and PTCH1, primarilyplayed pivotal roles in cellular processes. Moreover, theexpression patterns of six genes (CYFIP2, HOXB5, FOXN3,PTPN3, MARCKSL1, and FAS) were validated in an exter-nal noncoding RNA dataset. Additionally, CYFIP2, HOXB5,PTPN3, MARCKSL, CDC20, and PTCH1 had superior diag-nostic values for BCC prediction.

Overwhelming evidence has suggested that the patchedgene (PTCH) family played vital roles in BCC inductionand progression [19, 20]. A previous study highlighted thatPTCH could encode a receptor for the hedgehog signalingpathway which was important for vertebrate developmentand tumorigenesis [21]. Undén et al. found that the expres-sion level of PTCH mRNA was overexpressed in BCC cellscompared with nontumor epidermal cells, which was similarto our finding that PTCH1 was upregulated in BCC patients[22]. Interestingly, numerous researchers pointed out thatapproximately 90% of function mutations in PTCH1 were

Table 2: The functional analyses of differentially expressed genes in BCC.

Category Term Count P value Genes

GOTERM_BP (top 5)

Cellular component organization 183 3:92E − 10 RNH1, KDM1A, ZWILCH, DSCC1, HDAC11,INPPL1, TESK2, BUB1B, UBE3A, FNBP1L,…

Cellular process 507 1:22E − 09 CYFIP2, RPL7, HOXA5, HOXB5, EGFR,FOXN3, PTPN3, CDC20, FAS, PTCH1, IDH2,…

Cell cycle phase 49 8:98E − 09 ZWILCH, CDC20, EGFR, FOXN3, HSPA2,NDC80, ZWINT, TPX2, CENPF, KIF18A, MC1R,…

M phase 42 1:86E − 08 CDCA3, ZWILCH, DSCC1, CDCA8, BUB1B,TTK, MKI67, NCAPH, CDC20, PSMC3IP,…

Cell proliferation 48 2:47E − 08 ACHE, CSE1L, EGFR, PTCH1, WNT5A, EMP1,SOX11, CBFA2T3, EREG, TPX2,…

GOTERM_CC (top 5)

Intracellular part 552 1:83E − 12 CYFIP2, PTPN3, EGFR, CDC20, FAS, FOXN3,NUP107, RNH1, TESK2, NOC2L,…

Intracellular 560 5:23E − 11 CYFIP2, PTPN3, EGFR, CDC20, FAS, FOXN3,SPI1, NUP107, RNH1, TESK2,…

Cytoplasm 403 4:48E − 09 CYFIP2, PTPN3, EGFR, CDC20, FAS, RNH1,TESK2, FNBP1L, RPL7, AKT1,…

Intracellular organelle 473 4:91E − 09 PTPN3, EGFR, CDC20, FAS, FOXN3, SPI1,NUP107, RNH1, TESK2, NOC2L,…

Spindle 26 5:26E − 09 CDCA8, BUB1B, TTK, CDC14A, CDC20,CCNB1, RACGAP1, HAUS7, MYC, AKT1,…

GOTERM_MF

Protein binding 448 2:09E − 12 CYFIP2, EGFR, FOXN3, PTPN3, CDC20,FAS,PTCH1, MARCKSL1, CNTFR, SPI1,…

Binding 603 7:51E − 09 CYFIP2, EGFR, FOXN3, PTPN3, CDC20,FAS, PTCH1, MARCKSL1, HOXB5, CNTFR,…

Ribonuclease H activity 4 1:21E − 04 FEN1, RNASEH2A, RNH1, EXO1

GO: Gene Ontology; BP: biological process; MF: molecular function; CC: cellular component; BCC: basal cell carcinoma.

4 BioMed Research International

Page 5: HDBMR 2073690 1.downloads.hindawi.com/journals/bmri/2020/2073690.pdf2.4. Protein-Protein Interaction (PPI) Network. The Biologi-cal General Repository for Interaction Datasets (BioGRID)

strongly related to BCC initiation and progression [23].Gianferante et al. evaluated 18 nevoid BCC syndromeNational Cancer Institute families by whole exome sequenc-ing and found that 89% of families exhibited a pathogenicPTCH1 mutation [24]. A recent investigation has reportedthat PTCH1 had two mutational statuses (germinal andsomatic mutation). Moreover, there was a higher expressionlevel of PTCH1 in BCC with germinal and somatic PTCH1mutations than that only with germinal PTCH1 mutation[25]. Additionally, our functional analyses showed thatPTCH1 played significant roles in cell proliferation andBCC pathway. However, few reports investigated thedetailed molecular mechanisms of the biological roles ofPTCH1 on BCC cell proliferation and growth. Herein,we performed a ROC curve analysis of PTCH1, and theresult indicated that this gene was capable of discriminatingBCC patients and healthy controls with a relatively highAUC value (AUC = 0:981). Taken together, our findings sug-gested that PTCH1 might be involved in the pathogenesis ofBCC and has a great diagnostic value for BCC detection.

We found that four genes (upregulated CYFIP2 andMARCKSL1 and downregulated HOXB5 and PTPN3) werealso key players in cell biological processes. Furthermore,their expression patterns were validated by the differentialexpression analysis based on an external dataset. Besides,these four genes could clearly differentiate BCC patients fromnormal controls. CYFIP2 was reported to be a direct p53 tar-get gene, and its overexpression was closely linked with thedevelopment of various cancers [26, 27]. Ling et al. analyzed

p53 mutations in sporadic and hereditary BCC tumors, andgenetic alterations of p53 were responsible for BCC progres-sion [28]. Later on, Huang et al. stated that p53 activation byimiquimod contributed to cell apoptosis of a skinBCC/KMC1 cell line [29]. These findings provided indirectevidence for the hypothesis that CYFIP2 participated in thepathogenic mechanism of BCC. HOXB5 is a member of thehomeobox (HOX) gene B cluster and plays key roles in sev-eral cancers [30, 31]. Lee et al. suggested that HOXB5increased cell proliferation and invasiveness in estrogenreceptor- (ER-) positive breast cancer [32]. More interest-ingly, this research group also found that HOXB5 couldupregulate EGFR expression and thereby promote HOXB5-driven invasion in ER-positive breast cancer [33]. In thiswork, we noted that EGFR was involved in cell cycle and pro-liferation and served as a pivotal hub gene in PPI analysis.Whether the HOXB5/EGFR axis participated in BCC devel-opment remains to be illuminated in the following analysis.PTPN3 (also known as PTPH1) was also reported to bestrongly correlated with diverse cancers including colorectalcancer and gastric adenocarcinoma [34–36]. Furthermore,Li et al. unraveled that the PTPN3 depletion could block thedegradation of EGFR, which accelerated cell proliferationand tumorigenicity in lung cancer cells [37]. The potentialroles of the PTPN3/EGFR axis on BCC occurrence also needto be clarified in the future.MARCKSL1 is a cytoskeletal reg-ulator and implicated with the initiation of multiple cancers[38]. Our results implied that MARCKSL1 might be a prom-ising diagnostic maker for BCC prediction. However, the

DNA

Basal cell carcinoma

Normal keratinocyteCutaneous keratinocyte

Genetic alterations

Oncogenes :

Tumor suppressors : p53, PTCHI

SHH, SMO

SHH

Cholesterol KIF7SUFU

MicrotubuleBMR

DNAdamage

DNAp53

Cell cycle

p21

Bax BakUncontrolled proliferationIncreased survivalGenomic instability

GADD45

POLKp48

p53 signalingpathway

Proliferation

Target genesDNS

TCF/LEFAxin

+pGSK-3𝛽Dvl

Basal cell carcinoma

(c) Kanehisa laboratories05217 7/25/17

Wnt signalingpathway

APCProliferationinvasion

TGF signalingpathway

HIP1

GLI1

PTCHI,2

Wnt

Hedgehog signalingpathway

GLISMO

PTCH1

Frizzled 𝛽-Catenin

Figure 2: Basal cell carcinoma pathway from the Kyoto Encyclopedia of Genes and Genomes enrichment analysis. The yellow color shows theupregulated genes while blue color represents the downregulated genes.

5BioMed Research International

Page 6: HDBMR 2073690 1.downloads.hindawi.com/journals/bmri/2020/2073690.pdf2.4. Protein-Protein Interaction (PPI) Network. The Biologi-cal General Repository for Interaction Datasets (BioGRID)

ITPASH3GLB2

MLLT11

MRPL11 TXNL4ATOPBP1

S100A7

PSME4

NCAPHCLN6

SERPINB5 CCP110

SNRPF

CXorf57

ATAD2

GPN1

GADD45G

SMARCE1

DNPEP ATG16L1

MED28SLC20A1VAT1

FOXN3ICAM4MLLT4 SYNRG

BCL2L11BIDREL PTK6

ARID1A

FAM53CTSPYL2FAS

BCL2L2

GINS1

MYO1E

FAM208B

STK38L CTSVUGCG

AFF1ETV4

NOL11 C19orf66 PTPN21PARP4 IMPA2 KCTD9 SNRPGSOX4

RARA

NASPDNAJB2

WBP2

UBE3A

PHLPP1

MAT1A

MDK

AVEN

CDH10

FANCG

DEF8

FBXO11OSGEP

SORL1

WDR3

KLF4PFAS

CNOT2

COPS7A

ZFP36ROBO1

PSMG2

NECAP2

UBTD1

GPC4

NOTCH3

SOX13

PORPPP1R13L

ZFYVE21

VAPB

TOX4HMGB3

IFI35DCUN1D2

MX2

OSER1

ZCCHC14

AP3S1

RIN2SNRK

RAD51AP1

ZFP36L2REXO2

PFN2SEPHS1

EHBP1

N4BP3

ZNF224

NPEPPS

OCRL

ACSL1

ZC2HC1A

SLC26A2TNFRSF14

KIAA1107

ZNF324B

MKL2

MPST

DLGAP5

NFE2SLC35F6

FBXW4NDUFA4L2

SS18L2

LRP4

C9orf9

BACH2MINA

MAFF

NFE2L1

ZNF556

CORO1C

ZNF692

NF2

RALBP1

SETMAR

ZFC3H1PPIH

MNTFKBP5

VWA8

WASF3

ATG7

BUB1B

S100A9CENPF

SLC1A1

PSRC1

SMAD4

ST5

CUX1

GPRIN2

GAS2L1

ODF2

PAX6TUFT1

KIF18A

RACGAP1

CEP152

PRC1

MARCKSL1

MCM6

ACTR1B

SPP1

TP53BP2

FOXO1

PCDH12

PTGER4

STX8ZNF135

ARL15

SERINC1

OLFM4

DBI

UCP2

COL18A1

SLC9A6

NUP37

ATP2B4

MPDZ

AGO4GPRC5C

CTAGE5COMMD3

SETBP1

SERPINB4

CRIP1ALCAM

DSCC1

CTSB

ITIH5

TK1NMT2KCNE1

MRPL23ECI1

MC1RCOL9A3COL17A1

FZD7

TUSC3

MFSD5

PHKA1

UPK1APLEKHA1

SPRYD7

LPAR1 FZD3

TRPM4

GAS6

TMEM246

RNF170

SLC35F2

EXTL2 TYRO3

LPHN3

ADCY6

PTPRTNDC1

POLRMTCOG2

KLHL22

MST1R

ECM2

TAF1C

ANXA7NPR2

ZNF232

CDC45

RNF34

DNA2OXA1L

LPP

TAL1SF3A3

CAND1

EGLN3

CBFA2T3

KRT31

MAGED2

KLF5

PSMA5

IMPDH2

MRPS7

LSM8

CCHCR1ALDH18A1

CCNB1

SRSF7

MYC

CBX3

HERC5

AKT1

AIM1

SRSF6

HDAC1C2orf44

KRT8

PRPF8

TUBB

UGP2

MYBL2SNX4

KDM1A

ARNTL

NINL

TOP1

MEF2A

USP11

ATXN3L

NFE2L2

IRS2JUP

GTF2F2

MAGED1

DSC3

ICAM1DUSP2

SFXN1

CSNK2A2

DTL

KRT6B

SIK3

STIL

APRT TPX2

HMMR

PEA15RNF13

CCSER2

TCEB2CRYL1

PLEKHO1

VBP1

MAPK8IP3

POLR2H

TIA1GINS3

ANP32BCARM1

WDR45TRAK1

SLC7A5DOK4

GLO1

BRIP1MID2

DROSHA SNAI1

MKI67ERH

PRIM1

HNRNPA0

COPB1

NCL

SEL1L

CSE1L

XPO1

NOC2L

ZNF22XRCC5

EIF3HZAP70

FAM57A

METTL1

ETFA

PCGF2

BHLHE40

ETFB

SCAMP1

OSTF1

KIF18B

AHCYL2

SNAP25

GEM

CTSL

TESK2POLE2

RECK

PTOV1LRBA

RPS9

CLN5

LRP8MME

ZMYM4MCM2

ADRB2

INPPL1

CSNK2B

RNH1

MCM5

ASF1B

BAIAP2

ANXA2 SFNKRT9

RASSF9

TOP2ANUP107

NDC80

HK1PPP3CA

SLC1A5IDH2

CD4

CHD3

SLC3A2

LMNB1

PTPN3

CFTR

ITGA4

RPL7

PRKACAORC6

CEP55

TGFB3

ASS1

DPP3

EDIL3

COASY

PPP2R2B

PAK6

KIFC1

GAS2

TROAP

CDC14AGLI1

SHCBP1

KLF3

E2F1

CENPAMLX

HDAC11

SPI1

EFHC2

LONRF1

IRF8

HSPG2AMACR

LHX2MIPEP

CNKSR1

CDKL1

SDC4

SYBU

NUAK1

GCAT

PBK

TRIM16

SACS

RMND5B

PPME1

ISG20L2

CYFIP2

UBE2C

CAMK1PHKA2

TRO

NFYA

IRF1

GPX2

EML1

SNRNP25

GPR137BSPINK1

CDCA8

UBE2I

ICT1

LEF1

GCDH

HNRNPA3

COCH

ANG

AHNAK2PHC1CD44

CORO2A

CBX6KIFAP3

PSEN1DDB2

CD163 FEN1

ATP1B3

HSPA2

IVD

SEMA6AIL13RA2

CDIPT PTCH1

CNNM1

MAST2

SNCA

RRM2

SOX9

VAMP3

B4GALT5

LRRTM4

DHFR

SFTPCBEND5

SERPINB9

CCPG1

PARPBP

TRMT61B

SEMA4C

ACAA2

HERPUD1

SNCAIP

KALRN

NPTXR

MX1EGFR

BIRC5ILF2

EXO1

CHAF1B KIF20A

RAD51

VPS51

CAPRIN1

TRIP13 HAUS7SYNCAGTPBP1

ZWINT

MCM3

CDC7

RTF1

KIF23

RNASEH2A

SPHK2RAB9A

ZBTB43

RBP4BICD2

PIPOX C17orf59

CKS2 TACC2

DACT1

RFC4

TJP2TTK

CDC20

EHD1 SPC25

ELP4

SLC9A3R2TTC37

LIMCH1

MTG1

CNTFRELN

CLCF1BGN

NSG1

HOMER1

HOXA5

ZNF26

PLEKHG6

CRISP2

LIMK1

FRYLTACC3

C2CD2 MMP28 FAM49B FAM50B CDKN3 HYAL1KCNS3

ARHGAP26 TRIM68 FAM49A ARHGEF28AGMAT UBAC1RETSAT

EREG

KIAA0101DPP8

PLXNA2LAMB1

LAMA3FARP1 FRK

VCANMGST2AVPI1

HLTF DHX32OS9SDC2

(a)

MCM6

PRPF8

CDC45

NCL

ORC6

ITGA4

CHD3

DNA2

CDC7MCM3

(b)

CAND1

ILF2

UBE2I

S100A9

RPL7

JUP

MCM5HNRNPA3

ICAM1

TOP1

(c)

ARNTL

COG2

SNX4

TUFT1

VPS51

CCHCR1

(d)

Figure 3: The protein-protein interaction (PPI) network analysis: (a) PPI network of differentially expressed genes: (b) submodule 1; (c)submodule 2; (d) submodule 3. The submodules in the PPI network were extracted by MCODE based on PPI score > 3. The rosy red circularnodes denote upregulated genes. The green circular nodes represent significantly downregulated genes. The node size represents degree.

6 BioMed Research International

Page 7: HDBMR 2073690 1.downloads.hindawi.com/journals/bmri/2020/2073690.pdf2.4. Protein-Protein Interaction (PPI) Network. The Biologi-cal General Repository for Interaction Datasets (BioGRID)

underlying pathogenic mechanisms of the four-gene signa-ture (CYFIP2, MARCKSL1, HOXB5, and PTPN3) for BCCprogression have not been expatiated.

The FAS gene contains a highly conserved cytoplasmicdeath domain and encodes a transmembrane protein of thetumor necrosis factor receptor superfamily. Existing evidencehas shown that FAS could bind to a death ligand, FAS ligand(FASL), to induce the cell apoptotic pathway [39]. The earlyresearch indicated that FAS normally was underexpressedand even undetectable in BCC while there was a high FASLlevel, possibly inducing cell death and contributing to cancer-ization [40]. A later study examined the expression level ofFAS/FASL in BCCs using the immunohistochemical method.The results implied that FASL was located on the cell mem-brane of keratinocytes at the basal cell layers and FAS/FASLwas markedly decreased in BCC [41]. Wang et al. revealedthat FAS/FASL mRNA expression and protein levels werereduced in the BCC compared to the normal skin samplesand FASL immunostaining levels were strongly related tothe ability of tumor invasiveness and metastasis [42]. Simi-larly, we found that there was a lower FAS expression inBCC tissues than healthy controls, which was also confirmedby a bioinformatics analysis with another BCC dataset.Therefore, we speculated that FASmight be a key gene driverin BCC development. The other two genes’ (CDC20 andFOXN3) levels were increased in BCC tissues compared tonormal tissues, and functional analysis showed that they

were mainly involved in the cell cycle process. We alsoobserved that CDC20 exhibited a good performance forBCC diagnosis based on a ROC analysis. Therefore, weinferred that the aberrant expression of CDC20 and FOXN3was probably associated with BCC development. However,the influence of these two genes on BCC pathogenesis hasnot been fully investigated.

There are still limitations in our analysis. Firstly, only oneexternal noncoding RNA dataset GSE74858 was obtainedand used to evaluate expression levels of identified DEGsdue to lack of gene expression profile of BCC. Notably, theexpression patterns of six genes (CYFIP2, HOXB5, FOXN3,PTPN3, MARCKSL1, and FAS) were consistent with our ini-tial differential expression analysis in the training dataset.However, the expression levels of other key genes still needto be further evaluated by using a larger sample size. Sec-ondly, the corresponding experimental assays such as celland animal experiments were not performed to validate ourconclusion primarily due to lack of enough patients’ samplesand limited research funding. Thirdly, the underlying molec-ular mechanism between several signaling pathways involv-ing key genes and BCC development remains to bedeciphered. Fourthly, the relevant clinical characteristics arealso required to be collected to assess BCC prognosis.

In summary, nine gene signatures (CYFIP2, HOXB5,EGFR, FOXN3, PTPN3, CDC20, MARCKSL1, FAS, andPTCH1) may play central roles in the initiation and

Basal cell carcinoma Normal skin

5.5

6.0

6.5

7.0

7.5

8.0

GSE74858 CYFIP2p= 0.0224115966518463

Basal cell carcinoma Normal skin

9.2

9.4

9.6

9.8

10.0GSE74858 FAS

p= 0.0363963093133442

Basal cell carcinoma Normal skin

9.2

9.4

9.6

9.8

10.0GSE74858 FAS

p= 0.0363963093133442

Basal cell carcinoma Normal skin7.0

7.5

8.0

8.5

9.0

9.5

10.0

GSE74858 MARCKSL1p= 0.0204961661868877

Basal cell carcinoma Normal skin

7.0

7.2

7.4

7.6

7.8

8.0

8.2

8.4

GSE74858 PTPN3p= 0.0377593208674906

Basal cell carcinoma Normal skin

8.0

8.2

8.4

8.6

8.8

9.0

9.2

GSE74858 HOXB5p= 0.0102197391403201

Figure 4: Validation of the expression levels of six differentially expressed genes in T basal cell carcinoma based on GSE74858 dataset. The x-axis represents basal cell carcinoma and normal controls. The y-axis represents the expression read counts.

7BioMed Research International

Page 8: HDBMR 2073690 1.downloads.hindawi.com/journals/bmri/2020/2073690.pdf2.4. Protein-Protein Interaction (PPI) Network. The Biologi-cal General Repository for Interaction Datasets (BioGRID)

progression of BCC, which provides deeper insights into BCCmanagement. However, experimental verification and inte-grated bioinformatics analyses still need to be carried out inthe future.

Data Availability

The datasets used and/or analyzed during the current study areavailable from the corresponding author on reasonable request.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Supplementary Materials

The whole R sentences for data processing and identificationof differentially expressed genes in this study. (SupplementaryMaterials)

References

[1] R. Ballester-Sánchez, O. Pons-Llanas, C. Candela-Juan et al.,“Efficacy and safety of electronic brachytherapy for superficial

and nodular basal cell carcinoma,” Journal of ContemporaryBrachytherapy, vol. 3, no. 3, pp. 231–238, 2015.

[2] J. A. C. Verkouteren, K. H. R. Ramdas, M. Wakkee, andT. Nijsten, “Epidemiology of basal cell carcinoma: scholarlyreview,” The British Journal of Dermatology, vol. 177, no. 2,pp. 359–372, 2017.

[3] A. I. Rubin, E. H. Chen, and D. Ratner, “Basal-cell carcinoma,”The New England Journal of Medicine, vol. 353, no. 21,pp. 2262–2269, 2005.

[4] Y. Chen and J. Liu, “The prognostic roles of cyclooxygenase-2 forpatients with basal cell carcinoma,”Artificial Cells, Nanomedicine,and Biotechnology, vol. 47, no. 1, pp. 3053–3057, 2019.

[5] S. C. Flohil, I. Seubring, M. M. van Rossum, J. W. W. Coe-bergh, E. de Vries, and T. Nijsten, “Trends in basal cell carci-noma incidence rates: a 37-year Dutch observational study,”The Journal of Investigative Dermatology, vol. 133, no. 4,pp. 913–918, 2013.

[6] K. Tanese, “Diagnosis and management of basal cell carcinoma,”Current Treatment Options in Oncology, vol. 20, no. 2, p. 13, 2019.

[7] R. Wadlow and S. Ramaswamy, “DNA microarrays in clinicalCancer Research,” Current Molecular Medicine, vol. 5, no. 1,pp. 111–120, 2005.

[8] M. Fang, S. A.Wee, K. Ronski, H. Fan, S. Tao, and Q. Lin, “Evi-dence of EGR1 as a differentially expressed gene among

CDC20 ROC curve

1 – Specificity

Sens

itivi

ty

0.0 0.2 0.4 0.6 0.8 1.0

0.0

0.2

0.4

0.6

0.8

1.00.423 (0.847, 0.958)

AUC: 0.956

CYFIP2 ROC curve

1 – Specificity

Sens

itivi

ty0.0 0.2 0.4 0.6 0.8 1.0

0.0

0.2

0.4

0.6

0.8

1.0

−0.124 (1.000, 0.875)

AUC: 0.949

HOXB5 ROC curve

1 – Specificity0.0 0.2 0.4 0.6 0.8 1.0

0.0

0.2

0.4

0.6

0.8

1.0

−0.085 (0.906, 0.875)

AUC: 0.908

MARCKSL1 ROC curve

1 – Specificity

Sens

itivi

ty

0.0 0.2 0.4 0.6 0.8 1.0

0.0

0.2

0.4

0.6

0.8

1.01.435 (0.965, 0.938)

AUC: 0.962

PTCH1 ROC curve

1 – Specificity

Sens

itivi

ty

0.0 0.2 0.4 0.6 0.8 1.0

0.0

0.2

0.4

0.6

0.8

1.0−1.045 (0.882, 1.000)

AUC: 0.981

PTPN3 ROC curve

1 – Specificity

Sens

itivi

tySe

nsiti

vity

0.0 0.2 0.4 0.6 0.8 1.0

0.0

0.2

0.4

0.6

0.8

1.0

0.447 (0.918, 0.896)

AUC: 0.952

Figure 5: The receiver operating characteristic (ROC) curve analyzed the diagnostic value of six differentially expressed genes in basal cellcarcinoma. The ROC curves were used to evaluate the diagnostic value of these six differentially expressed genes in basal cell carcinomawith sensitivity and 1-specificity. The x-axis shows 1-specificity, and the y-axis shows sensitivity. AUC: area under the ROC curve.

8 BioMed Research International

Page 9: HDBMR 2073690 1.downloads.hindawi.com/journals/bmri/2020/2073690.pdf2.4. Protein-Protein Interaction (PPI) Network. The Biologi-cal General Repository for Interaction Datasets (BioGRID)

proliferative skin diseases,” Genomic Medicine, vol. 1, no. 1-2,pp. 75–85, 2007.

[9] E. R. Heller, A. Gor, D. Wang et al., “Molecular signatures ofbasal cell carcinoma susceptibility and pathogenesis: a geno-mic approach,” International Journal of Oncology, vol. 42,no. 2, pp. 583–596, 2013.

[10] T. Yunoki, Y. Tabuchi, T. Hirano, S. Miwa, J. Imura, andA. Hayashi, “Gene networks in basal cell carcinoma of the eye-lid, analyzed using gene expression profiling,” Oncology Let-ters, vol. 16, no. 5, pp. 6729–6734, 2018.

[11] B. A. Jee, H. Lim, S. M. Kwon et al., “Molecular classification ofbasal cell carcinoma of skin by gene expression profiling,”Molecular Carcinogenesis, vol. 54, no. 12, pp. 1605–1612, 2015.

[12] T. Barrett, D. B. Troup, S. E. Wilhite et al., “NCBI GEO: min-ing tens of millions of expression profiles–database and toolsupdate,” Nucleic Acids Research, vol. 35, Database, pp. D760–D765, 2007.

[13] A. Ramasamy, A. Mondry, C. C. Holmes, and D. G. Altman,“Key issues in conducting a meta-analysis of gene expressionmicroarray datasets,” PLoS Medicine, vol. 5, no. 9, articlee184, 2008.

[14] G. Marot, J. L. Foulley, C. D. Mayer, and F. Jaffrezic, “Moder-ated effect size and P-value combinations for microarray meta-analyses,” Bioinformatics, vol. 25, no. 20, pp. 2692–2699, 2009.

[15] 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.

[16] C. Xie, X. Mao, J. Huang et al., “KOBAS 2.0: a web server forannotation and identification of enriched pathways and dis-eases,” Nucleic Acids Research, vol. 39, Supplement_2,pp. W316–W322, 2011.

[17] 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,” Bio Systems, vol. 127, pp. 67–72,2015.

[18] Z. K. Liu, R. Y. Zhang, Y. L. Yong et al., “Identification of cru-cial genes based on expression profiles of hepatocellular carci-nomas by bioinformatics analysis,” PeerJ, vol. 7, article e7436,2019.

[19] H. Fan, A. E. Oro, M. P. Scott, and P. A. Khavari, “Induction ofbasal cell carcinoma features in transgenic human skinexpressing Sonic Hedgehog,” Nature Medicine, vol. 3, no. 7,pp. 788–792, 1997.

[20] J. P. Lacour, “Carcinogenesis of basal cell carcinomas: geneticsand molecular mechanisms,” The British Journal of Dermatol-ogy, vol. 146, Supplement 61, pp. 17–19, 2002.

[21] D. M. Stone, M. Hynes, M. Armanini et al., “The tumour-suppressor gene patched encodes a candidate receptor forSonic hedgehog,” Nature, vol. 384, no. 6605, pp. 129–134,1996.

[22] A. B. Undén, P. G. Zaphiropoulos, K. Bruce, R. Toftgård, andM. Ståhle-Bäckdahl, “Human patched (PTCH) mRNA is over-expressed consistently in tumor cells of both familial and spo-radic basal cell carcinoma,” Cancer Research, vol. 57, no. 12,pp. 2336–2340, 1997.

[23] S. C. Bresler, B. L. Padwa, and S. R. Granter, “Nevoid basal cellcarcinoma syndrome (Gorlin syndrome),” Head and NeckPathology, vol. 10, no. 2, pp. 119–124, 2016.

[24] D. M. Gianferante, M. Rotunno, M. Dean et al., “Whole-exomesequencing of nevoid basal cell carcinoma syndrome families

and review of Human Gene Mutation Database PTCH1 muta-tion data,” Molecular Genetics & Genomic Medicine, vol. 6,no. 6, pp. 1168–1180, 2018.

[25] M. F. Martinez, M. Romano, A. Martinez et al., “Nevoid basalcell carcinoma syndrome: PTCH1 mutation profile andexpression of genes involved in the hedgehog pathway inArgentinian patients,” Cells, vol. 8, no. 2, p. 144, 2019.

[26] E. Saller, E. Tom, M. Brunori et al., “Increased apoptosisinduction by 121F mutant p53,” The EMBO Journal, vol. 18,no. 16, pp. 4424–4437, 1999.

[27] R. S. Jackson II, Y.-J. Cho, S. Stein, and P. Liang, “CYFIP2, adirect p53 target, is leptomycin-B sensitive,” Cell Cycle, vol. 6,no. 1, pp. 95–103, 2007.

[28] G. Ling, A. Ahmadian, Å. Persson et al., “PATCHED and p53gene alterations in sporadic and hereditary basal cell cancer,”Oncogene, vol. 20, no. 53, pp. 7770–7778, 2001.

[29] S. W. Huang, S. H. Chang, S. W. Mu et al., “Imiquimod acti-vates p53-dependent apoptosis in a human basal cell carci-noma cell line,” Journal of Dermatological Science, vol. 81,no. 3, pp. 182–191, 2016.

[30] B. Zhang, N. Li, and H. Zhang, “Knockdown of homeobox B5(HOXB5) inhibits cell proliferation, migration, and invasion innon-small cell lung cancer cells through inactivation of theWnt/β-catenin pathway,” Oncology Research, vol. 26, no. 1,pp. 37–44, 2018.

[31] N. Xu, Y. P. Wu, H. B. Yin, X. Y. Xue, and X. Gou, “Molecularnetwork-based identification of competing endogenous RNAsandmRNA signatures that predict survival in prostate cancer,”Journal of Translational Medicine, vol. 16, no. 1, p. 274, 2018.

[32] J. Y. Lee, H. Hur, H. J. Yun et al., “HOXB5 promotes the pro-liferation and invasion of breast cancer cells,” InternationalJournal of Biological Sciences, vol. 11, no. 6, pp. 701–711, 2015.

[33] J. Y. Lee, J. M. Kim, D. S. Jeong, and M. H. Kim, “Transcrip-tional activation of EGFR by HOXB5 and its role in breast can-cer cell invasion,” Biochemical and Biophysical ResearchCommunications, vol. 503, no. 4, pp. 2924–2930, 2018.

[34] S. Han, S. Williams, and T. Mustelin, “Cytoskeletal proteintyrosine phosphatase PTPH1 reduces T cell antigen receptorsignaling,” European Journal of Immunology, vol. 30, no. 5,pp. 1318–1325, 2000.

[35] C. W. Wu, J. H. Chen, H. L. Kao et al., “PTPN3 and PTPN4tyrosine phosphatase expression in human gastric adenocarci-noma,” Anticancer Research, vol. 26, no. 2B, pp. 1643–1649,2006.

[36] Z. Wang, D. Shen, D.W. Parsons et al., “Mutational analysis ofthe tyrosine phosphatome in colorectal cancers,” Science,vol. 304, no. 5674, pp. 1164–1166, 2004.

[37] M. Y. Li, P. L. Lai, Y. T. Chou et al., “Protein tyrosine phospha-tase PTPN3 inhibits lung cancer cell proliferation and migra-tion by promoting EGFR endocytic degradation,” Oncogene,vol. 34, no. 29, pp. 3791–3803, 2015.

[38] Z. Chen, Y. Liu, L. Yao, S. Guo, Y. Gao, and P. Zhu, “The longnoncoding RNA lncZic2 drives the self-renewal of liver tumor-initiating cells via the protein kinase C substrates MARCKSand MARCKSL1,” The Journal of Biological Chemistry,vol. 293, no. 21, pp. 7982–7992, 2018.

[39] T. Dicker, G. Siller, and N. Saunders, “Molecular and cellularbiology of basal cell carcinoma,” The Australasian Journal ofDermatology, vol. 43, no. 4, pp. 241–246, 2002.

[40] C. Gutierrez-Steil, T. Wrone-Smith, X. Sun, J. G. Krueger,T. Coven, and B. J. Nickoloff, “Sunlight-induced basal cell

9BioMed Research International

Page 10: HDBMR 2073690 1.downloads.hindawi.com/journals/bmri/2020/2073690.pdf2.4. Protein-Protein Interaction (PPI) Network. The Biologi-cal General Repository for Interaction Datasets (BioGRID)

carcinoma tumor cells and ultraviolet-B-irradiated psoriaticplaques express Fas ligand (CD95L),” The Journal of ClinicalInvestigation, vol. 101, no. 1, pp. 33–39, 1998.

[41] T. J. Jang, “Expression of CD40 and Fas ligand in Bowen's dis-ease, squamous cell carcinoma and basal cell carcinoma,” Yon-sei Medical Journal, vol. 43, no. 3, pp. 304–308, 2002.

[42] X. Y.Wang, R. Zhang, and S. Lian, “Aberrant expression of Fasand FasL pro-apoptotic proteins in basal cell and squamouscell carcinomas,” Clinical and Experimental Dermatology,vol. 36, no. 1, pp. 69–76, 2011.

10 BioMed Research International