Network-Based Coexpression Analysis Identifies Functional ...TTs (d) Figure 1: The overview of the...

11
Research Article Network-Based Coexpression Analysis Identifies Functional and Prognostic Long Noncoding RNAs in Hepatocellular Carcinoma Jianguo Li, Jin Zhou, Shuangshuang Kai, Can Wang, Daijun Wang, and Jiying Jiang Schools of Basic Medicine and Pharmacy, Weifang Medical University, 7166 Baotong West Street, Weifang, 261053 Shandong Province, China Correspondence should be addressed to Jiying Jiang; [email protected] Received 19 July 2020; Revised 1 September 2020; Accepted 16 September 2020; Published 6 October 2020 Academic Editor: Tao Huang Copyright © 2020 Jianguo Li 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. Hepatocellular carcinoma (HCC) is a primary liver cancer associated with a growing incidence and extremely high mortality. However, the pathogenic mechanism is still not fully understood. In the present study, we identied 1,631 upregulated and 1,515 downregulated genes and found that cell cycle and metabolism-related pathways or biological processes highly dysregulated in HCC. To assess the biological importance of these DEGs, we carried out weighted gene coexpression network analysis (WGCNA) to identify the functional modules potentially involved in HCC pathogenesis or progression. The ve modules were detected with Dynamic Tree Cut algorithm, and GO enrichment analysis revealed that these modules exhibited dierent biological processes or signaling pathways, such as metabolism-related pathways, cell proliferation-related pathways, and molecules in tumor microenvironment. Moreover, we also observed two immune cells, namely, cytotoxic cells and macrophage enriched in modules grey and brown, respectively, while T helper cell-2 (Th2) was enriched in module turquoise. Among the WGCNA network, four hub long noncoding RNAs (lncRNAs) were identied to be associated with HCC prognostic outcomes, suggesting that coexpression network analysis could uncover lncRNAs with functional importance, which may be associated with prognostic outcomes of HCC patients. In summary, this study demonstrated that network-based analysis could identify some functional modules and some hub-lncRNAs, which may be critical for HCC pathogenesis or progression. 1. Introduction Hepatocellular carcinoma (HCC) is a primary liver cancer associated with a growing incidence and extremely high mortality, whose conrmed etiologic factors include hepa- titis B/C, alcohol use, nonalcoholic steatohepatitis, and obesity [1]. Also, cirrhosis is regarded as an important indi- cator in the screening and surveillance of HCC [2]. The rapid progression often leads to poor prognosis of HCC as most diagnoses are made at advanced disease stages [3]. With the advance in biotechnologies, genomic causes behind HCC have been gradually revealed. Genomic analyses of HCC have identied some recurrently mutated genes, such as TERT promoter, TP53, CTNNB1, and AXIN1 [4]. Previ- ous studies about microRNAs (miRNAs) show that miRNAs are closely related to HCC tumorigenesis, development and metastasis [5, 6]. For example, miR-188-5p can inhibit the proliferation and metastasis of HCC by targeting FGF5 [7]. Moreover, long noncoding RNAs (lncRNAs), which are gen- erally unable to encode proteins, are also involved in tumor formation, development, or metastasis. Overexpression of lncRNA HULC in liver cancer promotes HCC proliferation by downregulating tumor suppressor gene p18 [8]. With the development of the biomarkers of HCC, the therapeutics has been greatly improved. However, for patients with advanced stages, the traditional surgical resec- tion and chemotherapy are inadequacy. Transplantation, genomic-based, and immune therapies now become the cen- ter of attention as they exhibit a very promising eect on those virally induced cancers like HCC, and immunotherapy regarding immune checkpoint inhibitors has been applied clinically in cancers such as melanoma and non-small-cell lung cancer [9]. Inltrating immune cells play a critical role in the surveillance and immune response of various solid tumors and contribute greatly to the identication of immu- notherapy targets [10]. Inltrating immune cells mainly fall Hindawi BioMed Research International Volume 2020, Article ID 1371632, 11 pages https://doi.org/10.1155/2020/1371632

Transcript of Network-Based Coexpression Analysis Identifies Functional ...TTs (d) Figure 1: The overview of the...

  • Research ArticleNetwork-Based Coexpression Analysis Identifies Functional andPrognostic Long Noncoding RNAs in Hepatocellular Carcinoma

    Jianguo Li, Jin Zhou, Shuangshuang Kai, Can Wang, Daijun Wang, and Jiying Jiang

    Schools of Basic Medicine and Pharmacy, Weifang Medical University, 7166 Baotong West Street, Weifang,261053 Shandong Province, China

    Correspondence should be addressed to Jiying Jiang; [email protected]

    Received 19 July 2020; Revised 1 September 2020; Accepted 16 September 2020; Published 6 October 2020

    Academic Editor: Tao Huang

    Copyright © 2020 Jianguo Li 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.

    Hepatocellular carcinoma (HCC) is a primary liver cancer associated with a growing incidence and extremely high mortality.However, the pathogenic mechanism is still not fully understood. In the present study, we identified 1,631 upregulated and1,515 downregulated genes and found that cell cycle and metabolism-related pathways or biological processes highlydysregulated in HCC. To assess the biological importance of these DEGs, we carried out weighted gene coexpression networkanalysis (WGCNA) to identify the functional modules potentially involved in HCC pathogenesis or progression. The fivemodules were detected with Dynamic Tree Cut algorithm, and GO enrichment analysis revealed that these modules exhibiteddifferent biological processes or signaling pathways, such as metabolism-related pathways, cell proliferation-related pathways,and molecules in tumor microenvironment. Moreover, we also observed two immune cells, namely, cytotoxic cells andmacrophage enriched in modules grey and brown, respectively, while T helper cell-2 (Th2) was enriched in module turquoise.Among the WGCNA network, four hub long noncoding RNAs (lncRNAs) were identified to be associated with HCC prognosticoutcomes, suggesting that coexpression network analysis could uncover lncRNAs with functional importance, which may beassociated with prognostic outcomes of HCC patients. In summary, this study demonstrated that network-based analysis couldidentify some functional modules and some hub-lncRNAs, which may be critical for HCC pathogenesis or progression.

    1. Introduction

    Hepatocellular carcinoma (HCC) is a primary liver cancerassociated with a growing incidence and extremely highmortality, whose confirmed etiologic factors include hepa-titis B/C, alcohol use, nonalcoholic steatohepatitis, andobesity [1]. Also, cirrhosis is regarded as an important indi-cator in the screening and surveillance of HCC [2]. The rapidprogression often leads to poor prognosis of HCC as mostdiagnoses are made at advanced disease stages [3].

    With the advance in biotechnologies, genomic causesbehind HCC have been gradually revealed. Genomic analysesof HCC have identified some recurrently mutated genes, suchas TERT promoter, TP53, CTNNB1, and AXIN1 [4]. Previ-ous studies about microRNAs (miRNAs) show that miRNAsare closely related to HCC tumorigenesis, development andmetastasis [5, 6]. For example, miR-188-5p can inhibit theproliferation and metastasis of HCC by targeting FGF5 [7].

    Moreover, long noncoding RNAs (lncRNAs), which are gen-erally unable to encode proteins, are also involved in tumorformation, development, or metastasis. Overexpression oflncRNA HULC in liver cancer promotes HCC proliferationby downregulating tumor suppressor gene p18 [8].

    With the development of the biomarkers of HCC, thetherapeutics has been greatly improved. However, forpatients with advanced stages, the traditional surgical resec-tion and chemotherapy are inadequacy. Transplantation,genomic-based, and immune therapies now become the cen-ter of attention as they exhibit a very promising effect onthose virally induced cancers like HCC, and immunotherapyregarding immune checkpoint inhibitors has been appliedclinically in cancers such as melanoma and non-small-celllung cancer [9]. Infiltrating immune cells play a critical rolein the surveillance and immune response of various solidtumors and contribute greatly to the identification of immu-notherapy targets [10]. Infiltrating immune cells mainly fall

    HindawiBioMed Research InternationalVolume 2020, Article ID 1371632, 11 pageshttps://doi.org/10.1155/2020/1371632

    https://orcid.org/0000-0002-8729-6611https://creativecommons.org/licenses/by/4.0/https://doi.org/10.1155/2020/1371632

  • into two groups: lymphoid and myeloid [11]. Recent studystated that the degree of immune cell infiltration into HCCis associated with divergent immune cell types and correlatedto prognosis [12]. In the present study, we attempt to identifysome key genes, functional modules, and pathways for HCCtumorigenesis and progression using network-based algo-rithm. The immune cells infiltrated in HCC tissues were alsoevaluated, and some critical lncRNAs were identified by thecoexpression network. In summary, this study improvedour understanding of HCC tumorigenesis and providedsome potential therapeutic targets for HCC.

    2. Materials and Methods

    2.1. Data Collection. We collected RNA sequencing data of50 HCC and 50 paired nontumor tissues from theSequence Read Archive (SRA, https://www.ncbi.nlm.nih.gov/sra) database [13] with an accession numberSRP068976 [14]. The SRA files were preprocessed by fastq-dump with the option –split-files, which generated two pairedfastq files.

    2.2. Read Mapping and Gene Expression Quantification. TheRNA sequencing reads were first mapped to UCSC hg19human reference genome (http://www.genome.ucsc.edu/)using hisat2 [15], and the alignments in SAM file were thensorted by samtools. The gene expression was quantified byStringTie [16] and ballgown pipeline, with the gene annota-tion from GENCODE v19 [17].

    2.3. Differential Expression Analysis. The read count-basedexpression was used to identify the differentially expressedgenes (DEGs) by R/bioconductor DESeq2 package [18].The stably expressed genes were firstly identified ifFPKM ðfragment per kilobasemillionÞ > 1 in more than20% samples. The differentially expressed genes were identi-fied with the thresholds of BH (Benjamini and Hochberg)adjusted P value < 0.05 and fold change > 2 or < 1/2 [19, 20].

    2.4. Weighted Gene Coexpression Network Analysis(WGCNA). WGCNA [21] was performed to identify poten-tial functional modules. The soft threshold for scale-free net-work was determined based on the maximal R-square(power = 9). TOM similarity was used to evaluate the dis-tance between each gene pair. Moreover, hierarchical cluster-ing analysis with average method and dynamic method wasused to build the cluster tree and classify the genes into mod-ules, respectively. We finally identified 5 functional modules.

    2.5. KEGG, GO, and Immune Cell-Based OverrepresentationEnrichment Analysis. The KEGG (Kyoto Encyclopedia ofGenes and Genomes) [22], GO (Gene Ontology) [23, 24],and immune cell-based overrepresentation enrichment anal-ysis were implemented in R with clusterProfiler package [25],which used overrepresentation enrichment analysis (ORA)to identify enriched KEGG pathways, GO terms, andimmune cells. The gene markers for immune cells wereextracted from the previously published study [26]. Thethreshold for these gene sets was P value < 0.05.

    2.6. Cox Regression Proportional Hazard Model-BasedSurvival Analysis. Cox regression proportional hazard modelwas used to evaluate the differences of overall survivalbetween patients with two conditions, which was imple-mented in R programming software survival package withcoxph function. To visualize the overall survival for eachgroup, we used Kaplan-Meier curves to estimate the survivalprobability.

    3. Results

    3.1. Identification of Differentially Expressed Genes in HCCTumors and Healthy Tissues. To uncover the dysregulatedgenes associated with HCC, we compared the gene expres-sion profiles between tumor tissues and normal tissue adja-cent to the tumor (NAT). From the HCC gene expressionprofiles, a total of 15,186 genes were identified (fragmentper kilobase million, FPKM > 1 in more than 20% samples),while the number of protein coding genes (PCGs) andlncRNA genes significantly varied between tumor tissuesand NAT, as more PCGs and lncRNAs were observed intumor tissues compared with NAT (Wilcoxon rank-sum test,P < 0:05) (Figures 1(a) and 1(b)). Moreover, we observedquite dissimilar patterns regarding gene expressions betweenHCC tumor tissues and NAT (Figure 1(c), adjusted P value <0.05 and log2 fold change > 1 or < −1), and identified 1,631upregulated and 1,515 downregulated genes. Hierarchicalclustering analysis was performed to further visualize expres-sion patterns of the differentially expressed genes (DEGs),suggesting that there was a great difference between thesetwo groups (Figure 1(d)).

    3.2. Biological Interpretation of Differentially Expressed GeneSets Utilizing GO and KEGG-Based Enrichment Analysis.To investigate the dysregulated signaling pathways andbiological processes, we performed GSEA analysis of theDEGs based on using gene sets of KEGG and GO data-bases. KEGG-based enrichment analysis revealed thatupregulated genes exhibited significant enrichment inpathways regarding cell division, cell replication, and otherbiological processes related to cell cycle, while downregulatedgenes were mainly involved in metabolic and catabolicprocesses (Figure 2(a)). GO enrichment analysis further con-firmed our observations as upregulated genes were signifi-cantly enriched in cell cycle, DNA replication, andribosome, while terms including metabolic pathways, fattyacid degradation, chemical carcinogenesis, and PPAR signal-ing pathway were significantly enriched of the downregu-lated genes (Figure 2(b)). Based on the KEGG and GOenrichment analysis, we observed that cell cycle andmetabolism-related pathways or biological processes wereup- or downregulated in HCC, suggesting that cell prolifera-tion was hyperactivated and metabolic capability of liver wassignificantly decreased in HCC.

    3.3. Coexpression Network Analysis of the DEGs. In order toassess the biological importance of these DEGs and the corre-lation patterns among them, weighted gene coexpression net-work analysis (WGCNA) was carried out. We chose soft

    2 BioMed Research International

    https://www.ncbi.nlm.nih.gov/srahttps://www.ncbi.nlm.nih.gov/srahttp://www.genome.ucsc.edu/

  • power value 9 as it reflected the best scale independence andmean connectivity (Figure 3(a)). With this selected softpower, similarity matrices were calculated, and hierarchical

    clustering of these DEGs based on this dissimilarity measurewas performed. Five modules were detected with DynamicTree Cut algorithm and distinguished by different colors

    7000

    Nontumor Tumor

    Num

    ber o

    f pro

    tein

    -cod

    ing

    gene

    s8000

    9000

    10000

    11000

    P = 2.33e–7

    (a)

    Nontumor Tumor

    600

    Num

    ber o

    f Inc

    RNA

    gen

    es

    800

    1000

    1200

    P = 0.0014

    (b)

    –50

    50

    –Log

    10 (a

    djus

    ted P

    val

    ue)

    100

    Tumor vs. normal

    Downregulated genesUpregulated genes

    0

    Log2 (FC)

    5 10

    (c)

    –3

    –2

    –1

    0

    1

    2

    3

    Tissues

    Tissues

    NontumorTumor

    TTis

    (d)

    Figure 1: The overview of the protein-coding genes, lncRNAs, and differentially expressed genes. The distribution of number of protein-coding genes and lncRNAs was illustrated in (a) and (b). (c) The differentially expressed genes (DEGs) were represented by the pointswith colors red (upregulation) and green (downregulation). (d) The expression patterns of DEGs in tumor and nontumor tissues.

    3BioMed Research International

  • (Figure 3(b)). After obtaining these module gene listings, theGO enrichment analysis was performed to interpret eachmodule’s biological functions. Except for a few shared terms,these modules exhibited little similarity in functions(Figure 3(c)). Genes in module 1 (blue, 325 genes in total)were mainly associated with ribosome, peroxisome, andPPAR signaling pathway, module 2 (brown, 101 genes intotal) dealt with Toll-like receptor signaling pathway, salmo-nella infection, and phagosome, while module 5 (yellow, 103genes in total) consisted of genes concerned with focal adhe-sion, ECM-receptor interaction, and PI3K-Akt signalingpathway. Modules 3 and 4 (denoted in grey and turquoise,including 286 and 384 genes, respectively) were both associ-ated with retinol metabolism, metabolism of xenobiotics bycytochrome P450, drug metabolism-cytochrome P450, andchemical carcinogenesis. Specifically, module 2 (brown) wascharacterized by dysregulation of tumor microenvironment,such as ECM-receptor interaction and focal adhesion, whilemodule 3 was enriched by the metabolism-related pathways.

    Moreover, we also found that cell proliferation-related path-ways, such as cell cycle, DNA replication, and Fanconi ane-mia pathway, were enriched by the genes of module 4.Taken together, these modules were recognized as biologi-cally meaningful in HCC patients.

    3.4. Identification of Infiltrated Immune Cells in HCC Tissues.As the immune cells were infiltrated into the tumor tissues[27], we next investigated the infiltrated levels of the immunecells for the HCC tissues based on their marker gene sets. Weperformed gene set enrichment analysis (GSEA) to test theenrichment degree of the differentially expressed markergenes for each WGCNA module. Notably, two immune cells,namely, cytotoxic cells and macrophage, were enriched inmodules grey and brown, respectively, while T helper cell-2(Th2) was enriched in module turquoise (Figure 4(a)). Spe-cifically, the marker genes of cytotoxic cells and macrophagewere upregulated in HCC, and the marker genes of Th2 weredownregulated in HCC, suggesting that cytotoxic cells and

    Mitotic cell cycleMitotic cell cycle process

    Cell cycleCell cycle process

    Cell divisionChromosome organization

    Nuclear divisionMitotic nuclear division

    Cell cycle phase transitionMitotic cell cycle phase transition

    Organelle fissionChromosome segregation

    DNA metabolic processRegulation of cell cycle

    Cellular component organization or biogenesisOrganelle organization

    Nuclear chromosome segregationCellular component organization

    DNA replicationMitotic sister chromatid segregation

    2000 400 600

    P.adjust1e–42

    1e–21

    2e–21

    3e–21

    4e–21

    Small molecule metabolic processOrganic acid metabolic process

    Carboxylic acid metabolic processOxoacid metabolic process

    Organic acid catabolic processCarboxylic acid catabolic process

    Monocarboxylic metabolic processDurg metabolic process

    Oxidation–reduction processSmall molecule catabolic process

    Lipid metabolic processSmall molecule biosynthetic process

    Organic acid biosynthetic processCarboxylic acid biosynthetic process

    Fatty acid metabolic processAlpha–amino acid metabolic process

    Cellular response to xenobiotic stimulusCellular amino acid catabolic process

    Cellular amino acid metabolic processResponse to xenobiotic stimulus

    1e–42

    1e–21

    2e–21

    3e–21

    4e–21

    Small molecule metabolic processOrganic acid metabolic process

    Carboxylic acid metabolic processOxoacid metabolic process

    Organic acid catabolic processCarboxylic acid catabolic process

    Monocarboxylic metabolic processDurg metabolic process

    Oxidation–reduction processSmall molecule catabolic process

    Lipid metabolic processSmall molecule biosynthetic process

    Organic acid biosynthetic processCarboxylic acid biosynthetic process

    Fatty acid metabolic processAlpha–amino acid metabolic process

    Cellular response to xenobiotic stimulusCellular amino acid catabolic process

    Cellular amino acid metabolic processResponse to xenobiotic stimulus

    P.adjust1e–79

    1e–31

    3e–31

    4e–31

    6e–310 100 200 300

    (a)

    Cell cycleDNA replication

    RibosomeSystemic lupus erythematosus

    Pathogenic Escherichia coli infectionBiosynthesis of amino acids

    Fanconi anemia pathwayViral carcinogenesis

    Pyrimidine metabolismp53 signaling pathway

    ECM–receptor pathwayFructose and mannose metabolism

    Small cell lung cancerHuman papillomavirus infection

    Drug metabolism – other enzymesMismatch repair

    Protein digestion and absorptionCellular senescence

    Oocyte meiosisGap junction

    Metabolic pathwaysRetinol metabolism

    Chemical carcinogenesisDrug metabolism – cytochrome P450

    Fatty acid degradationMetabolism of xenobiotics by cytochrome P450

    Complement and coagulation cascadesPPAR signaling pathwayTryptophan metabolism

    Valine, leucine and isoleucine degradationSteroid hormone biosynthesis

    Glycine, serine and threonine metabolismButanoate metabolism

    PeroxisomeCarbon metabolism

    Bile secretionDrug metabolism – other enzymes

    Arginine biosynthesisAlanine, aspartate and glutamate metabolism

    Tyrosine metabolism

    P.adjust

    4.0e–07

    8.0e–07

    1.2e–06

    1.6e–06

    Metabolic pathwaysRetinol metabolism

    Chemical carcinogenesisDrug metabolism – cytochrome P450

    Fatty acid degradationMetabolism of xenobiotics by cytochrome P450

    Complement and coagulation cascadesPPAR signaling pathwayTryptophan metabolism

    g g p yg g p

    Valine, leucine and isoleucine degradationSteroid hormone biosynthesis

    Glycine, serine and threonine metabolismButanoate metabolism

    PeroxisomeCarbon metabolism

    Bile secretionDrug metabolism – other enzymes

    Arginine biosynthesisAlanine, aspartate and glutamate metabolism

    Tyrosine metabolism

    P.adjust

    0.02

    0.04

    0.06

    0 10 20 30 40 0 50 100 150 200

    (b)

    Figure 2: The GO biological processes and KEGG pathways enriched by the differentially expressed genes. (a) The GO biological processesenriched by the DEGs. The bars on the left and right represent the enriched GO terms enriched by the upregulated and downregulated genes,respectively. (b) The enriched KEGG pathways by the DEGs. The up- and downregulated genes were enriched in the pathways represented bythe bars on the left and right, respectively.

    4 BioMed Research International

  • 5

    –0.2Scal

    e-fre

    e top

    olog

    y m

    odel

    fit,

    signe

    d R

    2

    0

    200

    400

    600

    800

    Mod

    el co

    nnec

    tivity

    0.0

    0.2

    0.4

    0.6

    0.8

    10Soft threshold (power)

    Scale independence Mean connectivity

    15 20 5 10Soft threshold (power)

    15 20

    1

    1

    2

    3

    4 56

    7 89101112 14 16 18 20

    2

    34

    5 6 7 8 9101112 14 16 18 20

    (a)

    DynamicTree Cut

    0.4

    0.6

    Hei

    ght

    0.8

    1.0Gene dendrogram and module colors

    (b)

    Ribosome

    Peroxisome

    Primary bile acid biosynthesis

    PPAR signaling pathway

    Alanine, aspartate and glutamate metabolism

    Valine, leucine and isoleucine degradation

    Toll–like receptor signaling pathway

    Salmonella infection

    Phagosome

    Leishmaniasis

    Fc gamma R–mediated phagocytosisFocal adhesion

    ECM–receptor interation

    Chemical carcinogenesis

    Drug metabolism – cytochrome P450

    Metabolism of xenobiotics by cytochrome P450

    Retinol metabolism

    Cell cycle

    DNA replication

    Fanconi anemia pathway

    Systemic lupus erythematosus

    PI3K–Akt signaling pathway

    Malaria

    Cytokine–cytokine receptor interaction

    Blue(325)

    Brown(101)

    Grey(286)

    Turquoise(384)

    Yellow(103)

    0.04Gene ratio

    0.08

    0.12

    0.16

    P.adjust

    0.01

    0.02

    0.03

    0.04

    (c)

    Figure 3: The weighted gene coexpression network analysis (WGCNA) of the DEGs. (a) The scale independence and mean connectivity usedfor the selection of soft power. (b) The hierarchical clustering analysis of the DEGs based on the TOM similarity. (c) The KEGG pathwaysenriched by the genes of WGCNA modules.

    5BioMed Research International

  • Th2 cells

    Macrophages

    Cytotoxic cells

    Blue Brown Grey Turquoise Yellow

    Gene ratio

    0.30.20.1 P < 0.05

    (a)

    Tissues

    Tissues

    Gene expression

    TN

    BIRC5 3

    2

    1

    0

    –1

    –2

    –3

    CDC7CENPFCDC25CWDHD1RORAZBTB16CTSWKLRK1CD68GM2A

    Tissues GeBIRC5CDC7CENPFCDC25CWDHD1RORAZBTB16CTSWKLRK1CD68GM2A

    (b)

    Figure 4: Continued.

    6 BioMed Research International

  • macrophage were highly infiltrated in HCC, while the Th2cells were reduced in HCC tissues as compared with nontu-mor tissues. Moreover, the marker genes of the threeimmune cell types, including BIRC5, CDC7, CENPF,CDC25C, WDHD1, RORA, ZBTB16, CTSW, KLRK1,CD68, and GM2A, were observed to be dysregulated inHCC (Figure 4(b)). Correlation analysis revealed that markergenes within each immune cell were highly correlated witheach other, suggesting that they could cooperate with eachother to function in immune cell (Figure 4(c)). In addition,we also observed that the markers of cytotoxic cells and mac-rophages exhibited higher correlation, indicating that the twocell types may have interactions in HCC tissues.

    3.5. Identification of Critical Hub-lncRNAs and Evaluationof Their Prognostic Power in HCC Patients. To better sum-marize the functional roles of each module in HCC, it isimportant to recognize the intramodular interactions andrepresentative genes in a coexpression network. Thus, usingCytoscape [28], we visualized the interaction networks ofthese genes based on their coexpression and uncoveredhub-lncRNAs for each module, which may resemble func-tional importance (Figure 5(a)). We successfully identifiedSNHG3 in the blue module, LINC00152 in the brown mod-ule, TMEM220-AS1 and CTC-297N7.9 in the turquoisemodule, and RP11-286H15.1 in the yellow module as hub-lncRNAs. Notably, SNHG3 and LINC00152 were previouslyreported to function as competing endogenous RNA or reg-ulate essential pathways to promote tumorigenesis [29, 30].

    For each hub-lncRNA, samples were divided into high- andlow-expression groups based on the expression of this hub-lncRNA. Utilizing the survival data of HCC patients in corre-sponding high- and low-expression groups from TCGALIHC (liver hepatocellular carcinoma) datasets, Kaplan-Meier curves were plotted for each hub-lncRNA, and signif-icant differences in overall survival were observed betweenhigh- and low-expression groups (P < 0:05, Figure 5(b)).These findings not only suggested that the identification ofhub-lncRNA based on coexpression network could uncoverlncRNAs with critical function but also revealed that thesehub-lncRNAs had the power of evaluating prognostic out-comes in HCC patients.

    4. Discussion

    Hepatocellular carcinoma (HCC) is a primary liver cancerassociated with a growing incidence and extremely high mor-tality. However, the pathogenic mechanism is still not fullyunderstood. In the present study, we compared the geneexpression profiles between tumor tissues and NATs andidentified 1,631 upregulated and 1,515 downregulated genes.GSEA was subsequently performed to investigate the dysreg-ulated signaling pathways and biological processes, whichrevealed that DEGs exhibited significant enrichment in cellcycle and metabolism-related pathways or biological pro-cesses. The hyperactivated cell cycle and metabolism-related pathways indicated that uncontrolled tumor cell

    BIRC5

    Pearson correlation 1

    0.5

    0

    –0.5

    –1

    CDC7

    CENPF

    CDC25C

    WDHD1

    RORA

    ZBTB16

    CTSW

    KLRK1

    CD68

    GM2A

    BIRC

    5

    CDC7

    CEN

    PF

    CDC2

    5C

    WD

    HD

    1

    RORA

    ZBTB

    16

    CTSW

    KLRK

    1

    CD68

    GM

    2A

    Immune cellsCytotoxic cellsMacropahgesTh2

    BIRC5

    Pea

    CDC7

    CENPF

    CDC25C

    WDHD1

    RORA

    ZBTB16

    CTSW

    KLRK1

    CD68

    GM2A

    (c)

    Figure 4: The immune cells aberrantly infiltrated in HCC tissues. (a) The three immune cell types enriched by the module genes. (b) Theexpression patterns of the immune cell-related marker genes in tumor and nontumor tissues. (c) The Pearson correlation coefficientsbetween the markers within specific immune cell type or between the immune cells.

    7BioMed Research International

  • (a)

    Figure 5: Continued.

    8 BioMed Research International

  • proliferation and decreased metabolic capability may be thehallmark of HCC [31–33].

    In order to assess the biological importance of theseDEGs and the correlation patterns among them, weightedgene coexpression network analysis (WGCNA) was carriedout. Five modules were detected with Dynamic Tree Cutalgorithm (Figure 3(b)). After obtaining these module genelistings, the GO enrichment analysis was performed to inter-pret each module’s biological functions. Except for a fewshared terms, these modules exhibited little similarity infunctions (Figure 3(c)). Specifically, the module 2 (brown)was characterized by dysregulation of tumor microenviron-ment, such as ECM-receptor interaction and focal adhesion,while module 3 was enriched by the metabolism-relatedpathways. Moreover, we also found that cell proliferation-related pathways, such as cell cycle, DNA replication, andFanconi anemia pathway, were enriched by the genes ofmodule 4. Taken together, these modules were recognizedas biologically meaningful in HCC patients. In accordancewith the characteristics of HCC subtypes [34–36], the path-ways or biological processes characterized for the three mod-ules may also be associated with the signatures of previousHCC subtypes. To further characterize the features of theWGCNA modules, we further tested the enrichment degreeof the differentially expressed marker genes of immune cellsfor each WGCNA module. Notably, two immune cells,namely, cytotoxic cells and macrophage, were enriched inmodules grey and brown, respectively, while T helper cell-2(Th2) was enriched in module turquoise (Figure 4(a)). Eventhough cytotoxic cells and macrophages were highly infil-

    trated in HCC tissues, their immune activities were sup-pressed, indicating that the immune checkpoint inhibitorssuch as PD1/PDL1 and CTLA-4/B7-1/B7-2 may function inHCC tissues [37, 38]. Consistently, CTLA4 was highlyexpressed in HCC (P < 0:05).

    Among the network constructed by WGCNA, SNHG3 inthe blue module, LINC00152 in the brown module,TMEM220-AS1 and CTC-297N7.9 in the turquoise module,and RP11-286H15.1 in the yellow module were identified ashub-lncRNAs. SNHG3 and LINC00152 were previouslyreported to function as competing endogenous RNA or reg-ulate essential pathways to promote tumorigenesis [29, 30].Survival analysis of these hub-lncRNAs revealed that thesehub-lncRNAs were closely associated with the HCC overallsurvival (P < 0:05, Figure 5(b)). These findings suggested thatcoexpression network analysis could uncover lncRNAs withfunctional importance, which may be associated with prog-nostic outcomes of HCC patients.

    In addition, the present study also has some limitations.First, the relationship between suppressed activities of cyto-toxic cells and macrophages and CTLA4 and the functionalroles of the hub-lncRNAs should be further validated byexperiments. Second, the prognostic values of the hub-lncRNAs should be validated in independent datasets. Insummary, this study demonstrated that network-based anal-ysis could identify some functional modules and some hub-lncRNAs, which may be critical for HCC pathogenesis orprogression.

    In summary, this study demonstrated that network-based analysis could identify some functional modules and

    High-expressionLow-expression

    0

    P = 0.00068

    SNHG3 (blue)

    TMEM220-AS1 (turquoise) CTC-297N7.9 (turquoise) RP11-286H15.1 (yellow)

    LINC00152 (brown)

    0.00

    0.25

    Surv

    ival

    pro

    babi

    lity

    0.50

    0.75

    1.00

    1000 2000Time

    3000 4000 0

    P < 0.0001

    0.00

    0.25

    Surv

    ival

    pro

    babi

    lity

    0.50

    0.75

    1.00

    1000 2000Time

    3000 4000

    0

    P < 0.0001

    0.00

    0.25

    Surv

    ival

    pro

    babi

    lity

    0.50

    0.75

    1.00

    1000 2000Time

    3000 4000 0

    P < 0.0001

    0.00

    0.25

    Surv

    ival

    pro

    babi

    lity

    0.50

    0.75

    1.00

    1000 2000Time

    3000 4000 0

    P < 0.0001

    0.00

    0.25

    Surv

    ival

    pro

    babi

    lity

    0.50

    0.75

    1.00

    1000 2000Time

    3000 4000

    (b)

    Figure 5: The hub-lncRNAs in WGCNA network and their prognostic association with HCC overall survival. (a) The visualization of theWGCNA modules with hub-lncRNAs by Cytoscape. (b) The KM curves of the hub-lncRNAs in TCGA-LIHC cohort.

    9BioMed Research International

  • some hub-lncRNAs, which may be critical for HCC patho-genesis or progression.

    Data Availability

    We collected RNA sequencing data of 50 HCC and 50paired nontumor tissues from Sequence Read Archive(SRA, https://www.ncbi.nlm.nih.gov/sra) database with anaccession number SRP068976.

    Conflicts of Interest

    The authors declare that they have no conflict of interest.

    Authors’ Contributions

    JJ is the guarantor of integrity of the entire study; JL studythe concepts; JL study the design; JL is responsible for thedefinition of intellectual content; JZ is for the literatureresearch; SK is for the clinical studies; SK is for the exper-imental studies; CW is for the data acquisition; CW is forthe data analysis; CW is for the statistical analysis; DW isfor the manuscript preparation; DW is for the manuscriptediting; and JJ is for the manuscript review.

    Acknowledgments

    This work was funded by the National Natural Science Foun-dation of China (grant no. 81760567), Key R&D Program ofShandong Province (grant no. GG201809200094), andNatural Science Foundation of Inner Mongolia AutonomousRegion of China (grant no. 2018MS08114).

    References

    [1] M. S. Grandhi, A. K. Kim, S. M. Ronnekleiv-Kelly, I. R. Kamel,M. A. Ghasebeh, and T.M. Pawlik, “Hepatocellular carcinoma:from diagnosis to treatment,” Surgical Oncology, vol. 25, no. 2,pp. 74–85, 2016.

    [2] J. Hartke, M. Johnson, and M. Ghabril, “The diagnosis andtreatment of hepatocellular carcinoma,” Seminars in Diagnos-tic Pathology, vol. 34, no. 2, pp. 153–159, 2017.

    [3] D. Dimitroulis, C. Damaskos, S. Valsami et al., “From diagno-sis to treatment of hepatocellular carcinoma: an epidemicproblem for both developed and developing world,” WorldJournal of Gastroenterology, vol. 23, no. 29, pp. 5282–5294,2017.

    [4] J. Zucman-Rossi, A. Villanueva, J. C. Nault, and J. M. Llovet,“Genetic landscape and biomarkers of hepatocellular carci-noma,” Gastroenterology, vol. 149, no. 5, pp. 1226–1239.e4,2015.

    [5] A. Wojcicka, M. Swierniak, O. Kornasiewicz et al., “Next gen-eration sequencing reveals microRNA isoforms in liver cirrho-sis and hepatocellular carcinoma,” The International Journalof Biochemistry & Cell Biology, vol. 53, pp. 208–217, 2014.

    [6] S. R. Selitsky, T. A. Dinh, C. L. Toth et al., “Transcriptomicanalysis of chronic hepatitis B and C and liver cancer revealsmicroRNA-mediated control of cholesterol synthesis pro-grams,” mBio, vol. 6, no. 6, pp. e01500–e01515, 2015.

    [7] F. Fang, R. M. Chang, L. Yu et al., “MicroRNA-188-5p sup-presses tumor cell proliferation and metastasis by directly tar-

    geting FGF5 in hepatocellular carcinoma,” Journal ofHepatology, vol. 63, no. 4, pp. 874–885, 2015.

    [8] Y. Du, G. Kong, X. You et al., “Elevation of highly up-regulatedin liver cancer (HULC) by hepatitis B virus X protein promoteshepatoma cell proliferation via down-regulating p18,” TheJournal of Biological Chemistry, vol. 287, no. 31, pp. 26302–26311, 2012.

    [9] L. Dyck and K. H. G. Mills, “Immune checkpoints and theirinhibition in cancer and infectious diseases,” European Journalof Immunology, vol. 47, no. 5, pp. 765–779, 2017.

    [10] N. Rohr-Udilova, F. Klinglmuller, R. Schulte-Hermann et al.,“Deviations of the immune cell landscape between healthyliver and hepatocellular carcinoma,” Scientific Reports, vol. 8,no. 1, 2018.

    [11] T. A. Barnes and E. Amir, “HYPE or HOPE: the prognosticvalue of infiltrating immune cells in cancer,” British Journalof Cancer, vol. 117, no. 4, pp. 451–460, 2017.

    [12] M. Garnelo, A. Tan, Z. Her et al., “Interaction betweentumour-infiltrating B cells and T cells controls the progressionof hepatocellular carcinoma,” Gut, vol. 66, no. 2, pp. 342–351,2017.

    [13] R. Leinonen, H. Sugawara, and M. Shumway, “Internationalnucleotide sequence database, the sequence read archive,”Nucleic acids research, vol. 39, pp. D19–D21, 2016.

    [14] G. Liu, G. Hou, L. Li, Y. Li, W. Zhou, and L. Liu, “Potentialdiagnostic and prognostic marker dimethylglycine dehydroge-nase (DMGDH) suppresses hepatocellular carcinoma metas-tasis in vitro and in vivo,” Oncotarget, vol. 7, no. 22,pp. 32607–32616, 2016.

    [15] D. Kim, B. Langmead, and S. L. Salzberg, “HISAT: a fastspliced aligner with low memory requirements,” NatureMethods, vol. 12, no. 4, pp. 357–360, 2015.

    [16] M. Pertea, G. M. Pertea, C. M. Antonescu, T. C. Chang, J. T.Mendell, and S. L. Salzberg, “StringTie enables improvedreconstruction of a transcriptome from RNA-seq reads,”Nature Biotechnology, vol. 33, no. 3, pp. 290–295, 2015.

    [17] J. Harrow, A. Frankish, J. M. Gonzalez et al., “GENCODE: thereference human genome annotation for The ENCODE Pro-ject,” Genome research, vol. 22, no. 9, pp. 1760–1774, 2012.

    [18] M. I. Love, W. Huber, and S. Anders, “Moderated estimationof fold change and dispersion for RNA-seq data with DESeq2,”Genome Biology, vol. 15, no. 12, 2014.

    [19] W. S. Noble, “How does multiple testing correction work?,”Nature Biotechnology, vol. 27, no. 12, pp. 1135–1137, 2009.

    [20] C. Gu, X. Shi, Z. Huang et al., “A comprehensive study of con-struction and analysis of competitive endogenous RNA net-works in lung adenocarcinoma,” Biochimica et BiophysicaActa (BBA)-Proteins and Proteomics, vol. 1868, no. 8, article140444, 2020.

    [21] P. Langfelder and S. Horvath, “WGCNA: an R package forweighted correlation network analysis,” BMC Bioinformatics,vol. 9, no. 1, 2008.

    [22] M. Kanehisa and S. Goto, “KEGG: kyoto encyclopedia of genesand genomes,” Nucleic Acids Research, vol. 28, no. 1, pp. 27–30, 2000.

    [23] TheGeneOntologyConsortium, “The gene ontology resource:20 years and still GOing strong,”Nucleic acids research, vol. 47,no. D1, pp. D330–D338, 2019.

    [24] X. Shi, T. Huang, J. Wang et al., “Next-generation sequencingidentifies novel genes with rare variants in total anomalous

    10 BioMed Research International

    https://www.ncbi.nlm.nih.gov/sra

  • pulmonary venous connection,” eBioMedicine, vol. 38,pp. 217–227, 2018.

    [25] G. Yu, L. G. Wang, Y. Han, and Q. Y. He, “clusterProfiler: an Rpackage for comparing biological themes among gene clus-ters,” OMICS, vol. 16, no. 5, pp. 284–287, 2012.

    [26] G. Bindea, B. Mlecnik, M. Tosolini et al., “Spatiotemporaldynamics of intratumoral immune cells reveal the immunelandscape in human cancer,” Immunity, vol. 39, no. 4,pp. 782–795, 2013.

    [27] Y. Kurebayashi, H. Ojima, H. Tsujikawa et al., “Landscape ofimmune microenvironment in hepatocellular carcinoma andits additional impact on histological and molecular classifica-tion,” Hepatology, vol. 68, no. 3, pp. 1025–1041, 2018.

    [28] 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.

    [29] P. Chen, X. Fang, B. Xia, Y. Zhao, Q. Li, and X. Wu, “Longnoncoding RNA LINC00152 promotes cell proliferationthrough competitively binding endogenous miR-125b withMCL-1 by regulating mitochondrial apoptosis pathways inovarian cancer,” Cancer Medicine, vol. 7, no. 9, pp. 4530–4541, 2018.

    [30] W. Huang, Y. Tian, S. Dong et al., “The long non-coding RNASNHG3 functions as a competing endogenous RNA to pro-mote malignant development of colorectal cancer,” OncologyReports, vol. 38, no. 3, pp. 1402–1410, 2017.

    [31] Q. Huang, Y. Tan, P. Yin et al., “Metabolic characterization ofhepatocellular carcinoma using nontargeted tissue metabolo-mics,” Cancer Research, vol. 73, no. 16, pp. 4992–5002, 2013.

    [32] T. Masaki, Y. Shiratori, W. Rengifo et al., “Hepatocellular car-cinoma cell cycle: study of Long-Evans cinnamon rats,” Hepa-tology, vol. 32, no. 4, pp. 711–720, 2000.

    [33] A. P. Sutter, K. Maaser, M. Hopfner, A. Huether, D. Schuppan,and H. Scherubl, “Cell cycle arrest and apoptosis induction inhepatocellular carcinoma cells by HMG-CoA reductase inhib-itors. Synergistic antiproliferative action with ligands of theperipheral benzodiazepine receptor,” Journal of hepatology,vol. 43, no. 5, pp. 808–816, 2005.

    [34] The Cancer Genome Atlas Research Network, “Comprehen-sive and integrative genomic characterization of hepatocellularcarcinoma,” Cell, vol. 169, no. 7, pp. 1327–1341.e23, 2017.

    [35] Y. Hoshida, S. M. Nijman, M. Kobayashi et al., “Integrativetranscriptome analysis reveals common molecular subclassesof human hepatocellular carcinoma,” Cancer Research,vol. 69, no. 18, pp. 7385–7392, 2009.

    [36] Chinese Human Proteome Project (CNHPP) Consortium,Y. Jiang, A. Sun et al., “Proteomics identifies new therapeutictargets of early-stage hepatocellular carcinoma,” Nature,vol. 567, no. 7747, pp. 257–261, 2019.

    [37] M. Kudo, “Immune checkpoint inhibition in hepatocellularcarcinoma: basics and ongoing clinical trials,” Oncology,vol. 92, no. 1, pp. 50–62, 2017.

    [38] F. Xu, T. Jin, Y. Zhu, and C. Dai, “Immune checkpoint therapyin liver cancer,” Journal of Experimental & Clinical CancerResearch, vol. 37, no. 1, 2018.

    11BioMed Research International

    Network-Based Coexpression Analysis Identifies Functional and Prognostic Long Noncoding RNAs in Hepatocellular Carcinoma1. Introduction2. Materials and Methods2.1. Data Collection2.2. Read Mapping and Gene Expression Quantification2.3. Differential Expression Analysis2.4. Weighted Gene Coexpression Network Analysis (WGCNA)2.5. KEGG, GO, and Immune Cell-Based Overrepresentation Enrichment Analysis2.6. Cox Regression Proportional Hazard Model-Based Survival Analysis

    3. Results3.1. Identification of Differentially Expressed Genes in HCC Tumors and Healthy Tissues3.2. Biological Interpretation of Differentially Expressed Gene Sets Utilizing GO and KEGG-Based Enrichment Analysis3.3. Coexpression Network Analysis of the DEGs3.4. Identification of Infiltrated Immune Cells in HCC Tissues3.5. Identification of Critical Hub-lncRNAs and Evaluation of Their Prognostic Power in HCC Patients

    4. DiscussionData AvailabilityConflicts of InterestAuthors’ ContributionsAcknowledgments