13
Research Article Identification of Potential Prognostic Biomarkers for Breast Cancer Based on lncRNA-TF-Associated ceRNA Network and Functional Module Xinrong Li , Junquan Zhu , and Jian Qiu Department of Integrative Medicine & Medical Oncology, Shengzhou Peoples Hospital (The First Aliated Hospital of Zhejiang University Shengzhou Branch), 312400, Shengzhou, Zhejiang, China Correspondence should be addressed to Jian Qiu; [email protected] Received 30 April 2020; Revised 23 June 2020; Accepted 29 June 2020; Published 29 July 2020 Academic Editor: David A. McClellan Copyright © 2020 Xinrong 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. Breast cancer leads to most of cancer deaths among women worldwide. Systematically analyzing the competing endogenous RNA (ceRNA) network and their functional modules may provide valuable insight into the pathogenesis of breast cancer. In this study, we constructed a lncRNA-TF-associated ceRNA network via combining all the signicant lncRNA-TF ceRNA pairs and TF-TF PPI pairs. We computed important topological features of the network, such as degree and average path length. Hub nodes in the lncRNA-TF-associated ceRNA network were extracted to detect dierential expression in dierent subtypes and tumor stages of breast cancer. MCODE was used for identifying the closely connected modules from the ceRNA network. Survival analysis was further used for evaluating whether the modules had prognosis eects on breast cancer. TF motif searching analysis was performed for investigating the binding potentials between lncRNAs and TFs. As a result, a lncRNA-TF-associated ceRNA network in breast cancer was constructed, which had a scale-free property. Hub nodes such as MDM4, ZNF410, AC0842-19, and CTB-89H12 were dierentially expressed between cancer and normal sample in dierent subtypes and tumor stages. Two closely connected modules were identied to signicantly classify patients into a low-risk group and high-risk group with dierent clinical outcomes. TF motif searching analysis suggested that TFs, such as NFAT5, might bind to the promoter and enhancer regions of hub lncRNAs and function in breast cancer biology. The results demonstrated that the synergistic, competitive lncRNA-TF ceRNA network and their functional modules played important roles in the biological processes and molecular functions of breast cancer. 1. Introduction Breast cancer is one of the most common female cancers worldwide, which is also the second leading cause of female cancer death [1]. Adjuvant therapy has been an eective way to improve patient survival and promote the quality of life [2]. However, tumor metastasis and drug resistance are still a concern during breast cancer therapy. Thus, there is an urgent need to identify key biomarkers and uncover potential molecular mechanisms for breast cancer diagnosis and therapy. Many studies have identied some important genes that participated in the occurrence, development, and metastasis of breast cancer. For exam- ple, two well-known cancer genes, BRCA1 and BRCA2, were the major genes associated with the genetic etiology of breast cancer. Women with BRCA1/BRCA2 mutations had very high risk to develop breast cancer [3]. Mutations or variants of other genes such as TP53, ATM, BARD1, CHECK2, FGFR2, GSTM1, and MAP3K1 have also been reported to increase the risk of breast cancer [4]. Long noncoding RNAs (lncRNAs) are a type of RNA transcript of more than 200 nucleotides, which have been considered eective disease biomarkers in cancers [5]. Abnormal expression of several lncRNAs has been shown to be involved in breast cancer. For example, lncRNA HOTAIR was overexpressed and acted as a powerful predic- tor of metastasis in breast cancer [6]. The depletion of lncRNA MALAT1 decreased the tumorigenesis and Hindawi BioMed Research International Volume 2020, Article ID 5257896, 13 pages https://doi.org/10.1155/2020/5257896

Identification of Potential Prognostic Biomarkers for

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Research ArticleIdentification of Potential Prognostic Biomarkers for BreastCancer Based on lncRNA-TF-Associated ceRNA Network andFunctional Module

Xinrong Li , Junquan Zhu , and Jian Qiu

Department of Integrative Medicine & Medical Oncology, Shengzhou People’s Hospital (The First Affiliated Hospital of ZhejiangUniversity Shengzhou Branch), 312400, Shengzhou, Zhejiang, China

Correspondence should be addressed to Jian Qiu; [email protected]

Received 30 April 2020; Revised 23 June 2020; Accepted 29 June 2020; Published 29 July 2020

Academic Editor: David A. McClellan

Copyright © 2020 Xinrong 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.

Breast cancer leads to most of cancer deaths among women worldwide. Systematically analyzing the competing endogenousRNA (ceRNA) network and their functional modules may provide valuable insight into the pathogenesis of breast cancer. Inthis study, we constructed a lncRNA-TF-associated ceRNA network via combining all the significant lncRNA-TF ceRNA pairsand TF-TF PPI pairs. We computed important topological features of the network, such as degree and average path length.Hub nodes in the lncRNA-TF-associated ceRNA network were extracted to detect differential expression in different subtypesand tumor stages of breast cancer. MCODE was used for identifying the closely connected modules from the ceRNA network.Survival analysis was further used for evaluating whether the modules had prognosis effects on breast cancer. TF motifsearching analysis was performed for investigating the binding potentials between lncRNAs and TFs. As a result, alncRNA-TF-associated ceRNA network in breast cancer was constructed, which had a scale-free property. Hub nodes suchas MDM4, ZNF410, AC0842-19, and CTB-89H12 were differentially expressed between cancer and normal sample indifferent subtypes and tumor stages. Two closely connected modules were identified to significantly classify patients into alow-risk group and high-risk group with different clinical outcomes. TF motif searching analysis suggested that TFs, suchas NFAT5, might bind to the promoter and enhancer regions of hub lncRNAs and function in breast cancer biology. Theresults demonstrated that the synergistic, competitive lncRNA-TF ceRNA network and their functional modules playedimportant roles in the biological processes and molecular functions of breast cancer.

1. Introduction

Breast cancer is one of the most common female cancersworldwide, which is also the second leading cause offemale cancer death [1]. Adjuvant therapy has been aneffective way to improve patient survival and promotethe quality of life [2]. However, tumor metastasis and drugresistance are still a concern during breast cancer therapy.Thus, there is an urgent need to identify key biomarkersand uncover potential molecular mechanisms for breastcancer diagnosis and therapy. Many studies have identifiedsome important genes that participated in the occurrence,development, and metastasis of breast cancer. For exam-ple, two well-known cancer genes, BRCA1 and BRCA2,

were the major genes associated with the genetic etiologyof breast cancer. Women with BRCA1/BRCA2 mutationshad very high risk to develop breast cancer [3]. Mutationsor variants of other genes such as TP53, ATM, BARD1,CHECK2, FGFR2, GSTM1, and MAP3K1 have also beenreported to increase the risk of breast cancer [4].

Long noncoding RNAs (lncRNAs) are a type of RNAtranscript of more than 200 nucleotides, which have beenconsidered effective disease biomarkers in cancers [5].Abnormal expression of several lncRNAs has been shownto be involved in breast cancer. For example, lncRNAHOTAIR was overexpressed and acted as a powerful predic-tor of metastasis in breast cancer [6]. The depletion oflncRNA MALAT1 decreased the tumorigenesis and

HindawiBioMed Research InternationalVolume 2020, Article ID 5257896, 13 pageshttps://doi.org/10.1155/2020/5257896

metastasis of breast cancer [7]. lncRNA AGAP2-AS1 couldpromote breast cancer cell growth by upregulating theexpression of MyD88 and activating the NF-κB signalingpathway [8]. In addition to these important functions inbreast cancer, many recent studies have reported thatlncRNAs might interact with mRNAs, competitively bindto their common microRNAs (miRNAs), and then functionas competing endogenous RNAs (ceRNAs) [9]. TheceRNA-related network could link the functions of lncRNAs,miRNAs, and mRNAs. Dysfunction of these molecules in thenetwork was highly related to the occurrence and develop-ment of human diseases, including breast cancer [10].

Although a single gene can function in the study of path-ogenesis, detection of individual gene expression can still notpromote the overall understanding of human diseases [11].Recently, the application of biological networks for identify-ing biomarkers and understanding cancer biology hasbecome increasingly urgent [12]. Networks specific to diseasecontext could help in improving the understanding of theunderlying biology from a global perspective [13]. Transcrip-tion factors (TFs) are a kind of genes that could function inthe regulation of gene expression via binding to their DNAregulatory elements, such as promoters or enhancers [14].The miRNAs, TFs, and the mRNAs or lncRNAs regulatedby them could be integrated for constructing global regula-tory networks. More intriguingly, network module centralityanalysis provided more information to understand biologicalproblems [15]. However, some regulatory patterns such aslncRNA-TF interactions in breast cancer remainedunknown. More important molecular mechanisms underly-ing breast cancer still need more comprehensive molecularand biological studies.

In the present study, we are working to construct alncRNA-TF-associated ceRNA network for revealing theirpotential interaction in breast cancer using bioinformaticstools. This network contained TFs, lncRNAs, and theirinteractions based on ceRNAs and protein-protein interac-tions (PPIs). First, we performed a comprehensive analysisof the network and computed important topological fea-tures, such as degree and average path length. Hub nodeswith the highest degrees in the lncRNA-TF-associatedceRNA network were selected to detect differential expres-sion in different subtypes/tumor stages of breast cancer.Then, closely connected modules were identified fromthe lncRNA-TF-associated ceRNA network. Survival analy-sis was performed to evaluate whether the modules hadprognosis effects on breast cancer. Furthermore, in orderto investigate the binding potential between TFs andlncRNAs, we performed TF motif searching to indicatethe promoter and enhancer regions of lncRNAs. In con-clusion, our study could help explain the biological pro-cesses and molecular mechanism of breast cancer from aglobal network perspective.

2. Materials and Methods

2.1. Breast Cancer-Related Datasets. We downloaded breastcancer-related gene expression profile from TCGA (https://xenabrowser.net/datapages/) and converted transcript-level

RNA-seq data into lncRNA/protein-coding gene-levelRNA-seq data using GENCODE (https://www.gencodegenes.org/human/) [16]. TFs that we obtainedbefore were further mapped to the protein-coding gene-level RNA-seq data. Data preprocessing and log transforma-tion were performed to these RNA-seq data. Finally, breastcancer-related lncRNA/TF RNA-seq expression profiles wereobtained. These data involved 1,215 samples with clinicalinformation. All the raw expression data are supported inSupplementary Tables S1 and S2.

2.2. Construct a lncRNA-TF-Associated ceRNA Network.Based on ceRNA theory, we comprehensively analyzelncRNA and TF RNA-seq expression profiles of breast cancerand constructed a lncRNA-TF-associated ceRNA network.First, we downloaded all the miRNA-mRNA interactionsthat were curated from StarBase (http://starbase.sysu.edu.cn/), which contained 386 miRNAs and 13,861 mRNAs(supported in Supplementary Table S3). The miRNA-TFinteractions were further extracted by mapping TFs to themRNAs obtained previously. In addition, we used miRandatools for identifying significant miRNA-lncRNAinteractions by inputting lncRNA and miRNA sequences(default parameters) [16]. Second, we counted the numberof the shared miRNAs for each lncRNA-TF pair based onthe miRNA-TF interactions and miRNA-lncRNAinteractions and indicate the shared miRNAs withstatistical significance for all the lncRNA-TF pairs usinghypergeometric test. The lncRNA-TF pairs with thethreshold of hypergeometric test p value < 0.05 wereconsidered statistically significant (Supplementary Table S4).

Third, using breast cancer-related lncRNA and TF-levelRNA-seq expression profiles, Pearson correlation coefficients(PCC) were further calculated for those lncRNA-TF pairswith a hypergeometric test p value < 0.05. And thelncRNA-TF pairs with PCC > 0:6were finally considered sig-nificant lncRNA-TF pairs (Supplementary Table S5).

In addition, TF-related PPI interactions were extractedfrom the HPRD database. Then, a breast cancer-relatedlncRNA-TF ceRNA network was formed by combining allthe significant lncRNA-TF pairs and TF-TF PPI pairs (Sup-plementary Table S6).

2.3. Identify Closely Connected Network Modules. We usedthe Molecular Complex Detection (MCODE) plug-in inCytoscape to identify closely connected modules from thelncRNA-TF-associated ceRNA network. The MCODEalgorithm is based on graph-theoretical analysis, whichclusters a given network by topology for finding denselyconnected regions [17]. The criteria that we used for iden-tifying functional modules were as follows: MCODEscores > 5, degree cutoff = 2, node score cutoff = 0:2, maxdepth = 100, and k − score = c2.

2.4. Survival Analysis. Our gene expression profile contained1,215 breast cancer patients with clinical information. Sub-type classification is defined from TCGA clinical matrix.Based on these data, the univariate Cox regression was usedto identify breast cancer-related prognostic signatures. We

2 BioMed Research International

accumulated the regression coefficient and the expressionvalues of each gene and computed the risk score of eachpatient as follows:

Risk score = 〠n

i=1riExp ið Þ, ð1Þ

where n is the number of genes in a gene set, ri is the regres-sion coefficient of gene i, and ExpðiÞ is the expression value ofgene i for a corresponding patient.

We classified breast cancer patients into two groups byusing the mean risk score as a cutoff. That is, patients withthe risk score greater than the mean value were classifiedinto a high-risk group. Patients with the risk score lessthan the mean value were classified into a low-risk group.These high-risk group and low-risk group patients werethen used to perform Kaplan-Meier survival analysis.Log-rank test with a p value < 0.05 was used to generatestatistical significance. The raw TCGA clinical matrix issupported in Supplementary Table S7.

2.5. TF Motif Searching Analysis. For investigating the bind-ing potential between TFs and lncRNAs, we performed TFmotif searching analysis to the promoter and enhancerregions of lncRNAs. Promoters were defined as +/-2 kb fromtranscription start site (TSS). Enhancers were downloadedfrom FANTOM5 [18, 19]. FIMO was used to scan promoterand enhancer regions with a p value < 1e–4 [20].

3. Results

3.1. Construction of a lncRNA-TF-Associated ceRNANetwork. lncRNAs that contained miRNA-response ele-ments could competitively bind miRNAs with mRNAsand then function as ceRNAs to participate in multiplebiological processes of complex diseases. In this study, alncRNA-TF-associated ceRNA network in breast cancerwas constructed by combining all significant lncRNA-TFceRNA pairs and TF-TF PPI pairs (Figure 1, details inmethods). This network consisted of 164 lncRNA nodes,91 TF nodes, and 644 edges (Figure 2(a)). To evaluatethe importance of network nodes, we performed topologi-cal analyses for the lncRNA-TF-associated ceRNA network(Supplementary Table S8). First, we computed the degreeof network nodes and found that all the nodes followeda power law distribution, which indicated that thenetwork had the scale-free property (Figure 2(b), R2 =0:94). Next, we calculated the average path length of thelncRNA-TF-associated ceRNA network. Simultaneously,we also chose 1,000 degree-conserved random networksto calculate their average path length and counted thenumber of average path length in a random networkshorter than that in the real network. p values werecalculated by the number divided by 1,000. The resultshowed that the average path length of the real networkwas significantly shorter than that of random networks(Figure 2(c), p < 0:01). These results suggested that hubgenes of the lncRNA-TF-associated ceRNA networkplayed important roles in the local region of the network.

3.2. Detection of Breast Cancer-Related Hub Genes. Numer-ous studies found that genes connected by a large numberof other genes (also known as high degree) in biologicalnetwork tended to play vital roles in pathological pro-cesses. These genes with high degree in network weredefined as hub genes. Here, we detected breast cancer-related hub genes from the lncRNA-TF ceRNA network.We defined the genes with top 10% node degree as hubgenes, including 14 TFs and 11 lncRNAs (Figure 3(a)).Results showed that these hubs not only had high degreesbut also had high betweenness, closeness, and low shortestpath length, indicating that these genes might maintainthe basic biological processes in cancer pathology(Figure 3(a)). We further extracted the hub-hub subnet-work from the lncRNA-TF-associated ceRNA network.As a result, the hub-hub subnetwork was composed ofall these hubs and their 103 edges, including the knowncancer-related lncRNAs MALAT1 and XIST(Figure 3(b)). Then, we tested the prognosis effects ofthe 14 hub TFs. Results showed that hazard ratios of theseTFs in breast cancer of TCGA BRCA cohorts were notsignificant (Figure 3(c)). However, in luminal A subtype,these hub TFs showed a strong prognosis effect(Figure 3(d)). These results inspired us to investigate thefunction of hub genes in subtypes of breast cancer.

The results mentioned above showed that ourlncRNA-TF-associated ceRNA network had the scale-freeproperty, representing a small subset of high-degree nodes(also called hubs) that were connected by the most ofother nodes. Thus, we selected 2 TF hubs (MDM4 andZNF410) and 2 lncRNA hubs (AC084219 and CTB-89H12) with the highest degrees from the lncRNA-TF-associated ceRNA network and detected their expressionin various subtypes of breast cancer. The results showedthat they could significantly be distinguished betweenbreast cancer samples and normal samples (Figures 4(a)–4(d)) in different subtypes. Actually, MDM4 has beenemerging as an important breast cancer biomarker andoncoprotein [21]. MDM4 was found highly expressed notonly in normal breast epithelial cells but also in mostluminal breast cancer [22]. MDM4 has also been suggestedto promote triple-negative breast cancer metastasis [23].Cancer cells and stromal/immune cells, such as cancer-associated fibroblasts, were the important parts of tumormicroenvironment. ZNF410, also known as APA-1, was aTF that regulated the expression of matrix-remodelinggenes during fibroblast senescence [24]. Du et al. haveshown the tumor-suppressive role of lncRNA CTB-89H12and the expression regulation ability of PTEN in prostatecancer [25]. The above studies suggested that hub nodesin global lncRNA-TF network might play important rolesin biological processes and molecular functions of breastcancer. We further calculated the expression of the twolncRNAs (AC084219 and CTB-89H12) in different tumorstages and found that they were differentially expressedin the advanced stage of tumor (Figure 4(e)).

3.3. Identification of Closely Connected Network Modules.Biological networks are often too large to interpret the

3BioMed Research International

biological phenomena accurately. Functional modules of anetwork may be more useful for reflecting the relevant bio-logical importance. Functional modules have been widelyapplied to explore the mechanism involved in various biolog-ical processes, such as miRNA regulation, disease occurrence,and drug action [26]. We used “MCODE” in the Cytoscapesoftware to identify closely connected network modules fromour lncRNA-TF-associated ceRNA network. As a result, twoclosely connected modules linked to breast cancer wereidentified.

Module 1 was composed of 43 nodes (26 lncRNAs and 17TFs) and 120 edges (Figure 5(a)). Some lncRNAs and TFs ofmodule 1 have been reported to function in breast cancer.For example, MDM4 negatively regulated the major tumorsuppressor gene p53 and further modulated stress responses,which had been considered a biomarker that may drivemetastasis and progression of breast cancer [27]. The alteredexpression ofDMTF1 proteins was highly related to the path-ophysiology of cancer. In response to oncogenic stresses,DMTF1 bind to the promoter of ARF and governed theARF-p53 tumor suppressor pathway activity [28]. LncRNAPURA was an evolutionarily conserved cellular protein par-ticipating in processes of DNA replication, transcription,and RNA transport, which functioned in human cancer[29]. To evaluate whether module 1 had prognosis effectson luminal A breast cancer, we calculated linear combinationof expression values of lncRNAs/TFs in module 1 weighted

by the regression coefficient of univariate Cox regression toperform survival analysis. As a result, we significantly classi-fied luminal A breast cancer patients into low-risk group andhigh-risk group with different clinical outcomes(Figure 5(b)).

Module 2 was composed of 36 nodes (29 lncRNAs and 7TFs) and 45 edges (Figure 5(c)). We also found that severallncRNAs and TFs in module 2 were highly associated withbreast cancer.MATR3 was a highly conserved nuclear matrixprotein, which was widely expressed in various tissues andinvolved in breast cancer-related biological processes, suchas transcription, translation, RNA processing, DNA replica-tion, apoptosis, and chromatin remodeling [30]. Axitinib, aclinically approved drug, could effectively treat cancerpatients with aberrant activity of nuclear β-catenin. The E3ubiquitin ligase SHPRH was identified as the direct target ofaxitinib. Treatment with axitinib stabilized SHPRH andincreased the ubiquitination and degradation of β-catenin[31]. Furthermore, we also calculated linear combination ofexpression values of lncRNAs/TFs in module 2 weighted bythe regression coefficient of univariate Cox regression inorder to evaluate whether module 2 had prognosis effectson luminal A and luminal B breast cancer. As shown inFigure 5(d), luminal A and luminal B breast cancer patientswere significantly classified into low-risk group and high-risk group with different clinical outcomes, respectively.These results suggested that the integration of lncRNAs and

. . .

. . .

. . .

. . .

. . .

. . .

. . .

. . .

. . .

. . .

miRNA-lncRNA pairs

miRNA-TF pairs

Hypergeometric test Pearson correlation

ceRNA pairs

Step

1St

ep 2

Step

3

Breast cancer Expression matrix(TCGA BRCA)

HPRD PPI

Merged network

lncRNAmiRNAmRNA

Figure 1

4 BioMed Research International

YAF2

CTD-2516F10

ZNF571

RP11-148B6

AC007566

RP11-797A18RP11-730K11

RP11-267N12

RP3-323N1

MDM4 RP11-549J18

RP11-518L10

RP1-102H19

MEG8

RP11-29G8 DLEU2

CTD-2574D22ZNF283

AC005154

RP11-415J8AC084219

CTD-2270L9RP11-174G6

KCNQ1OT1RP1-59D14 BAZ2BRP11-227G15

RP5-1085F17

RP11-492E3RP11-382A18

ZNF91MALAT1

RP4-717I23

ZNF382

RP1-5O6

RP11-676J12

ZNF564ZBTB37

RP11-701H24

AF131215

RP11-206L10

AC003104 RORA

ZNF236

RP11-10O17CTD-2561J22 DMTF1

CTC-459F4

CTD-3064H18

ZNF124

RP1-20N2

AP001172

BDP1

ZNF483REL

AC007036RP11-119K6AC005519

PIAS2

ZNF417

RP11-1006G14 MATR3

GS1-251I9AC093375

RP11-715F3

CTD-3252C9

NEAT1

NFAT5

HMBOX1

ZNF117

AC000120

ZNF493

CTD-2017D11

RP11-37B2

RP11-65L3

ZNF451RP11-21A7A

RP11-902B17

RP11-509J21ERCC6

RP11-18F14

RP11-57H14

SHPRH RP11-658F2

RP11-378J18

RP11-64K12

RP4-714D9

ZNF586SCARNA9

AC016683

RP11-761E20

RP5-837J1

RP11-229P13

AC007038

RP11-552M11

ELK4ZNF519

RP4-665N4KB-318B8

RP11-819C21

RP11-119F19

CTA-217C2

RP4-614O4

TFEC

KB-431C1

RP5-991G20

CTD-2047H16

MIR155HG

ZNF546

ZNF235

RUSC1-AS1

ZNF510

FENDRRRP11-452L6HOTAIR AC034220RP11-141O11

RP11-197P3

HOXD11ZNF747 FOXF1GABPB2 ANKRA2HOXC11

RP11-383J24

RP11-923I11 RP11-21J18

XXbac-B461K10

AP001469

AC058791

RP11-425I13

FOXP2CTD-2353F22

ZBTB20

CTD-2622I13RP11-33N16 RP11-290D2

AP000662

CTD-3138B18

AP000525

MIR17HG

RP11-566E18

APC

ZNF566

CHD2RP11-73M18

RP11-445F12ZNF573

ZNF81

ZNF141

RP4-802A10

AATFRP11-478C19

RP11-540O11

ATM

CTB-102L5

NR2C2 CTD-2083E4RP11-397D12

HOXD3XIST

RP11-258C19HCG18

PURA

RP11-115C21CTB-89H12

RP11-158H5

RP11-342K6ZNF248

MAGI1-IT1RP1-184J9

RP11-457M11

AC078852AC009299

ZNF410RBM15

ZNF445

RP11-181E10RP11-264L1

RP11-174G17RP11-429D19

GS1-124K5RP11-43D2

RUNX1T1

ZDHHC21

RP11-752G15

RP11-46A10

RP11-231E4

RP11-282K24

RP11-151N17

BRWD1

MEIS2

RP11-5L12

AP000265

RP3-368A4

RP11-102F4

RP11-79N23

RP5-837I24

RP11-421L21

RP11-35G9

HCP5HCG11 SNHG7CTB-92J24RP11-405O10 A1BG-AS1

HMGA2RP11-126O1

AC092535

RP11-154J22MEG3

RP11-227D2

KLF11 STAT1RREB1ZNF254 ZNF324 REXO4

NR3C2RP11-276H19

ARID5B

HOTAIRM1

HOXA5

HOXA4HOXA3

HOXA-AS2

HABP4

ERGRP11-175K6

EMX2OS

TEAD1KLF4

EBF3

POU2F2

RP11-284N8

IRF4

PRDM1

POU2AF1

EDF1

EZH1 EPAS1

RP1-193H18PPARG

AC113189

ZNF524 CEBPA

EOMES

CTC-524C5

RP11-331F9

ZNF26

GFI1

HCLS1 CTC-308K20

RP4-564F22

RNU12

(a)

Figure 2: Continued.

5BioMed Research International

TFs in our functional modules had significant prognosiscapability and could be used as prognostic signatures ofbreast cancer.

3.4. Identification of Core lncRNA-TF Crosstalks. TFs maycontrol the activity of lncRNAs via binding to the DNAregulatory elements of lncRNAs. In this study, we con-ducted motif searching to the promoter and enhancerregions of lncRNAs for investigating the binding potentialbetween TFs and lncRNAs. The results showed multipleTF binding sites in the promoters and enhancers oflncRNAs, respectively (Figures 6(a) and 6(b)). For exam-ple, NFAT5 has been implicated in cancer cell proliferationand invasion [32]. In this study, NFAT5 had ceRNA rela-tionships with lncRNAs under the threshold of hypergeo-metric test p value < 0.05 and PCC > 0:6, which werefurther validated to have multiple motifs binding in thepromoters of lncRNAs (Figure 6(a)).

Because hub genes often play more important roles inthe biological network, we focused on the motif searchingresults of top 20% hub lncRNAs in our breast cancer-related lncRNA-TF ceRNA network. Those lncRNA-TFpairs with TFs binding in the promoters and enhancersof top 20% hub lncRNAs were extracted to form a newnetwork (Figure 6(c)). That is, lncRNA nodes and TFnodes in this network had not only significant ceRNArelationships but also strong motif binding. The resultsimplied that TFs might bind to the promoter andenhancer regions of important hub lncRNAs and form“feedback loops” to function in cancer biology. The resultsof KEGG pathway enrichment showed that TFs of the net-work were associated with basal functions, such as “Thy-roid hormone signaling pathway,” “Hepatitis B,”“Transcriptional misregulation in cancer,” “Pathways incancer,” and “Cell cycle” (Figure 6(d)). These pathwayswere all demonstrated to be closely associated with breastcancer [33–35]. For example, breast cancer patients duringor after chemotherapy were found to have a remarkableclinical problem of hepatitis B virus [36]. In normal cells,thyroid hormones could regulate the normal physiologicalprocesses. However, once signaling pathways became dys-regulated, thyroid hormones would induce cancer cell pro-

liferation [37]. Insulin resistance that attenuated biologicalresponse to insulin circulation was reported to be associ-ated with a series of pathological conditions and someendocrine tumors, including breast cancer [38]. All theseresults showed that TFs could crosstalk with lncRNAsvia binding to the promoter and enhancer regions oflncRNAs, which were involved in breast cancer-relatedbiological processes and molecular functions.

4. Discussion

Breast cancer is accountable for the plurality of cancerdeaths among women worldwide. Metastatic breast canceris even considered an incurable disease with poor prog-nosis [39]. There is an urgent need to investigate themolecular mechanism and find the significant risk factorsfor diagnosis and prognosis of breast cancer. The ceRNAregulation may represent a widespread layer of gene reg-ulation which is important for pathogenesis such asbreast cancer [40]. Systematically analyzing the lncRNA-related ceRNA network may provide valuable insight intothe function of lncRNAs and the molecular mechanismof diseases. Thus, in this study, we constructed a globallncRNA-TF network for revealing their potential interac-tion in breast cancer using bioinformatics tools. This net-work was constructed by combining all significantlncRNA-TF ceRNA pairs and TF-TF PPI pairs. First, wemade a comprehensive analysis of the network and com-puted important topological features, such as degree andaverage path length. We found that all the nodesfollowed power law distribution and average path lengthof the real network was substantially shorter than thatof random networks. We selected hub nodes with thehighest degrees in the global lncRNA-TF network andfound that they could significantly distinguish betweentumor samples of different subtypes/tumor stages andnormal samples. The literature evidences further sug-gested the importance of hub nodes in the globallncRNA-TF network. Then, two closely connected mod-ules containing some hub genes such as MDM4, DMTF1,RORA, and MATR3 were identified from the globallncRNA-TF network, which represented significant different

Degree of genes

Num

ber o

f gen

es

10

10

100

11

R2 = 0.94

(b)

3.2 3.4 3.6 3.8 4.0 4.2

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

Den

sity

Average path length

Observed: 3.45p < 0.01

(c)

Figure 2

6 BioMed Research International

MDM4 55 0.17 0.4 2.48MATR3 50 0.16 0.4 2.52ZNF410 44 0.14 0.38 2.62

HMBOX1 41 0.1 0.38 2.62ZBTB20 39 0.15 0.39 2.6SHPRH 29 0.09 0.35 2.83ZNF26 25 0.06 0.35 2.85

DMTF1 22 0.04 0.35 2.84BAZ2B 22 0.02 0.35 2.84NFAT5 21 0.04 0.36 2.82ZNF91 21 0.02 0.34 2.96RORA 20 0.01 0.32 3.12

AC084219 20 0.05 0.39 2.55ZNF236 16 0.02 0.34 2.95

CTB-89H12 16 0.03 0.39 2.55MALAT1 16 0.03 0.39 2.57

ZNF124 15 0.02 0.31 3.2HCG18 15 0.03 0.39 2.56

RP3-368A4 15 0.03 0.36 2.75RP11-206L10 14 0.04 0.38 2.6

AC000120 14 0.03 0.37 2.73RP11-342K6 13 0.08 0.39 2.59KCNQ1OT1 13 0.02 0.38 2.63

RP11-540O11 13 0.02 0.36 2.75XIST 13 0.09 0.38 2.66

TFlncRNA

Deg

ree

Betw

eenn

ess

Clos

enes

s

Shor

test

path

High

Low

(a)

ZBTB20

KCNQ1OT1

MATR3

CTB-89H12

ZNF91

SHPRHZNF410

AC000120

RP11-540O11

MDM4

RORA

RP11-206L10

ZNF236 ZNF26

RP11-342K6

XIST MALAT1

HCG18

NFAT5

HMBOX1

BAZ2B

RP3-368A4

DMTF1

ZNF124

AC084219

(b)

MDM4

MATR3

ZNF410

HMBOX1

ZBTB20

SHPRH

ZNF26

DMTF1

BAZ2B

NFAT5

ZNF91

RORA

ZNF236

ZNF124

BRCA

−0.10

−0.05

0.00

0.05

0.10Log10(HR)Hazard ratio

(c)

0 50 100 150 200 250

0.0

0.2

0.4

0.6

0.8

1.0

Overall survival

Months

Perc

ent s

urvi

val

Logrank p = 0.047HR(high) = 1.7p(HR) = 0.05n(high) = 206n(low) = 206

Luminal Asubtype

Low signature groupHigh signature group

(d)

Figure 3

7BioMed Research International

1.5

2.0

2.5

3.0

3.5

4.0

4.5

1.5

2.0

2.5

3.0

3.5

4.0

4.5

BRCA(num(T) = 135; num(N) = 112)

2

3

4

5

6

2

3

4

5

6

BRCA

(a)

(b)

(c)

(d)

(e)

(num(T) = 66; num(N) = 112)

2

3

4

5

6

2

3

4

5

6

BRCA(num(T) = 415; num(N) = 112)

2.0

2.5

3.0

3.5

4.0

4.5

5.0

2.0

2.5

3.0

3.5

4.0

4.5

5.0

BRCA(num(T) = 194; num(N) = 112)

Her2 Luminal A Luminal BBasal/triple negative

Expr

essio

n−lo

g 2(TP

M+1

)M

DM

4

Expr

essio

n−lo

g 2(TP

M+1

)

Expr

essio

n−lo

g 2(TP

M+1

)

Expr

essio

n−lo

g 2(TP

M+1

)

TumorNormal

⁎⁎

3.5

4.0

4.5

5.0

5.5

6.0

6.5

3.5

4.0

4.5

5.0

5.5

6.0

6.5

BRCA

3.5

4.0

4.5

5.0

5.5

6.0

3.5

4.0

4.5

5.5

6.0

BRCA

3.5

4.0

4.5

5.0

5.5

6.0

6.5

7.0

3.5

4.0

4.5

5.0

5.5

6.0

6.5

7.0

BRCA

3

4

5

6

7

3

4

5

6

7

BRCA

ZNF4

10

Expr

essio

n−lo

g 2(TP

M+1

)

Expr

essio

n−lo

g 2(TP

M+1

)

Expr

essio

n−lo

g 2(TP

M+1

)

Expr

essio

n−lo

g 2(TP

M+1

)

⁎⁎

BRCA BRCA BRCA BRCA

0

1

2

3

4

0

1

2

3

4

0.0

0.5

1.0

1.5

2.0

2.5

3.0

0.0

0.5

1.0

1.5

2.0

2.5

3.0

AC0

8421

9

Expr

essio

n−lo

g 2(TP

M+1

)

Expr

essio

n−lo

g 2(TP

M+1

)

0

1

2

3

4

0

1

2

3

4

Expr

essio

n−lo

g 2(TP

M+1

)

0

1

2

3

Expr

essio

n−lo

g 2(TP

M+1

)

⁎ ⁎

BRCA BRCA BRCA BRCA

1.0

1.5

2.0

2.5

3.0

3.5

4.0

4.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

4.5

1

2

3

4

5

1

2

3

4

5

CTB-

89H

12

Expr

essio

n−lo

g 2(TP

M+1

)

1

2

3

4

5

1

2

3

4

5

Expr

essio

n−lo

g 2(TP

M+1

)

Expr

essio

n−lo

g 2(TP

M+1

)

1.0

1.5

2.0

2.5

3.0

3.5

4.0

4.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

4.5

Expr

essio

n−lo

g 2(TP

M+1

) ⁎ ⁎ ⁎ ⁎

0.0

0.2

0.4

0.6

0.8

1.0

1.2 F value = 2.04

Stage I Stage II Stage III Stage IV Stage X Stage I Stage II Stage III Stage IV Stage X

1

2

3

4

5 F value = 3.67

AC084219 CTB-89H12

Exp

Exp

Figure 4

8 BioMed Research International

RP11-290D2

RORA

ZNF141

RP11-151N17

ZNF493

RP11-540O11

ZNF117

RP1-20N2

AC000120

RP3-368A4

BRWD1

RP11-258C19

DMTF1AC003104

RP1-102H19

BDP1

MDM4

CTD-2270L9

RP11-518L10

ATM

RP11-478C19

CTB-89H12 RP11-148B6

ZBTB37AC084219

MALAT1

PURA

RP11-549J18PIAS2

RP11-115C21

RP11-282K24

AP001172

CTD-2574D22

MAGI1-IT1KCNQ1OT1

BAZ2B

ZNF91

RP11-126O1AATF

ZNF283

RP11-445F12

ZNF586RP11-154J22

(a)

Luminal A

0 50 100 150 200 2500.0

0.2

0.4

0.6

0.8

1.0Overall survival

Months

Perc

ent s

urvi

val

Low signature groupHigh signature group

Logrank p = 0.034HR(high) = 1.8p(HR) = 0.037n(high) = 206n(low) = 206

(b)

RP11-119K6

RP11-5L12

MATR3 GS1-251I9RP11-1006G14

RP11-65L3

ZNF483

ZNF417

RP11-33N16

RP11-715F3

KB-318B8

ZNF235

CTA-217C2AC093375

RP11-819C21

CTD-3252C9RP4-717I23

CTD-2047H16

ZDHHC21

RP11-229P13

AC005519

RP11-378J18

RP4-665N4

AC007036

GS1-124K5

RP5-837J1

ZNF519

RP11-10O17

SHPRHRP11-174G6

AC016683

RP11-552M11

RP11-227G15

RP11-18F14

RP11-761E20

RP5-991G20

(c)

0 50 100 150

0.0

0.2

0.4

0.6

0.8

1.0Overall survival

Months

Perc

ent s

urvi

val

Logrank p = 0.016HR(high) = 0.42p(HR) = 0.018n(high) = 95n(low) = 95

0.0

0.2

0.4

0.6

0.8

1.0Overall survival

Perc

ent s

urvi

val

Logrank p = 0.015HR(high) = 2p(HR) = 0.016n(high) = 206n(low) = 206

Luminal A

Luminal B

0 50 100 150 200 250Months

Low signature groupHigh signature group

(d)

Figure 5

9BioMed Research International

clinical outcomes between the breast cancer patients in thelow-risk group and high-risk group classified by thesurvival analysis.

Here, as a point of innovation, we identified somesubtype-specific prognosis factors in breast cancer. InFigure 3(c), results showed that TFs have the weak prognosis

ATM

BDP1

EOMES

HMBOX1

IRF4

NFAT5

NR2C2

POU2F2

PRDM1

PURA

REL

RORA

ZNF410

lncRNAslog10(motif_num + 0.1)

TF

Promoters

0.0

0.2

0.4

0.6

0.8cor_value

−1.0−0.50.0

0.51.0

(a)

Enhancers

BDP1

EOMES

HMBOX1

IRF4

NFAT5

NR2C2

POU2F2

PRDM1

PURA

RORA

TFEC

ZNF410

TF0.0

0.2

0.4

0.6

0.8cor_value

lncRNAslog10(motif_num + 0.1)

−1.0−0.5

0.0

0.51.0

1.5

(b)

IRF4

PRDM1

RP11-284N8TFEC

EOMES

POU2F2

RP11-206L10

XIST

ATM

RP11-342K6

PURA

NR2C2

HCG18

NFAT5

RP11-73M18RP11-148B6

BDP1RP3-368A4

AC084219 RP5-1085F17

RP11-258C19

RP11-540O11

AC007038RP11-549J18

AC003104 REL

MALAT1RP11-151N17

HMBOX1

AC005154

KCNQ1OT1

CTD-2270L9ZNF410

RORA

CTB-89H12

PromoterEnhancer

(c)

−log10(q−value)

KEG

G p

athw

ays

0 1 2 3 4 5

Adipocytokine signaling pathway

Insulin resistanceHomologous recombinationMeaslesNF-kappa B signaling pathwayChronic myeloid leukemiaViral carcinogenesis

Cell cycleHerpes simplex infectionHTLV−I infection

Transcriptional misregulation in cancerPathways in cancer

Hepatitis BThyroid hormone signaling pathway

(d)

Figure 6

10 BioMed Research International

effects on panbreast cancer. However, combining these fac-tors showed a strong prognosis effect in the luminal A sub-type, which indicated that these crucial genes have animportant clinical value in luminal A breast cancer. As previ-ously mentioned, MDM4, which is a negative regulator ofp53, not only played crucial roles in regulation of normalbreast development but also contributed to the relapsingand metastasis of breast cancer. Intriguingly,MDM4 was sig-nificantly overexpressed in the luminal A subtype of breastcancer [41]. Thus, several anticancer therapeutic strategiessuch as SAR405838 [42], DS-3032b [43], and ALRN-6924[44] were explored with the purpose of restoring the normalactivity of p53. As for the DMTF1 in module 1, Tian et al.found thatDMTF1β, a major subtype ofDMTFs, was overex-pressed in breast cancer tissues and promotes tumorigenesisin a transgenic mouse model [45]. Niklaus et al. indicated thecisplatin resistance of breast cancer cells is associated withexpression of DMTF1-β by using SKBR3 (cisplatin sensitive)andMCF7 (cisplatin resistant) breast cancer cell lines in vitro[46]. Maglic et al. demonstrated that overexpression ofDMTF1β was associated with poor clinical outcomes, byexamining the expression ofDMTF1β in the cancer and adja-cent tissue from twenty breast cancer patients, which sug-gested that DMTF1β could be considered a potentialdiagnostic index for patients with breast cancer [47]. Whenit comes to RORA, viewed as a member of the circadiangenes, it could disrupt endogenous homeostasis and therebypromote endocrine tumor development and accelerate pro-gression resulting from the dysfunction of this gene [48].Taheri et al. found that one functional polymorphism(rs4774388) of RORA was associated with breast cancer riskafter performing a comparative analysis between the breastcancer patients and the healthy persons in Iran [49]. Besides,Du and Xu [50] observed that RORA suppressed the expres-sion of malignant phenotypes in breast cancer cell lines bothin vitro and in vivo, which indicated that RORA could beconsidered an ideal potential diagnostic biomarker and ther-apeutic target of breast cancer. In the module 2, MATR3,known as a vital pathogenic gene of amyotrophic lateral scle-rosis, is still poorly understood in the process of canceriza-tion [51]. Just a few studies were conducted; for example,Yang et al. performed Western blot and RNA immunopre-cipitation assay to find that the lncRNA SNHG1 was directlyinteracted with MATR3 to promote neuroblastoma progres-sion [52]. Nho et al. observed the “Licochalcone H” couldsuppress cell viability and induce apoptosis in human oralsquamous cell lines by suppression ofMATR3 [53]. In short,previous articles indicated that the hub genes involved in thetwo modules showed a variety of physiological and patholog-ical functions in breast cancer as an integrated interactionnetwork including lncRNAs and TFs, which have significantprognosis capability and could be used as prognostic signa-tures of breast cancer.

Furthermore, TF motif searching analysis was performedto demonstrated that TFs might bind to the enhancers orpromoters of important hub lncRNAs and form “feedbackloops” to participate in cancer biology. The enriched path-ways were shown to be closely associated with breast cancer;for example, the thyroid hormone signaling pathway ranked

as having the highest degree of enrichment. Numerous stud-ies were conducted to study the close relationship betweenthe thyroid hormone and breast cancer. Hercbergs et al. indi-cated that thyroid hormone promoted the proliferation of thebreast cells in vitro and breast cancer cases with hypothyroidfunction were less likely to be associated with lymph nodemetastases [54]. Søgaard et al. pointed out that hyperthyroid-ism was a risk factor for the incidence of breast cancer basedon a population-based cohort study [55]. Besides, the NF-kappa B signaling pathway ranked the top 10 signaling path-way in our analysis, indicating a vital role in the regulation ofbreast cancerization. Liu et al. showed that lncRNA NKILAcould block the phosphorylation of IκB in vitro and suppressthe metastasis of breast cancer by comparison of the differentexpressions of NKILA between the benign breast tissues andinvasive carcinomas [56].

In summary, we provided a comprehensive analysis ofbreast cancer-related lncRNA-TF ceRNA crosstalk. Theresults demonstrated that the synergistic, competitivelncRNA-TF pairs played important roles in pathological pro-cesses of breast cancer and had strong effect on the prognosisof breast cancer patients. Although our study showed valu-able results associated with breast cancer, there were stillsome limitations. First, we integrated hypergeometric testand PCC computed by gene expression profile to identify sig-nificant lncRNA-TF interactions. A stricter measure willdecrease false-positive rate and increase accuracy and reli-ability of our results. Second, we only used FANTOM5enhancer data to investigate the regulatory loops betweenTFs and lncRNA enhancers. If we can download the same-sample multiomics data from TCGA, the core lncRNA-TFfeedback loops would be more accurate. Third, in this study,we conducted a bioinformatics analysis to identify the crucialfactors in breast cancer; results indicated that some genes(TFs or lncRNAs) might play vital roles in the subtype can-cers. These results also encouraged us to validate the biolog-ical function and mechanism. In further study, we willconduct the biological experiments to investigate thesepotential factors. In a word, the identified lncRNAs andTFs in the global lncRNA-TF subnetwork and closely con-nected modules would provide important information forfurther breast cancer studies and be worth the experimentalvalidations.

Data Availability

The raw data used to support the findings of this study areavailable from the Supplementary Materials.

Conflicts of Interest

The authors declare that they have no conflicts of interest todisclose.

Authors’ Contributions

Jian Qiu is responsible for ensuring that the descriptions areaccurate and agreed by all authors. Jian Qiu designed this

11BioMed Research International

study. Xinrong Li and Junquan Zhu collected and processeddata. Xinrong Li wrote the manuscript.

Supplementary Materials

Supplementary 1. Supplementary Table S1: raw data for proc-essed mRNA expression from TCGA.

Supplementary 2. Supplementary Table S2: raw data for proc-essed lnc expression from TCGA.

Supplementary 3. Supplementary Table S3: raw data formiRNA-mRNA interactions from StarBase.

Supplementary 4. Supplementary Table S4: raw data forhypergeometric results between lncRNAs and TFs.

Supplementary 5. Supplementary Table S5: raw data for PCCresults between lncRNAs and TFs.

Supplementary 6. Supplementary Table S6: raw data for net-work interactions in Figure 2(a).

Supplementary 7. Supplementary Table S7: raw data for clin-ical information of TCGA breast cancer.

Supplementary 8. Supplementary Table S8: topology featureof genes of the lncRNA-TF-associated ceRNA network.

References

[1] C. E. DeSantis, J. Ma, A. Goding Sauer, L. A. Newman, andA. Jemal, “Breast cancer statistics, 2017, racial disparity inmortality by state,” CA: a Cancer Journal for Clinicians,vol. 67, no. 6, pp. 439–448, 2017.

[2] B. Weigelt, J. L. Peterse, and L. J. van’t Veer, “Breast cancermetastasis: markers and models,” Nature Reviews Cancer,vol. 5, no. 8, pp. 591–602, 2005.

[3] Y. Miki, J. Swensen, D. Shattuck-Eidens et al., “A strong candi-date for the breast and ovarian cancer susceptibility geneBRCA1,” Science, vol. 266, no. 5182, pp. 66–71, 1994.

[4] M. R. Stratton and N. Rahman, “The emerging landscape ofbreast cancer susceptibility,” Nature Genetics, vol. 40, no. 1,pp. 17–22, 2008.

[5] S. Jathar, V. Kumar, J. Srivastava, and V. Tripathi, “Technolog-ical developments in lncRNA biology,” Advances in Experi-mental Medicine and Biology, vol. 1008, pp. 283–323, 2017.

[6] R. A. Gupta, N. Shah, K. C. Wang et al., “Long non-codingRNA _HOTAIR_ reprograms chromatin state to promotecancer metastasis,” Nature, vol. 464, no. 7291, pp. 1071–1076, 2010.

[7] G. Arun, S. Diermeier, M. Akerman et al., “Differentiation ofmammary tumors and reduction in metastasis upon Malat1lncRNA loss,” Genes & Development, vol. 30, no. 1, pp. 34–51, 2016.

[8] H. Dong, W. Wang, S. Mo et al., “SP1-induced lncRNAAGAP2-AS1 expression promotes chemoresistance of breastcancer by epigenetic regulation of MyD88,” Journal of Experi-mental & Clinical Cancer Research, vol. 37, no. 1, p. 202, 2018.

[9] S. Ergun and S. Oztuzcu, “Oncocers: ceRNA-mediated cross-talk by sponging miRNAs in oncogenic pathways,” TumorBiology, vol. 36, no. 5, pp. 3129–3136, 2015.

[10] X. Qi, D. H. Zhang, N. Wu, J. H. Xiao, X. Wang, and W. Ma,“ceRNA in cancer: possible functions and clinical implica-

tions,” Journal of Medical Genetics, vol. 52, no. 10, pp. 710–718, 2015.

[11] X. Wang, S. Zhang, and H. Qian, “Discovery of significantpathways in breast cancer metastasis via module extractionand comparison,” IET Systems Biology, vol. 8, no. 2, pp. 47–55, 2014.

[12] A. L. Barabási, N. Gulbahce, and J. Loscalzo, “Network medi-cine: a network-based approach to human disease,” NatureReviews Genetics, vol. 12, no. 1, pp. 56–68, 2011.

[13] S. M. Hill, The HPN-DREAM Consortium, L. M. Heiser et al.,“Inferring causal molecular networks: empirical assessmentthrough a community-based effort,” Nature Methods, vol. 13,no. 4, pp. 310–318, 2016.

[14] C. J. Holtz-Heppelmann, A. Algeciras, A. D. Badley, and C. V.Paya, “Transcriptional regulation of the human FasLpromoter-enhancer region,” The Journal of Biological Chemis-try, vol. 273, no. 8, pp. 4416–4423, 1998.

[15] D. Vella, S. Marini, F. Vitali, D. Di Silvestre, G. Mauri, andR. Bellazzi, “MTGO: PPI network analysis via topologicaland functional module identification,” Scientific Reports,vol. 8, no. 1, p. 5499, 2018.

[16] A. Frankish, M. Diekhans, A. M. Ferreira et al., “GENCODEreference annotation for the human and mouse genomes,”Nucleic Acids Research, vol. 47, no. D1, pp. D766–d773, 2019.

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

[18] R. Andersson, The FANTOM Consortium, C. Gebhard et al.,“An atlas of active enhancers across human cell types and tis-sues,” Nature, vol. 507, no. 7493, pp. 455–461, 2014.

[19] The FANTOM Consortium and the RIKEN PMI and CLST(DGT), “A promoter-level mammalian expression atlas,”Nature, vol. 507, no. 7493, pp. 462–470, 2014.

[20] C. E. Grant, T. L. Bailey, andW. S. Noble, “FIMO: scanning foroccurrences of a given motif,” Bioinformatics, vol. 27, no. 7,pp. 1017-1018, 2011.

[21] M.Wade, Y.-C. Li, and G. M.Wahl, “MDM2, MDMX and p53in oncogenesis and cancer therapy,” Nature Reviews Cancer,vol. 13, no. 2, pp. 83–96, 2013.

[22] S. Haupt, D. Buckley, J. M. Pang et al., “Targeting Mdmx totreat breast cancers with wild-type p53,” Cell Death & Disease,vol. 6, no. 7, article e1821, 2015.

[23] C. Gao, G. Xiao, A. Piersigilli, J. Gou, O. Ogunwobi, andJ. Bargonetti, “Context-dependent roles of MDMX (MDM4)and MDM2 in breast cancer proliferation and circulatingtumor cells,” Breast Cancer Research, vol. 21, no. 1, p. 5, 2019.

[24] J. A. Benanti, D. K. Williams, K. L. Robinson, H. L. Ozer, andD. A. Galloway, “Induction of extracellular matrix-remodelinggenes by the senescence-associated protein APA-1,”Molecularand Cellular Biology, vol. 22, no. 21, pp. 7385–7397, 2002.

[25] Z. Du, T. Sun, E. Hacisuleyman et al., “Integrative analysesreveal a long noncoding RNA-mediated sponge regulatorynetwork in prostate cancer,” Nature Communications, vol. 7,no. 1, article 10982, 2016.

[26] M. Li, X. Wu, J. Wang, and Y. Pan, “Towards the identificationof protein complexes and functional modules by integratingPPI network and gene expression data,” BMC Bioinformatics,vol. 13, no. 1, p. 109, 2012.

[27] S. Haupt, J. O. Mejía-Hernández, R. Vijayakumaran, S. P.Keam, and Y. Haupt, “The long and the short of it: the

12 BioMed Research International

MDM4 tail so far,” Journal of Molecular Cell Biology, vol. 11,no. 3, pp. 231–244, 2019.

[28] E. A. Fry and K. Inoue, “c-MYB and DMTF1 in cancer,” Can-cer Investigation, vol. 37, no. 1, pp. 46–65, 2019.

[29] M. J. Wortman, L. K. Hanson, L. Martínez-Sobrido et al., “Reg-ulation of PURA gene transcription by three promoters gener-ating distinctly spliced 5-prime leaders: a novel means of finecontrol over tissue specificity and viral signals,” BMC Molecu-lar Biology, vol. 11, no. 1, p. 81, 2010.

[30] M. J. Zeitz, K. S. Malyavantham, B. Seifert, and R. Berezney,“Matrin 3: chromosomal distribution and protein interac-tions,” Journal of Cellular Biochemistry, vol. 108, no. 1,pp. 125–133, 2009.

[31] Y. Qu, N. Gharbi, X. Yuan et al., “Axitinib blocks Wnt/β-catenin signaling and directs asymmetric cell division incancer,” Proceedings of the National Academy of Sciencesof the United States of America, vol. 113, no. 33,pp. 9339–9344, 2016.

[32] C. Küper, F. X. Beck, and W. Neuhofer, “NFAT5-mediatedexpression of S100A4 contributes to proliferation and migra-tion of renal carcinoma cells,” Frontiers in Physiology, vol. 5,p. 293, 2014.

[33] G. Gu, L. Gelsomino, K. R. Covington et al., “Targeting thyroidhormone receptor beta in triple-negative breast cancer,” BreastCancer Research & Treatment, vol. 150, no. 3, pp. 535–545,2015.

[34] A. Kaymak, S. Sayols, T. Papadopoulu, and H. Richly, “Role forthe transcriptional activator ZRF1 in early metastatic events inbreast cancer progression and endocrine resistance,” Oncotar-get, vol. 9, no. 47, pp. 28666–28690, 2018.

[35] K. L. Thu, I. Soria-Bretones, T. W. Mak, and D. W. Cescon,“Targeting the cell cycle in breast cancer: towards the nextphase,” Cell Cycle, vol. 17, no. 15, pp. 1871–1885, 2018.

[36] Z. Liu, L. Jiang, G. Liang et al., “Hepatitis B virus reactivationin breast cancer patients undergoing chemotherapy: a reviewand meta-analysis of prophylaxis management,” Journal ofViral Hepatitis, vol. 24, no. 7, pp. 561–572, 2017.

[37] Y. C. Liu, C. T. Yeh, and K. H. Lin, “Molecular functions ofthyroid hormone signaling in regulation of cancer progressionand anti-apoptosis,” International Journal of Molecular Sci-ences, vol. 20, no. 20, article 4986, 2019.

[38] M. Mirabelli, E. Chiefari, B. Arcidiacono et al., “Mediterraneandiet nutrients to turn the tide against insulin resistance andrelated diseases,” Nutrients, vol. 12, no. 4, p. 1066, 2020.

[39] M. Samson, N. Porter, O. Orekoya et al., “Progestin and breastcancer risk: a systematic review,” Breast Cancer Research andTreatment, vol. 155, no. 1, pp. 3–12, 2016.

[40] R. Yang, L. Xing, M. Wang, H. Chi, L. Zhang, and J. Chen,“Comprehensive analysis of differentially expressed profilesof lncRNAs/mRNAs and miRNAs with associated ceRNAnetworks in triple-negative breast cancer,” Cellular Physiol-ogy and Biochemistry, vol. 50, no. 2, pp. 473–488, 2018.

[41] S. Haupt, R. Vijayakumaran, P. J. Miranda, A. Burgess, E. Lim,and Y. Haupt, “The role of MDM2 and MDM4 in breast can-cer development and prevention,” Journal of Molecular CellBiology, vol. 9, no. 1, pp. 53–61, 2017.

[42] V. A. de Weger, M. de Jonge, M. H. G. Langenberg et al., “Aphase I study of the HDM2 antagonist SAR405838 combinedwith the MEK inhibitor pimasertib in patients with advancedsolid tumours,” British Journal of Cancer, vol. 120, no. 3,pp. 286–293, 2019.

[43] M. M. Gounder, T. M. Bauer, G. K. Schwartz et al., “A phase 1study of the MDM2 inhibitor DS-3032b in patients (pts) withadvanced solid tumors and lymphomas,” Journal of ClinicalOncology, vol. 34, article 2581, Supplement 15, 2016.

[44] F. Meric-Bernstam, M. N. Saleh, J. R. Infante et al., “Phase Itrial of a novel stapled peptide ALRN-6924 disruptingMDMX- and MDM2-mediated inhibition ofWT p53inpatients with solid tumors and lymphomas,” Journal of Clini-cal Oncology, vol. 35, 15_suppl, p. 2505, 2017.

[45] N. Tian, J. Li, J. Shi, and G. Sui, “From general aberrant alter-native splicing in cancers and its therapeutic application to thediscovery of an oncogenic DMTF1 isoform,” InternationalJournal of Molecular Sciences, vol. 18, no. 3, p. 191, 2017.

[46] N. J. Niklaus, M. Humbert, and M. P. Tschan, “Cisplatin sen-sitivity in breast cancer cells is associated with particularDMTF1 splice variant expression,” Biochemical and Biophysi-cal Research Communications, vol. 503, no. 4, pp. 2800–2806,2018.

[47] D. Maglic, D. B. Stovall, J. M. Cline et al., “DMP1β, a splice iso-form of the tumour suppressor DMP1 locus, induces prolifer-ation and progression of breast cancer,” The Journal ofPathology, vol. 236, no. 1, pp. 90–102, 2015.

[48] S. Morales-Santana, S. Morell, J. Leon, A. Carazo-Gallego, J. C.Jimenez-Lopez, and M. Morell, “An overview of the polymor-phisms of circadian genes associated with endocrine cancer,”Frontiers in Endocrinology, vol. 10, p. 104, 2019.

[49] M. Taheri, M. D. Omrani, R. Noroozi, S. Ghafouri-Fard, andA. Sayad, “Retinoic acid-related orphan receptor alpha(RORA) variants and risk of breast cancer,” Breast Disease,vol. 37, no. 1, pp. 21–25, 2017.

[50] J. Du and R. Xu, “RORα, a potential tumor suppressor andtherapeutic target of breast cancer,” International Journal ofMolecular Sciences, vol. 13, no. 12, pp. 15755–15766, 2012.

[51] C. S. Leblond, Z. Gan-Or, D. Spiegelman et al., “Replicationstudy of MATR3 in familial and sporadic amyotrophic lateralsclerosis,” Neurobiol Aging, vol. 37, pp. 209.e217–209.e221,2016.

[52] T. W. Yang, D. Sahu, Y. W. Chang, C. L. Hsu, and C. H. Hsieh,“RNA-binding proteomics reveals MATR3 interacting withlncRNA SNHG1 to enhance neuroblastoma progression,”Journal of Proteome Research, vol. 18, no. 1, pp. 406–416, 2018.

[53] S. H. Nho, G. Yoon, J. H. Seo et al., “Licochalcone H inducesthe apoptosis of human oral squamous cell carcinoma cellsvia regulation of matrin 3,” Oncology Reports, vol. 41, no. 1,pp. 333–340, 2019.

[54] A. Hercbergs, S. A. Mousa, M. Leinung, H. Y. Lin, and P. J.Davis, “Thyroid hormone in the clinic and breast cancer,”Hormones and Cancer, vol. 9, no. 3, pp. 139–143, 2018.

[55] M. Søgaard, D. K. Farkas, V. Ehrenstein, J. O. Jørgensen, O. M.Dekkers, and H. T. Sørensen, “Hypothyroidism and hyperthy-roidism and breast cancer risk: a nationwide cohort study,”European Journal of Endocrinology, vol. 174, no. 4, pp. 409–414, 2016.

[56] B. Liu, L. Sun, Q. Liu et al., “A cytoplasmic NF-κB interactinglong noncoding RNA blocks IκB phosphorylation and sup-presses breast cancer metastasis,” Cancer Cell, vol. 27, no. 3,pp. 370–381, 2015.

13BioMed Research International