Open Access

Identification of co‑expression modules and hub genes of retinoblastoma via co‑expression analysis and protein‑protein interaction networks

  • Authors:
    • Yukun Mao
    • Qingbin Nie
    • Yang Yang
    • Gengsheng Mao
  • View Affiliations

  • Published online on: May 27, 2020     https://doi.org/10.3892/mmr.2020.11189
  • Pages: 1155-1168
  • Copyright: © Mao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Retinoblastoma is a common intraocular malignant tumor in children. However, the molecular and genetic mechanisms of retinoblastoma remain unclear. The gene expression dataset GSE110811 was retrieved from Gene Expression Omnibus. After preprocessing, coexpression modules were constructed by weighted gene coexpression network analysis (WGCNA), and modules associated with clinical traits were identified. In addition, functional enrichment analysis was performed for genes in the indicated modules, and protein‑protein interaction (PPI) networks and subnetworks were constructed based on these genes. Eight coexpression modules were constructed through WGCNA. Of these, the yellow module had the highest association with severity and age (r=0.82 and P=3e‑07; r=0.72 and P=3e‑05). The turquoise module had the highest association with months (r=‑0.63 and P=5e‑04). The genes in the two modules participate in multiple pathways of retinoblastoma, and by combining the PPI network and subnetworks; 10 hub genes were identified in the two modules. The present study identified coexpression modules and hub genes associated with clinical traits of retinoblastoma, providing novel insight into retinoblastoma progression.

Introduction

Retinoblastoma is a retinal embryo malignancy that occurs in childhood and is triggered by mutations in the retinoblastoma gene (RB1) in cells of the developing retina (1,2). Retinoblastoma accounts for 3% of all cancer types in children, and is the most common intraocular malignancy in children (3). Moreover, children with a family history of retinoblastoma have a significantly increased risk of developing retinoblastoma (4). When diagnosed at an early stage and treated with standard regimens of systemic chemotherapy and topical consolidation therapy it is possible to successfully treat retinoblastoma, with most patients maintaining normal vision in at least one eye (5). However, retinoblastoma can cause blindness or death if not treated at an early stage (6). In addition, the use of standard chemotherapy regimens may result in notable toxicities and the development of secondary malignancies, such as soft tissue sarcomas, brain tumors and osteosarcoma (79). Therefore, there is an urgent need to understand the molecular characteristics of RB1 expression in order to improve the quality of life of patients with retinoblastoma (4,10).

It has been confirmed that clinical risk assessment in retinoblastoma requires combining clinical traits. For example, tumor staging, pathological grade and tumor laterality show associations with the patient's age at diagnosis, overall survival and second malignancy in retinoblastoma (11). High-risk histopathologic features (HRPFs) are closely related to poor prognosis, including tumor invasion of the optic nerve, choroid or anterior chamber, suggesting the need for postenucleation adjuvant chemotherapy (12). Therefore, it is necessary to identify gene coexpression modules associated with these clinical traits.

Network-based analysis has been used to characterize clinically significant genes and constitutes a means of presenting a variety of biological data, such as protein-protein interactions, gene regulation, cellular pathways and signal transduction. The core elements of biological networks may be identified by measuring nodes based on their network features. In addition, weighted gene coexpression network analysis (WGCNA) has been widely applied to develop gene coexpression modules of various diseases. For example, Zhou et al (13) successfully applied coexpression analysis to identify gene coexpression modules associated with the prognosis of pancreatic carcinoma. However, there are few studies on the gene coexpression modules associated with the clinical traits of retinoblastoma using WGCNA.

In this study in order to comprehensively analyze the molecular characteristics of RB1 expression, the aim was to identify coexpression modules associated with the clinical traits of retinoblastoma based on WGCNA and pathways involving the genes in these modules, as well as identifying hub genes related to clinical traits by combining the findings with protein-protein interaction (PPI) network. This study will contribute to an improved understanding of the development of retinoblastoma.

Materials and methods

Raw data and pretreatment

The GSE110811 dataset was downloaded from Gene Expression Omnibus (https://www.ncbi.nlm.nih.gov/geo/). The dataset contains the gene expression profiles of 28 retinoblastoma samples based on the GPL16686 platform (Human Gene 2.0 ST array; Affymetrix; Thermo Fisher Scientific, Inc.) (12). Annotation information of microarray data was used to match probes with corresponding gene information. Probes matching with more than one gene were excluded, and average expression values were calculated for genes matching with more than one probe. Corresponding clinical information was also obtained. The workflow chart is shown in Fig. 1.

Construction of the gene coexpression network by WGCNA

A gene coexpression network was built with the WGCNA algorithm (14). Before constructing the network, the number of genes with different expression threshold was calculated, and the appropriate soft threshold value was then determined. Cluster analysis of 28 samples was performed at the appropriate threshold value using the flashclust package in R. The function pickSoftThreshold was applied to the calculate scale-free topology model fitting index (R2) and mean connectivity values corresponding to different soft threshold values (β; ranging from 1–20). The connectivity suggests the degree of correlation to which a gene is related to other genes in the network. The appropriate power value was estimated when R2 was 0.85. Furthermore, the gene expression matrix was transformed into an adjacency matrix and a topological overlap matrix (TOM). The corresponding dissimilarity of TOM (1-TOM) was calculated. The topological overlap is a measure of gene biological similarity based on pairwise gene coexpression correlation. A cluster dendrogram was constructed based on 1-TOM, and the DynamicTreeCut package (http://www.genetics.ucla.edu/labs/horvath/CoexpressionNetwork/BranchCutting/) was used to assign branches and generate modules. The heatmap package was employed to visualize the TOM among 400 randomly selected genes. For module construction, the default for maxBlockSize was 5,000, and deepSplit was set to 2. After calculating module eigengenes (MEs) using the function moduleEigengenes, modules with similar expression patterns were merged by the function mergeCloseModules. During this process, MEs are the major components of the first principal component in a module with the same expression profile, which can reflect the entire features of module genes.

Module-trait relationship construction

Correlation between MEs and clinical traits was analyzed by Pearson correlation analysis. R>0.8 was considered to indicate a strong correlation. For each expression profile, gene significance (GS) was calculated as the absolute value of the correlation between the expression profile and each clinical trait. Module membership (MM) was defined as the correlation between the expression profile and each ME (15). Thus, genes with a high significance for clinical traits and MM were identified by two parameters.

Functional and pathway enrichment analyses

To explore the biological processes and pathways in which the genes indicated in the module participate, functional enrichment analysis was performed via the clusterProfiler package in R language, including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses (16). GO terms include ‘biological process’, ‘molecular function’ and ‘cellular component’. P<0.05 after correction was set as the cutoff criterion.

PPI network and core subnetwork construction

PPI network analysis is a powerful tool for understanding the biological responses occurring in retinoblastoma. In the PPI network, a protein is defined as a node, and the interaction between two nodes is defined as an edge. The size of a node represents a degree: The larger the node, the larger the degree (17). The thickness of an edge indicates a correlation: The thicker the edge, the higher the correlation. Genes in the indicated modules were entered into the STRING online database (http://string-db.org/), and the cutoff value was set to 0.17. Related pairs of genes were retrieved and visualized with Cytoscape (version 3.4.0) (18). As an open platform with numerous plugins, the function of Cytoscape is to expand visualization options and network analysis power. Cytoscape cytoHubba plugin was used to select the top ten hub nodes according to maximal clique centrality (MCC) (19), and the Cytoscape MCODE plugin was applied to identify core subnetworks with highly interconnected nodes (20).

Results

Gene expression data

The box plot shows the expression value of each gene in 28 retinoblastoma samples (Fig. 2A). The average expression value of all mRNAs in each sample was nearly consistent, suggesting that the GSE110811 dataset was well normalized. As shown in Fig. 2B, two outlier samples (GSM3017126 and GSM3017124) were removed.

Coexpression module construction

When the soft threshold power value β was equal to eight, the connectivity between genes conformed to a scale-free network distribution (Fig. 3A-D). Different coexpression modules of retinoblastoma were identified by hierarchical clustering and dynamic branch cutting, which are shown as different colors in Fig. 3E. After merging similar modules, eight coexpression modules were generated. The number of genes in the eight-coexpression modules is listed in Table I. The gray module represents the set of genes that were not assigned to any module. Eigengenes were used as representative gene expression profiles, quantifying module similarity through eigengene correlation (Fig. 3F).

Table I.

Number of genes in the eight-coexpression modules.

Table I.

Number of genes in the eight-coexpression modules.

Module colorsFrequency
Black44
Blue836
Brown146
Green91
Grey697
Red49
Turquoise3,039
Yellow98
Module-trait relationship construction

To explore meaningful modules associated with clinical traits, correlations between MEs and the following clinical traits were analyzed: Age; HRPF; metastasis; mild; moderate; severe; unilateral; survival time (months); mutation germline (mutation G); mutation not known or test not done (mutation ND); mutation none; and mutation nongermline (mutation NG; Fig. 4). Among them, HRPF has been confirmed to be associated with a poor prognosis in retinoblastoma. Moreover, anaplastic grades (mild, moderate and severe) are able to predict risk stratification for retinoblastoma. As shown in Fig. 4, different colors represent different correlation coefficients; furthermore, the green suggests negative correlation and the red stands for positive correlation. It was found that the yellow module had the highest association with severe anaplasia and age (r=0.82 and P=3e-07; r=0.72 and P=3e-05) and that the turquoise module had the highest association with months (r=−0.63 and P=5e-04). The eigengene dendrogram and heatmap showed that the MEyellow module was highly related to severe anaplasia and age, and the MEturquoise module was highly related to months (Fig. 5A-J). Therefore, the two modules were selected as meaningful modules associated with clinical traits. The scatterplots of GS vs. MM in the MEyellow and MEturquoise modules are shown in Figs. 6A-F and 7A-G, respectively. The above results revealed that the yellow and turquoise modules were significantly associated with clinical traits.

Functional enrichment analysis of genes in the yellow and turquoise modules

KEGG and GO enrichment analyses were performed for the genes in the yellow and turquoise modules, and a significant difference in the biological processes enriched by the genes in the two-coexpression modules was observed. For GO analysis, genes in the yellow module are mainly enriched in negative regulation of neuron differentiation, axon guidance, neuron projection guidance, negative regulation of neurogenesis and negative regulation of nervous system development (Fig. 8A). However, no KEGG pathways enriched with genes in the yellow module were found. For GO analysis, genes in the turquoise module are mainly enriched in chromosome segregation, organelle fission, chromosomal region, centrosome, tubulin binding, ATPase activity and coupling (Fig. 8B). In KEGG enrichment analysis, genes in the turquoise module are enriched in protein processing in the endoplasmic reticulum, cell cycle, proteasome, DNA replication, mismatch repair and protein export (Fig. 8C).

PPI network construction

Genes in the yellow modules were used to construct the PPI network (Fig. 9A); subnetworks were then constructed to explore more specific and detailed information in the PPI network, as identified using the Cytoscape MCODE plugin. Two subnetworks (scores=4.6 and 3) were obtained for the yellow module (Fig. 9B and C). Genes in the turquoise modules were used to construct the PPI network (Fig. 10A). In total, three subnetworks (scores=4, 3.714 and 3.111) were obtained for the turquoise module (Fig. 10B-D). The Cytoscape CytoHubba plugin was employed to find the top ten hub genes with a high degree of connectivity between the nodes, according to the MCC. The top 10 hub genes identified from the PPI network of genes in the yellow and turquoise modules are shown in Fig. 11A and B, respectively, and the top ten hub genes in the yellow and turquoise modules are listed in Tables II and III, respectively.

Table II.

Top 10 hub genes identified from the PPI network of genes in the yellow module.

Table II.

Top 10 hub genes identified from the PPI network of genes in the yellow module.

RankGene nameScore
1BCL11A3
2SSBP3-AS12
2EBF32
4KCNQ5-IT11
4LOC1019296331
4CA21
4ASAP21
4ROBO11
4INSM21
4LGI21

[i] BCL11A, BAF chromatin remodeling complex subunit BCL11A; SSBP3-AS1, SSBP3 antisense RNA 1; EBF3, EBF transcription factor 3; KCNQ5-IT1, KCNQ5 intronic transcript 1; LOC101929633, uncharacterized LOC101929633; CA2, carbonic anhydrase 2; ASAP2, ArfGAP with SH3 domain, ankyrin repeat and PH domain 2; ROBO1, roundabout guidance receptor 1; INSM2, INSM transcriptional repressor 2; LGI2, leucine rich repeat LGI family member 2.

Table III.

Top 10 hub genes identified from the PPI network of genes in the turquoise module.

Table III.

Top 10 hub genes identified from the PPI network of genes in the turquoise module.

RankGene nameScore
1NIPBL35
2LRPPRC31
3GPBP124
4UBE3A21
5HERC18
5RNF2148
7CCNH7
8DYRK1A4
9STAM3
9CCT23

[i] NIPBL, NIPBL cohesion loading factor; LRPPRC, leucine rich pentatricopeptide repeat containing; GPBP1, GC-rich promoter binding protein 1; UBE3A, ubiquitin protein ligase E3A; HERC1, HECT and RLD domain containing E3 ubiquitin protein ligase family member 1; RNF214, ring finger protein 214; CCNH, cyclin H; DYRK1A, dual specificity tyrosine phosphorylation regulated kinase 1Al; STAM, signal transducing adaptor molecule; CCT2, chaperonin containing TCP1 subunit 2.

Discussion

The present study systematically analyzed gene expression for retinoblastoma using WGCNA and PPI networks. These findings identified gene coexpression modules and hub genes associated with clinical traits of retinoblastoma, providing novel insight into retinoblastoma progression.

In the present study, 8 coexpression modules were identified based on WGCNA. Among them, the yellow module exhibited the highest association with histopathologic grading and age. Histopathologic grading included mild, moderate and severe. Histopathologic grading was determined based on tumor size, growth pattern, level of differentiation, degree of apoptosis, grade of anaplasia, tumor seeding, extent of tissue invasion and the presence of retinocytoma. High parental age is associated with increased risk of sporadic hereditary retinoblastoma (21). Therefore, early detection is important in curing the disease using surgical treatment (22). It has been recorded that a delay of more than 6 months from the first clinical sign to diagnosis is associated with 70% mortality in developing countries (23). The turquoise module had the highest association with survival time (months), indicating that the module could become a potential prognostic factor. Therefore, the two modules were considered important in retinoblastoma. GO analysis revealed that the genes in the yellow module were mainly enriched in the negative regulation of neuron differentiation, axon guidance, neuron projection guidance, neurogenesis and development of the nervous system. Increasing evidence suggests that neuronal differentiation of neuroblastoma cell lines is induced by a number of factors, such as p73, Tropomyosin receptor kinase A and Ubiquitin C-Terminal Hydrolase L1 (2426). According to GO enrichment analysis, the genes in the turquoise module are mainly enriched in chromosome segregation, organelle fission, chromosomal region, centrosome, tubulin binding, and ATPase activity and coupling. In KEGG enrichment analysis, it was shown that genes in the turquoise module were enriched in protein processing in the endoplasmic reticulum, cell cycle, proteasome, DNA replication, mismatch repair and protein export. Hub genes in the turquoise module were involved in these pathways. For example, among these pathways, hub gene CCNH was enriched in the cell cycle pathway. Overall, the genes in the two modules may play important roles in the development of retinoblastoma.

In this study, the top ten hub genes with a high degree of connectivity according to MCC in the yellow and turquoise modules were identified. The degree of a particular protein is related to the necessity of the gene, and proteins with higher numbers are more likely to be essential (27). As biological networks are heterogeneous, it is necessary to capture essential proteins using a variety of methods. The top 10 hub genes in the yellow module included: BAF chromatin remodeling complex subunit BCL11A (BCL11A); SSBP3 antisense RNA 1 (SSBP3-AS1); EBF transcription factor 3, KCNQ5 intronic transcript 1 (KCNQ5-IT1); uncharacterized LOC101929633 (LOC101929633); carbonic anhydrase 2 (CA2); ArfGAP with SH3 domain, ankyrin repeat and PH domain 2; roundabout guidance receptor 1 (ROBO1); INSM transcriptional repressor 2; and leucine rich repeat LGI family member 2 (LGI2). Those in the turquoise module included: NIPBL cohesion loading factor (NIPBL); leucine rich pentatricopeptide repeat containing (LRPPRC); GC-rich promoter binding protein 1; ubiquitin protein ligase E3A; HECT and RLD domain containing E3 ubiquitin protein ligase family member 1; ring finger protein 214; cyclin H (CCNH); dual specificity tyrosine phosphorylation regulated kinase 1Al (DYRK1A); signal transducing adaptor molecule; and chaperonin containing TCP1 subunit 2 (CCT2). However, the function of these genes in retinoblastoma remains unclear. A previous study found that BCL11A plays a critical role in several diseases as an oncogene (28). Consistent with the present study, it has been confirmed that in epigenetic complexes, the transcription factor BCL11A competes with histone H3 for binding to Retinoblastoma-binding proteins 4 and 7 (29). Therefore, BCL11A has a widespread role in the development of retinoblastoma (30). It has been reported that ROBO1 is localized to the cell membrane; in primary and metastatic prostate cancer its expression is lower (31). However, the role of ROBO1 in retinoblastoma has not yet been studied. The present results showed that ROBO1 in the yellow module may be associated with retinoblastoma, but this requires further validation. LGI2 is secreted by neurons and acts on members of the metalloproteinase-deficient ADAM neuronal receptor family, with roles in synaptic remodeling (32). Similarly, GO analysis in the present study indicated enrichment of the genes in the yellow module in synaptic guidance. Among the genes in the turquoise module, NIPBL plays a critical and regulatory role in cohesion loading on chromatin, as well as having roles in gene expression and transcriptional signaling (33,34). Zinc finger protein 609 may participate in the regulation of cortical neuron migration (35). Upregulated LRPPRC may promote tumourigenesis in various tumors, such as lung adenocarcinoma, esophageal squamous cell carcinoma, stomach, colon, mammary and endometrial adenocarcinoma, as well as lymphoma (36,37). Regardless, the present study is the first to report that LRPPRC may affect the development of retinoblastoma. Furthermore, CCNH, DYRK1A and CCT2 are involved in the regulation of the cell cycle, which is consistent with the present functional enrichment analysis results (3842). Due to the unclear molecular mechanisms of retinoblastoma, the ability to treat the disease remains limited (4345). Moreover, patients with hereditary retinoblastoma have an increased risk of subsequent malignant neoplasms after radiotherapy or chemotherapy (46,47). The present study revealed that these hub genes may be involved in the molecular mechanisms underlying the development and progression of retinoblastoma, though further validation is required.

Currently, WGCNA is widely used to analyze large-scale data sets and find modules for highly related genes. To our knowledge, the present study is the first to analyze the correlation between coexpression modules and clinical traits of retinoblastoma via WGCNA. In a previous study using WGCNA analysis four hub genes were identified as prognostic markers in another eye tumor, uveal melanoma (15) and another study identified four hub genes associated with the progression of uveal melanoma (48). However, identical genes associated with the two eye tumors, uveal melanoma and retinoblastoma, have not been found. The genes identified using WGCNA in uveal melanoma have been not used in a clinical setting; however, functional enrichment studies have not revealed the role of these genes. Since WGCNA has been widely used to construct coexpression modules, it is a useful method to study retinoblastoma.

However, the present study has several limitations that should be noted. First, this study was based on analysis of data extracted from a limited sample size; therefore, the genes identified cannot be generalized to the majority of patients with retinoblastoma. Second, the biological mechanisms of the hub genes, such as SSBP3-AS1, KCNQ5-IT1, LOC101929633 and CA2, are still unknown. Third, this study was limited by the absence of experimental evidence, so large-scale studies are needed for validation.

In the present study, two meaningful gene coexpression modules for retinoblastoma were identified based on WGCNA. Among the genes of the two-coexpression modules, hub genes were identified by PPI networks and subnetworks, and based on previous studies, these genes may play a critical role in retinoblastoma. Therefore, this research is helpful in understanding the molecular mechanisms of retinoblastoma.

In this study, 8 coexpression modules were constructed via WGCNA. The yellow module and turquoise module had highly significant associations with clinical traits. In addition, the genes in the two modules participate in multiple pathways in retinoblastoma. Ten hub genes in the two-coexpression modules were identified according to the PPI network. This study identified meaningful gene coexpression modules and hub genes associated with a number of clinical traits of retinoblastoma that may contribute to the development of the disease.

Acknowledgements

Not applicable.

Funding

This study was funded by the Beijing Municipal Science and Technology Commission (grant no. Z151100004015179).

Availability of data and material

The datasets analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YM and GM conceived and designed the study strategy and reviewed the manuscript. GM, YM, YY and QN conducted the data analysis and prepared the tables and figures. YM and QN finished the draft of the manuscript and managed the data. GM and YY collected the references and managed the data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

WGCNA

weighted gene coexpression network analysis

PPI

protein-protein interaction

HRPFs

high-risk histopathologic features

TOM

topological overlap matrix

ME

module eigengene

GS

gene significance

MM

module membership

GO

Gene Ontology

KEGG

Kyoto Encyclopedia of Genes and Genomes

MCC

maximal clique centrality

References

1 

Kamihara J, Bourdeaut F, Foulkes WD, Molenaar JJ, Mosse YP, Nakagawara A, Parareda A, Scollon SR, Schneider KW, Skalet AH, et al: Retinoblastoma and neuroblastoma predisposition and surveillance. Clin Cancer Res. 23:e98–e106. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Shang Y: LncRNA THOR acts as a retinoblastoma promoter through enhancing the combination of c-myc mRNA and IGF2BP1 protein. Biomed Pharmacother. 106:1243–1249. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Rao R and Honavar SG: Retinoblastoma. Indian J Pediatr. 84:937–944. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Skalet AH, Gombos DS, Gallie BL, Kim JW, Shields CL, Marr BP, Plon SE and Chévez-Barrios P: Screening children at risk for retinoblastoma: Consensus report from the American association of ophthalmic oncologists and pathologists. Ophthalmology. 125:453–458. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Broaddus E, Topham A and Singh AD: Survival with retinoblastoma in the USA: 1975–2004. Br J Ophthalmol. 93:24–27. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Stathopoulos C, Say EAT and Shields CL: Prenatal ultrasonographic detection and prenatal (prior to birth) management of hereditary retinoblastoma. Graefes Arch Clin Exp Ophthalmol. 256:861–862. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Bright CJ, Hawkins MM, Winter DL, Alessi D, Allodji RS, Bagnasco F, Bárdi E, Bautz A, Byrne J, Feijen EAM, et al: Risk of soft-tissue sarcoma among 69 460 five-year survivors of childhood cancer in Europe. J Natl Cancer Inst. 110:649–660. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Fidler MM, Reulen RC, Winter DL, Allodji RS, Bagnasco F, Bardi E, Bautz A, Bright CJ, Byrne J, Feijen EAM, et al: Risk of subsequent bone cancers among 69 460 five-year survivors of childhood and adolescent cancer in Europe. J Natl Cancer Inst. 110:22018. View Article : Google Scholar

9 

Qaddoumi I, Bass JK, Wu J, Billups CA, Wozniak AW, Merchant TE, Haik BG, Wilson MW and Rodriguez-Galindo C: Carboplatin-associated ototoxicity in children with retinoblastoma. J Clin Oncol. 30:1034–1041. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Sachdeva UM and O'Brien JM: Understanding pRb: Toward the necessary development of targeted treatments for retinoblastoma. J Clin Invest. 122:425–434. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Andreoli MT, Chau FY, Shapiro MJ and Leiderman YI: Epidemiological trends in 1452 cases of retinoblastoma from the surveillance, epidemiology, and end results (SEER) registry. Can J Ophthalmol. 52:592–598. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Hudson LE, Mendoza P, Hudson WH, Ziesel A, Hubbard GB III, Wells J, Dwivedi B, Kowalski J, Seby S, Patel V, et al: Distinct gene expression profiles define anaplastic grade in retinoblastoma. Am J Pathol. 188:2328–2338. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Zhou Z, Cheng Y, Jiang Y, Liu S, Zhang M, Liu J and Zhao Q: Ten hub genes associated with progression and prognosis of pancreatic carcinoma identified by co-expression analysis. Int J Biol Sci. 14:124–136. 2018. View Article : Google Scholar : PubMed/NCBI

14 

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

15 

Wan Q, Tang J, Han Y and Wang D: Co-Expression modules construction by WGCNA and identify potential prognostic markers of uveal melanoma. Exp Eye Res. 166:13–20. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Yu G, Wang LG, Han Y and He QY: ClusterProfiler: An R package for comparing biological themes among gene clusters. OMICS. 16:284–287. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Kohl M, Wiese S and Warscheid B: Cytoscape: Software for visualization and analysis of biological networks. Methods Mol Biol. 696:291–303. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Chin CH, Chen SH, Wu HH, Ho CW, Ko MT and Lin CY: CytoHubba: Identifying hub objects and sub-networks from complex interactome. BMC Syst Biol. 8 (Suppl 4):S112014. View Article : Google Scholar : PubMed/NCBI

20 

Bader GD and Hogue CW: An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinformatics. 4:22003. View Article : Google Scholar : PubMed/NCBI

21 

Moll AC, Imhof SM, Kuik DJ, Bouter LM, Den Otter W, Bezemer PD, Koten JW and Tan KE: High parental age is associated with sporadic hereditary retinoblastoma: The dutch retinoblastoma register 1862–1994. Hum Genet. 98:109–112. 1996. View Article : Google Scholar : PubMed/NCBI

22 

de Jong MC, Kors WA, de Graaf P, Castelijns JA, Kivelä T and Moll AC: Trilateral retinoblastoma: A systematic review and meta-analysis. Lancet Oncol. 15:1157–1167. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Dimaras H, Kimani K, Dimba EA, Gronsdahl P, White A, Chan HS and Gallie BL: Retinoblastoma. Lancet. 379:1436–1446. 2012. View Article : Google Scholar : PubMed/NCBI

24 

De Laurenzi V, Raschellá G, Barcaroli D, Annicchiarico-Petruzzelli M, Ranalli M, Catani MV, Tanno B, Costanzo A, Levrero M and Melino G: Induction of neuronal differentiation by p73 in a neuroblastoma cell line. J Biol Chem. 275:15226–15231. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Fell SM, Li S, Wallis K, Kock A, Surova O, Rraklli V, Höfig CS, Li W, Mittag J, Henriksson MA, et al: Neuroblast differentiation during development and in neuroblastoma requires KIF1Bβ-mediated transport of TRKA. Genes Dev. 31:1036–1053. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Gu Y, Lv F, Xue M, Chen K, Cheng C, Ding X, Jin M, Xu G, Zhang Y, Wu Z, et al: The deubiquitinating enzyme UCHL1 is a favorable prognostic marker in neuroblastoma as it promotes neuronal differentiation. J Exp Clin Cancer Res. 37:2582018. View Article : Google Scholar : PubMed/NCBI

27 

Jeong H, Mason SP, Barabási AL and Oltvai ZN: Lethality and centrality in protein networks. Nature. 411:41–42. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Lazarus KA, Hadi F, Zambon E, Bach K, Santolla MF, Watson JK, Correia LL, Das M, Ugur R, Pensa S, et al: BCL11A interacts with SOX2 to control the expression of epigenetic regulators in lung squamous carcinoma. Nat Commun. 9:33272018. View Article : Google Scholar : PubMed/NCBI

29 

Moody RR, Lo MC, Meagher JL, Lin CC, Stevers NO, Tinsley SL, Jung I, Matvekas A, Stuckey JA and Sun D: Probing the interaction between the histone methyltransferase/deacetylase subunit RBBP4/7 and the transcription factor BCL11A in epigenetic complexes. J Biol Chem. 293:2125–2136. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Sleven H, Welsh SJ, Yu J, Churchill MEA, Wright CF, Henderson A, Horvath R, Rankin J, Vogt J, Magee A, et al: De novo mutations in EBF3 cause a neurodevelopmental syndrome. Am J Hum Genet. 100:138–150. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Parray A, Siddique HR, Kuriger JK, Mishra SK, Rhim JS, Nelson HH, Aburatani H, Konety BR, Koochekpour S and Saleem M: ROBO1, a tumor suppressor and critical molecular barrier for localized tumor cells to acquire invasive phenotype: Study in African-American and Caucasian prostate cancer models. Int J Cancer. 135:2493–2506. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Seppala EH, Jokinen TS, Fukata M, Fukata Y, Webster MT, Karlsson EK, Kilpinen SK, Steffen F, Dietschi E, Leeb T, et al: LGI2 truncation causes a remitting focal epilepsy in dogs. PLoS Genet. 7:e10021942011. View Article : Google Scholar : PubMed/NCBI

33 

Zuin J, Casa V, Pozojevic J, Kolovos P, van den Hout MCGN, van Ijcken WFJ, Parenti I, Braunholz D, Baron Y, Watrin E, et al: Regulation of the cohesin-loading factor NIPBL: Role of the lncRNA NIPBL-AS1 and identification of a distal enhancer element. PLoS Genet. 13:e10071372017. View Article : Google Scholar : PubMed/NCBI

34 

Gao D, Zhu B, Cao X, Zhang M and Wang X: Roles of NIPBL in maintenance of genome stability. Semin Cell Dev Biol. 90:181–186. 2019. View Article : Google Scholar : PubMed/NCBI

35 

van den Berg DLC, Azzarelli R, Oishi K, Martynoga B, Urbán N, Dekkers DHW, Demmers JA and Guillemot F: Nipbl interacts with Zfp609 and the integrator complex to regulate cortical neuron migration. Neuron. 93:348–361. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Siira SJ, Spahr H, Shearwood AJ, Ruzzenente B, Larsson NG, Rackham O and Filipovska A: LRPPRC-mediated folding of the mitochondrial transcriptome. Nat Commun. 8:15322017. View Article : Google Scholar : PubMed/NCBI

37 

Tian T, Ikeda J, Wang Y, Mamat S, Luo W, Aozasa K and Morii E: Role of leucine-rich pentatricopeptide repeat motif-containing protein (LRPPRC) for anti-apoptosis and tumourigenesis in cancers. Eur J Cancer. 48:2462–2473. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Murali A, Nalinakumari KR, Thomas S and Kannan S: Association of single nucleotide polymorphisms in cell cycle regulatory genes with oral cancer susceptibility. Br J Oral Maxillofac Surg. 52:652–658. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Rajaraman P, Wang SS, Rothman N, Brown MM, Black PM, Fine HA, Loeffler JS, Selker RG, Shapiro WR, Chanock SJ and Inskip PD: Polymorphisms in apoptosis and cell cycle control genes and risk of brain tumors in adults. Cancer Epidemiol Biomarkers Prev. 16:1655–1661. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Litovchick L, Florens LA, Swanson SK, Washburn MP and DeCaprio JA: DYRK1A protein kinase promotes quiescence and senescence through DREAM complex assembly. Genes Dev. 25:801–813. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Earl RK, Turner TN, Mefford HC, Hudac CM, Gerdts J, Eichler EE and Bernier RA: Clinical phenotype of ASD-associated DYRK1A haploinsufficiency. Mol Autism. 8:542017. View Article : Google Scholar : PubMed/NCBI

42 

Minegishi Y, Nakaya N and Tomarev SI: Mutation in the zebrafish cct2 gene leads to abnormalities of cell cycle and cell death in the retina: A model of CCT2-related leber congenital amaurosis. Invest Ophthalmol Vis Sci. 59:995–1004. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Errico A: Cancer therapy: Retinoblastoma-chemotherapy increases the risk of secondary cancer. Nat Rev Clin Oncol. 11:6232014. View Article : Google Scholar : PubMed/NCBI

44 

Wong JR, Morton LM, Tucker MA, Abramson DH, Seddon JM, Sampson JN and Kleinerman RA: Risk of subsequent malignant neoplasms in long-term hereditary retinoblastoma survivors after chemotherapy and radiotherapy. J Clin Oncol. 32:3284–3290. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Temming P, Arendt M, Viehmann A, Eisele L, Le Guin CH, Schündeln MM, Biewald E, Mausert J, Wieland R, Bornfeld N, et al: How eye-preserving therapy affects long-term overall survival in heritable retinoblastoma survivors. J Clin Oncol. 34:3183–3188. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Habib LA, Francis JH, Fabius AW, Gobin PY, Dunkel IJ and Abramson DH: Second primary malignancies in retinoblastoma patients treated with intra-arterial chemotherapy: The first 10 years. Br J Ophthalmol. 102:272–275. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Temming P, Arendt M, Viehmann A, Eisele L, Le Guin CH, Schundeln MM, Biewald E, Astrahantseff K, Wieland R, Bornfeld N, et al: Incidence of second cancers after radiotherapy and systemic chemotherapy in heritable retinoblastoma survivors: A report from the German reference center. Pediatr Blood Cancer. 64:71–80. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Shi K, Bing ZT, Cao GQ, Guo L, Cao YN, Jiang HO and Zhang MX: Identify the signature genes for diagnose of uveal melanoma by weight gene co-expression network analysis. Int J Ophthalmol. 8:269–274. 2015.PubMed/NCBI

Related Articles

Journal Cover

August-2020
Volume 22 Issue 2

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Mao Y, Nie Q, Yang Y and Mao G: Identification of co‑expression modules and hub genes of retinoblastoma via co‑expression analysis and protein‑protein interaction networks. Mol Med Rep 22: 1155-1168, 2020
APA
Mao, Y., Nie, Q., Yang, Y., & Mao, G. (2020). Identification of co‑expression modules and hub genes of retinoblastoma via co‑expression analysis and protein‑protein interaction networks. Molecular Medicine Reports, 22, 1155-1168. https://doi.org/10.3892/mmr.2020.11189
MLA
Mao, Y., Nie, Q., Yang, Y., Mao, G."Identification of co‑expression modules and hub genes of retinoblastoma via co‑expression analysis and protein‑protein interaction networks". Molecular Medicine Reports 22.2 (2020): 1155-1168.
Chicago
Mao, Y., Nie, Q., Yang, Y., Mao, G."Identification of co‑expression modules and hub genes of retinoblastoma via co‑expression analysis and protein‑protein interaction networks". Molecular Medicine Reports 22, no. 2 (2020): 1155-1168. https://doi.org/10.3892/mmr.2020.11189