Exploration of bladder cancer molecular mechanisms based on miRNA-mRNA regulatory network

  • Authors:
    • Wenfeng Li
    • Jia Liu
    • Di Zou
    • Xiaye Cai
    • Jingying Wang
    • Jinmeng Wang
    • Li Zhu
    • Liang Zhao
    • Rongying Ou
    • Yunsheng Xu
  • View Affiliations

  • Published online on: February 7, 2017     https://doi.org/10.3892/or.2017.5433
  • Pages: 1461-1468
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

To explore the complex molecular mechanisms of bladder cancer, mRNA and miRNA expression profiles were combined for systematic analyses. A total of 18 common differentially expressed genes (DEGs) were identified from two mRNA expression datasets which consisted of 206 tumor and 74 normal tissues. Then, survival analysis based on the SurvExpress database showed that the common DEGs were able to significantly differentiate low- and high-risk groups in 4 public bladder cancer datasets (p<0.01). Notably, the tumor and normal samples were able to be almost clearly classified into 4 groups based on these identified common DEGs. In addition, 6 out of the 18 common DEGs, including ALDH1A1 and SRPX, are regulated by 6 reported miRNAs based on regulatory network analyses. Expression levels of the 6 DEGs were validated in 10 bladder cancer samples using RT-PCR, and the expression values were concordant with the microarray results. Collectively, our analyses indicated that various biological processes are involved in the development and progression of bladder cancer. Firstly, cell cycle checkpoints and DNA repair networks of cancer stem-like cells were regulated by high expression of ALDH1A1, and hence promoted tumor self-renewal or metastasis. Then, activation of HspB6 induced the angiogenesis process which provides necessary nutrition and oxygen for tumor cells. Moreover, downregulation of the expression of tumor-suppressor genes SRPX and FLNC further promoted apoptosis and metastasis. The identification of potential biological processes and genes can be helpful for the understanding of bladder cancer molecular mechanisms.

Introduction

Bladder cancer, as one of the most common types of cancer of the urinary tract, accounts for ~380,000 new cases and ~150,000 deaths every year, worldwide (1). Statistics show that the incidence of bladder cancer experience an increase of 19.4% adjusted for the increase in total world population from 1990 to 2010 (2). In the US, the 5-year survival rate is ~77% (3). Epidemiological research has shown that age is one of the most significant risk factor, and the average age at diagnosis of bladder cancer is ~70-year old (4). Apart from age, smoking is considered as the leading environmental risk factor, and more than half of bladder cancer cases can be attributed to smoking (4).

Considering the complexity of bladder cancer molecular mechanisms, extensive research has been carried out using microarray or next-generation sequencing technologies. In addition, several mRNAs and miRNAs have been identified to be differentially expressed based on RNA-seq data from 129 tumors. One of them is FGFR3 which is upregulated in the papillary-like bladder cancer cluster. Moreover, miR-99a and miR-100 that downregulate FGFR3 were found to be downregulated in bladder cancer (5). In addition, 32 significant somatic mutations including TP53, RB1, FGRF3, were identified in 130 paired tumor and normal samples using whole-exome sequencing (5). Researches also showed that several pathways were frequently dysregulated in bladder cancer such as cell cycle regulation, kinase and phosphatidylinositol-3-OH kinase and chromatin remodeling (5).

Although research has been carried to explore bladder cancer molecular mechanisms, research based on the integration of mRNA and miRNA expression profiles has not been widely explored. With the development of microarray and sequencing technology, more and more datasets have been submitted to the Gene Expression Omnibus (GEO) database, and hence re-analyses of the deposited datasets with advanced bioinformatic methods can further promote the understanding of bladder cancer molecular mechanisms (6). In the present study, DEGs in bladder cancer were firstly identified based on two mRNA expression datasets from different laboratories. Then, the common DEGs were subjected to function and pathway analyses. Furthermore, mRNA and miRNA regulatory networks were constructed. Finally, several critical DEGs were validated using SurvExpress database and RT-PCR method.

Materials and methods

Acquisition of microarray data

Public available datasets were used in the present study. Gene expression profiles GSE13507 and GSE37815 were downloaded from the GEO database (http://www.ncbi.nlm.nih.gov/geo/). These two datasets were submitted by Kim et al in 2008 (7) and Kim et al in 2012 (8), respectively. Dataset GSE13507 consists of 188 tumor and 68 normal tissues, and dataset GSE37815 consists of 24 expression profiles including 6 normal and 18 tumor tissues. RNAs were extracted and hybridized to Illumina human-6 v2.0 expression beadchip according to the manufacturer's instructions. Detailed information concerning the experiment design and samples were documented in previous studies (8,9).

Identification of differentially expressed mRNAs

To identify DEGs, raw data were systematically analyzed using in-house R script. Firstly, mRNA expression values were subjected to 1og2 transformation, background correction and normalization using GeneChip Robust Multi-Array Analysis (GC-RMA) (10) algorithm with default parameters. Then, uninformative control probe sets were filtered out, and the average expression value was calculated for genes with multiple probes. Finally, DEGs were identified using Linear Models for Microarray Analysis (Limma) package (11) with criteria of adjusted p≤0.01 and |log2 fold-change (FC)| ≥2. Common DEGs between the two datasets were presented using Venn diagram. Heat map of DEGs was constructed using heatmap.2 method within ggplot package.

Function and pathway enrichment analysis

To illuminate the biology functions related to the identified DEGs, Gene Ontology (GO) and KEGG pathway enrichment for the common DEGs were carried out using Database for Annotation, Visualization and Integrated Discovery (DAVID) online tools (12). GO terms, consisting of biological process (BP), cellular component (CC) and molecular function (MF), were screened out with p<0.05.

miRNA-mRNA regulatory network

Research has shown that miRNAs can regulate carcinogenesis, malignant transformation, and metastatic processes by preventing mRNA expression or directly participating in those processes (13). In the present study, 6 miRNAs documented in previous research were used for the construction of miRNA-mRNA pairings contributing to bladder cancer. Combination of the 6 miRNAs has been demonstrated to be the best predictor to distinguish patients with urothelial bladder cancer from normal controls (14). miRNA-mRNA regulatory network was constructed using CyTargetLinker plugin (15) of Cytoscape (16). The prediction of miRNA targets were based on microcosm, miRTarBase and TargetScan databases. Common target genes from the 3 databases were selected, and the interaction between the 6 miRNAs and the common DEGs were constructed.

Virtual validation and RT-PCR validation

Clinical outcome validation for the common DEGs were carried out using online tool SurvExpress, which is based on a cancer-wide gene expression database with clinical outcomes (17). Four bladder cancer datasets in this database were used for virtual validation. Parameter settings were carefully selected according to the manufacturer's instructions.

Six DEGs including ALDH1A1, FLNC, CNN1, SRPX, HSPB6 and FAM107A were selected for further RT-PCR validation based on the criterion that DEGs can be regulated by miRNAs and were identified in the SurvExpress analysis. Total RNAs were extracted from 10 bladder cancer and adjacent normal tissues with TRIzol reagent from ThermoFisher (Waltham, MA, USA). Extraction was based on the standard protocol and manufacturer's instructions. Then, cDNA were obtained with M-MLV Reverse Transcriptase from Promega (Madison, WI, USA). mRNA expression values were detected using 7500 Real-Time PCR System (ThermoFisher). The internal GAPDH mRNA expression was used for normalization and relative quantification was calculated using the 2−ΔCt method. Primer sequences for the 6 genes were as follows: HSPB6, 5′-TTGCTGTCAAGGTGGTGGGC-3′ (forward) and 5′-CGGTAGCGACGGTGGAACT-3′ (reverse); SRPX, 5′-CCCACAGCCCGAAACCT-3′ (forward) and 5′-TGCTCCTATCCTGCCAATG-3′ (reverse); CNN1, 5′-ACCCTCCTGGCTTTGGC-3′ (forward) and 5′-AATGATGTTCCGCCCTTCT-3′ (reverse); FLNC, 5′-AGAGAAGTTGGAGAGGAGAGTAG-3′ (forward) and 5′-AACCRCTATTATTCATCATACTAAC-3′ (reverse); ALDH1A1, 5′-CTTGGAATTTCCCGTTGG-3′ (forward) and 5′-TTGCTCTGCTGGTTTGACA-3′ (reverse); FAM107A, 5′-AGCAACACGCTCCTGACTT-3′ (forward) and 5′-TGGCGGCCTTATTGTCTA-3′ (reverse).

Results

DEGs in bladder cancer

Background correction and normalization were applied to the two datasets, and the medians of the gene expression values were almost at the same level indicating that the data were appropriate for subsequent analysis (data not shown). After independent DEG analysis, a total of 21 and 72 DEGs were screened out for GSE13507 and GSE37815, respectively. In addition, 18 DEGs (Table I) were identified to be differentially expressed in the two datasets (Fig. 1A). Among these common DEGs, 8 DEGs were upregulated and 10 DEGs were downregulated. In addition, there was a high correlation (R2=0.97) for the common DEG expression values between GSE13507 and GSE37815 (Fig. 1B).

Table I.

The identified 18 common DEGs in GSE37817 and GSE13507.

Table I.

The identified 18 common DEGs in GSE37817 and GSE13507.

GSE37817GSE13507


GeneP-valueFCP-valueFC
ALDH1A10.001223.575.44E-192.050259
C2orf400.0028172.362.34E-162.045515
CNN10.009863.365.03E-183.006849
COL16A10.004753−2.52.52E-23−2.16685
CPED10.001365−2.52.70E-23−2.00774
CRYAB0.0042643.311.23E-152.154952
DCN0.0048072.581.26E-222.253045
FAM107A0.000146−3.175.11E-28−2.28816
FLNC0.009383−3.691.94E-19−3.02438
HSPB60.002658−2.962.14E-19−2.41159
PCP40.000849−4.013.03E-14−2.51219
PDLIM30.0071892.121.96E-162.15556
PLAC90.000661−2.812.15E-20−2.03664
PRUNE20.0013653.443.27E-152.298053
SMOC20.007966−2.391.16E-20−2.21085
SRPX0.000761−3.521.77E-27−2.68146
SYNM0.0020993.111.33E-142.198553
TCEAL20.000251−3.063.26E-20−2.01581

[i] DEGs, differentially expressed genes. FC, fold change.

Furthermore, hierarchical clustering of the tumor and normal tissues was carried out based on the common DEGs. As indicated in Fig. 2, the tumor and normal tissues were able to be almost clearly classified into different clusters. Error assignment of several samples may have been caused by tumor heterogeneity or smaller expression value variation.

GO and KEGG pathway enrichment analysis

To explore the biology functions of the common DEGs, GO and KEGG pathway enrichment were conducted using DAVID online tool. Results showed that 3 KEGG pathways were identified. ALDH1A1 participates in retinol metabolism pathway, DCN is involved in the TGF-β signaling pathway, and FLNC plays a role in the MAPK signaling pathway. In addition, the common DEGs were mainly related to the cellular component of myofibril (p=1.0E-04), contractile fiber part (p=1.0E-04) and Z-disc (p=1.2E-04) (Table II). Several common DEGs, such as CRYAB, PDLIM3, SYNM, CNN1, COL16A1 and FLNC, were significantly enriched in the cytoskeletal protein binding function (p=2.4E-04), structural molecule activity function (p=0.007), and structural constituent of muscle function (p=0.02) (Table II). However, the biological processes of cytoskeleton organization (p=0.004) and response to heat (p=0.04) were also enriched (Table II).

Table II.

GO enrichment analysis results for the common DEGs

Table II.

GO enrichment analysis results for the common DEGs

CategoryIDGO termP-valueGenes
BPGO:0007010Cytoskeleton organization0.00CRYAB, PDLIM3, SYNM, CNN1
GO:0009408Response to heat0.04HSPB6, CRYAB
CCGO:0030016Myofibril0.00CRYAB, PDLIM3, SYNM, FLNC
GO:0044449Contractile fiber part0.00CRYAB, PDLIM3, SYNM, FLNC
GO:0030018Z-disc0.00CRYAB, PDLIM3, FLNC
MFGO:0008092Cytoskeletal protein binding0.00CRYAB, PDLIM3, SYNM, CNN1, FLNC
GO:0005198Structural molecule activity0.01CRYAB, PDLIM3, SYNM, COL16A1
GO:0008307Structural constituent of muscle0.03PDLIM3, SYNM

[i] GO, Gene Ontology; DEGs, differentially expressed genes; BP, biological process; CC, cellular component; MF, molecular function.

miRNA-mRNA regulatory network

To generate a catalog of miRNA-mRNA pairings, the predicted target mRNAs of 6 reported miRNAs were intersected with the defined common DEGs. Predication results showed that 4,279, 1,818 and 3,086 target mRNAs, in microcosm, miRTarBase and TargetScan database, respectively, were able to be regulated by the 6 miRNAs. Among the target mRNAs, 93 common mRNAs were identified after intersection between the 3 databases. In addition, 8 miRNA-mRNA pairings were identified with the criteria that the prediction of target mRNAs were also differentially expressed in the microarray analysis. The regulatory network for the 8 miRNA-mRNA pairings are displayed in Fig. 3. Based on Fig. 3, it is evident that miRNA hsa-miR-34a-5p regulates FAM107A and HSPB6, respectively. ALDH1A1 and CNN1 are regulated by hsa-miR-221-3p, hsa-miR-21-5p and hsa-miR-16-5p.

Virtual validation and RT-PCR validation

It is critical for the validation of multi-gene biomarkers in the study of the molecular mechanisms of cancer. To verify the prognostic performance of the identified common DEGs, we firstly employed the SurvExpress online tool which provides survival analysis and risk assessment. Results are shown in Fig. 4 and are summarized in Table III. Survival analysis using Kaplan-Meier curves indicated that the common DEGs were able to significantly differentiate low- and high-risk groups in the 4 datasets, and the p-values were 0.0028, 0.00056, 0.0011 and 0.021, respectively. A higher concordance index (CI) value (Table III; CI >50) also demonstrated that better predication was achieved between low- and high-risk groups.

Table III.

Virtual validation results of the common DEGs in 4 bladder cancer datasets.

Table III.

Virtual validation results of the common DEGs in 4 bladder cancer datasets.

DEG between risk groups
DatasetSamplesGenes foundRisk groups (p-value)CINo.DEGs
TCGA54180.0002987.28C2orf40, CRYAB, DCN, FLNC, HSPB6, PDLIM3, SRPX, SYNM
GSE13507246180.0005678.11COL16A1
GSE528730150.001177.31HSPB6
GSE3168493140.02168.67CNN1, COL16A1, CRYAB, DCN, PDLIM3, SRPX, TCEAL2

[i] DEGs, differentially expressed genes; CI, concordance index.

Furthermore, RT-PCR validation was carried out using 10 specimens from patients to validate the microarray analysis results. Based on the criterion that the DEGs can be regulated by miRNAs and identified in the SurvExpress analysis, a total of 6 DEGs were selected for RT-PCR validation including HSPB6, SRPX, CNN1, FLNC, ALDH1A1 and FAM107A. The mRNA expression values for those selected DEGs were measured in tumor and adjacent normal tissues. The results indicated that the expression levels of HSPB6, FLNC and SRPX were slightly lower in the tumor tissues when compared with these levels in the adjacent normal tissues (Fig. 5); whereas, mRNA expression levels for the remaining 3 DEGs were significant higher in the tumor tissues (Fig. 5). All these results were nearly concordant with the results of the microarray analysis.

Discussion

Microarray and next generation sequencing technologies have significantly advanced the understanding of the molecular mechanisms of bladder cancer. Extensive research concerning mRNA or miRNA expression, and whole-genome or exome sequencing, has been widely carried out in the past few years. However, efforts that attempt to unveil the complex mechanisms of bladder cancer using the integration of different omic data are rare. In the present study, a total of 18 genes were found to be simultaneously differentially expressed in 206 tumor tissues, and 10 were upregulated and 8 were downregulated. Notably, the tumor and normal samples were able to be clearly classified into 3 groups based on these common DEGs. Several normal samples were assigned into the tumor cluster incorrectly, which was probably caused by tumor heterogeneity or sample quality.

Pathway enrichment analysis revealed that the common DEGs were involved in the TGF-β signaling pathway. Although the molecular mechanism between bladder cancer and the TGF-β signaling pathway is unclear, TGF-β1 production has been demonstrated to be significantly associated with the phenotype of bladder cancer (18). RT-PCR results showed that BMP2 and INHBB within the TGF-β signaling pathway were significantly associated with tumorigenicity (p=0.02) and invasiveness (p=0.04) (19). Another important MAPK pathway was also enriched based on the common DEGs. Jebar et al found that mutually exclusive mutation of FGFR3 or RAS (HRAS or KRAS) can activate the MAPK pathway, and the exact role of FGFR3 in activation of the MAPK pathway warrants further studies (20).

Further analyses based on the mRNA and miRNA regulatory network partially unveiled the complex mechanisms of bladder cancer. The development and progression of bladder cancer probably involve multi-path processes including cancer stem cell processes, downregulation of the expression of tumor-suppressor genes, and promotion of cancer cell migration and angiogenesis (Fig. 6).

Recently, extensive research has focused on the study of cancer stem cells, which are believed to contribute to tumor self-renewal and metastasis (21). In the present study, it was found that putative cancer stem cell biomarker ALDH1A1 probably contributes to the poor prognosis of bladder cancer. In addition, based on the miRNA regulation network, the upregulation of ALDH1A1 is likely to be the consequence of the downregulation of the expression of miR-221-3p and miR-21-5p. ALDH1A1 combined with CD44 has been identified as a promising cancer stem cell biomarker in various types of cancers (2224). Keymoosi et al demonstrated that ALDH1A1 was highly expressed in almost 16% (25/159) of bladder cancer cases based on immunohistochemistry. In addition, high expression of ALDH1A1 was found to be significantly correlated with clinical characteristics such as tumor size (p=0.002), pathologic stage (T1, p=0.007 and T2, p-value<0.001) and high recurrence rate (p=0.013) (22). In ovarian cancer, knockdown of ALDH1A1 lead to S and G2 phase cell accumulation via marked decrease in KLF4 and p21 (25). In addition, DNA damage was also increased after ALDH1A1 knockdown evidenced by induction of γ-H2AX and BAX-mediated apoptosis (25). All of these findings indicate that ALDH1A1 participates in the regulation of cell cycle checkpoints and DNA repair networks in cancer stem-like cells (25).

Moreover, downregulation of the expression of tumor-suppressor genes also plays an important role in tumor development and metastasis. In the present study, downregulation of the expression of SRPX and FLNC was identified, which probably was inhibited by miR-200c-5p or miR-16-5p and miR-205-5p, respectively. SRPX was firstly isolated as a novel transformation suppressor gene, and SRPX expression is downregulated by retroviral oncogenes such as v-src or v-ras (26). Research has demonstrated that SRPX expression is markedly reduced in various human cancer cell lines (26,27). Tambe et al documented that the C-terminal region and the 3 consensus repeats in the N-terminal region of SRPX are critical in SRPX-induced apoptosis. Low expression of SRPX can sequentially activate caspase-12, −9 and −3 rather than the mitochondrial pathway and induce apoptosis (28). Furthermore, SRPX can interact with apoptosis-inducing protein ASY/Nogo-B/RTN-xS leading to apoptosis (28). FLNC as one member of the filamin protein family can crosslink actin filaments into orthogonal networks (29). The promoter of FLNC has been frequently identified to be methylated in several types of human cancers such as prostate (30,31), ovarian (32,33) and gastric cancer (34). Kaneda et al showed that >66% of gastric cancer cell lines are methylated in one CpG island of the FLNC promoter, and the methylation is considered as the result of H. pylori infection (34). In addition, the methylation of the FLNC promoter is expected to impair its mRNA expression (35). In 2014, Qiao et al reported that downregulation of the expression of endogenous FLNC can promote cancer cell migration and invasion, and opposite effects were observed with high expression of FLNC (36). Moreover, the downregulation of the expression of FLNC can induce the upregulation of matrix metallopeptidase 2 which participates in the degradation of extracellular matrix and cancer metastasis (37).

In addition, another important factor that can promote cancer cell proliferation and metastasis is angiogenesis which can provide adequate oxygen and nutrient supplies (38). In the present study, angiogenesis was activated by the high expression of HspB6. In addition, based on the miRNA regulation network, the upregulation of HspB6 was likely to be the consequence of miR-34a-5p. HspB6, also referred to as hsp20, has been reported to be activated in physiological or pathological stress (39,40) and non-small cell lung cancer (41). Wang et al showed that HspB6 can promote growth factor secretion including VEGF and bFGF and induce myocardial angiogenesis (42). The angiogenesis process may be promoted via activation of VEGFR2 by HspB6 based on protein binding assay and immunostaining results (43). However, the complex regulatory mechanism of HspB6 in bladder cancer remains to be explored.

In summary, the development and progression of bladder cancer are induced via various processes. Firstly, the cell cycle checkpoints and DNA repair networks in cancer stem-like cell are regulated by high expression of ALDH1A1, and hence promote tumor self-renewal or metastasis. Then, activation of HspB6 promotes angiogenesis which can provide necessary nutrients and oxygen for tumor cells. Finally, downregulation of the expression of SRPX and FLNC further promote apoptosis and metastasis.

Acknowledgements

The present study was supported by grants from the National Natural Science Foundation of China (nos. 81571395, 81371748 and 81373075).

References

1 

Ferlay J, Shin HR, Bray F, Forman D, Mathers C and Parkin DM: Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 127:2893–2917. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, Abraham J, Adair T, Aggarwal R, Ahn SY, et al: Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: A systematic analysis for the Global Burden of Disease Study 2010. Lancet. 380:2095–2128. 2012. View Article : Google Scholar : PubMed/NCBI

3 

National Cancer Institute Surveillance, . Epidemiology, and End Results Program: SEER stat fact sheets. Bladder Cancer; 2014, http://seer.cancer.gov/statfacts/html/urinb.html

4 

Knowles MA and Hurst CD: Molecular biology of bladder cancer: New insights into pathogenesis and clinical diversity. Nat Rev Cancer. 15:25–41. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Cancer Genome Atlas Research Network: Comprehensive molecular characterization of urothelial bladder carcinoma. Nature. 507:315–322. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Berger B, Peng J and Singh M: Computational solutions for omics data. Nat Rev Genet. 14:333–346. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Kim WJ, Kim EJ, Kim SK, Kim YJ, Ha YS, Jeong P, Kim MJ, Yun SJ, Lee KM, Moon SK, et al: Predictive value of progression-related gene classifier in primary non-muscle invasive bladder cancer. Mol Cancer. 9:32010. View Article : Google Scholar : PubMed/NCBI

8 

Kim YJ, Yoon HY, Kim JS, Kang HW, Min BD, Kim SK, Ha YS, Kim IY, Ryu KH, Lee SC, et al: HOXA9, ISL1 and ALDH1A3 methylation patterns as prognostic markers for nonmuscle invasive bladder cancer: Array-based DNA methylation and expression profiling. Int J Cancer. 133:1135–1142. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Lee JS, Leem SH, Lee SY, Kim SC, Park ES, Kim SB, Kim SK, Kim YJ, Kim WJ and Chu IS: Expression signature of E2F1 and its associated genes predict superficial to invasive progression of bladder tumors. J Clin Oncol. 28:2660–2667. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, Ellis B, Gautier L, Ge Y, Gentry J, et al: Bioconductor: Open software development for computational biology and bioinformatics. Genome Biol. 5:R802004. View Article : Google Scholar : PubMed/NCBI

11 

Kerr MK: Linear models for microarray data analysis: Hidden similarities and differences. J Comput Biol. 10:891–901. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC and Lempicki RA: DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol. 4:P32003. View Article : Google Scholar : PubMed/NCBI

13 

Acunzo M and Croce CM: MicroRNA in cancer and cachexia - a mini-review. J Infect Dis. 212 Suppl 1:S74–S77. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Sapre N, Macintyre G, Clarkson M, Naeem H, Cmero M, Kowalczyk A, Anderson PD, Costello AJ, Corcoran NM and Hovens CM: A urinary microRNA signature can predict the presence of bladder urothelial carcinoma in patients undergoing surveillance. Br J Cancer. 114:454–462. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Kutmon M, Kelder T, Mandaviya P, Evelo CT and Coort SL: CyTargetLinker: A cytoscape app to integrate regulatory interactions in network analysis. PLoS One. 8:e821602013. View Article : Google Scholar : PubMed/NCBI

16 

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

17 

Aguirre-Gamboa R, Gomez-Rueda H, Martínez-Ledesma E, Martínez-Torteya A, Chacolla-Huaringa R, Rodriguez-Barrientos A, Tamez-Peña JG and Treviño V: SurvExpress: An online biomarker validation tool and database for cancer gene expression data using survival analysis. PLoS One. 8:e742502013. View Article : Google Scholar : PubMed/NCBI

18 

Zhuang J, Lu Q, Shen B, Huang X, Shen L, Zheng X, Huang R, Yan J and Guo H: TGFβ1 secreted by cancer-associated fibroblasts induces epithelial-mesenchymal transition of bladder cancer cells through lncRNA-ZEB2NAT. Sci Rep. 5:119242015. View Article : Google Scholar : PubMed/NCBI

19 

Hung TT, Wang H, Kingsley EA, Risbridger GP and Russell PJ: Molecular profiling of bladder cancer: Involvement of the TGF-beta pathway in bladder cancer progression. Cancer Lett. 265:27–38. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Jebar AH, Hurst CD, Tomlinson DC, Johnston C, Taylor CF and Knowles MA: FGFR3 and Ras gene mutations are mutually exclusive genetic events in urothelial cell carcinoma. Oncogene. 24:5218–5225. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Kreso A and Dick JE: Evolution of the cancer stem cell model. Cell Stem Cell. 14:275–291. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Keymoosi H, Gheytanchi E, Asgari M, Shariftabrizi A and Madjd Z: ALDH1 in combination with CD44 as putative cancer stem cell markers are correlated with poor prognosis in urothelial carcinoma of the urinary bladder. Asian Pac J Cancer Prev. 15:2013–2020. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Steg AD, Bevis KS, Katre AA, Ziebarth A, Dobbin ZC, Alvarez RD, Zhang K, Conner M and Landen CN: Stem cell pathways contribute to clinical chemoresistance in ovarian cancer. Clin Cancer Res. 18:869–881. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Marcato P, Dean CA, Giacomantonio CA and Lee PW: Aldehyde dehydrogenase: Its role as a cancer stem cell marker comes down to the specific isoform. Cell Cycle. 10:1378–1384. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Meng E, Mitra A, Tripathi K, Finan MA, Scalici J, McClellan S, da Silva L Madeira, Reed E, Shevde LA, Palle K, et al: ALDH1A1 maintains ovarian cancer stem cell-like properties by altered regulation of cell cycle checkpoint and DNA repair network signaling. PLoS One. 9:e1071422014. View Article : Google Scholar : PubMed/NCBI

26 

Mukaisho K, Suo M, Shimakage M, Kushima R, Inoue H and Hattori T: Down-regulation of drs mRNA in colorectal neoplasms. Jpn J Cancer Res. 93:888–893. 2002. View Article : Google Scholar : PubMed/NCBI

27 

Kim CJ, Shimakage M, Kushima R, Mukaisho K, Shinka T, Okada Y and Inoue H: Down-regulation of drs mRNA in human prostate carcinomas. Hum Pathol. 34:654–657. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Tambe Y, Isono T, Haraguchi S, Yoshioka-Yamashita A, Yutsudo M and Inoue H: A novel apoptotic pathway induced by the drs tumor suppressor gene. Oncogene. 23:2977–2987. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Duff RM, Tay V, Hackman P, Ravenscroft G, McLean C, Kennedy P, Steinbach A, Schöffler W, van der Ven PF, Fürst DO, et al: Mutations in the N-terminal actin-binding domain of filamin C cause a distal myopathy. Am J Hum Genet. 88:729–740. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Vanaja DK, Ehrich M, van den Boom D, Cheville JC, Karnes RJ, Tindall DJ, Cantor CR and Young CY: Hypermethylation of genes for diagnosis and risk stratification of prostate cancer. Cancer Invest. 27:549–560. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Mahapatra S, Klee EW, Young CY, Sun Z, Jimenez RE, Klee GG, Tindall DJ and Donkena KV: Global methylation profiling for risk prediction of prostate cancer. Clin Cancer Res. 18:2882–2895. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Imura M, Yamashita S, Cai LY, Furuta J, Wakabayashi M, Yasugi T and Ushijima T: Methylation and expression analysis of 15 genes and three normally-methylated genes in 13 Ovarian cancer cell lines. Cancer Lett. 241:213–220. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Zeller C, Dai W, Steele NL, Siddiq A, Walley AJ, Wilhelm-Benartzi CS, Rizzo S, van der Zee A, Plumb JA and Brown R: Candidate DNA methylation drivers of acquired cisplatin resistance in ovarian cancer identified by methylome and expression profiling. Oncogene. 31:4567–4576. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Kaneda A, Kaminishi M, Yanagihara K, Sugimura T and Ushijima T: Identification of silencing of nine genes in human gastric cancers. Cancer Res. 62:6645–6650. 2002.PubMed/NCBI

35 

Oue N, Mitani Y, Motoshita J, Matsumura S, Yoshida K, Kuniyasu H, Nakayama H and Yasui W: Accumulation of DNA methylation is associated with tumor stage in gastric cancer. Cancer. 106:1250–1259. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Qiao J, Cui SJ, Xu LL, Chen SJ, Yao J, Jiang YH, Peng G, Fang CY, Yang PY, Liu F and Filamin C: A dysregulated protein in cancer revealed by label-free quantitative proteomic analyses of human gastric cancer cells. Oncotarget. 6:1171–1189. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Kessenbrock K, Plaks V and Werb Z: Matrix metalloproteinases: Regulators of the tumor microenvironment. Cell. 141:52–67. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Nishida N, Yano H, Nishida T, Kamura T and Kojiro M: Angiogenesis in cancer. Vasc Health Risk Manag. 2:213–219. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Latchman DS: Heat shock proteins and cardiac protection. Cardiovasc Res. 51:637–646. 2001. View Article : Google Scholar : PubMed/NCBI

40 

Willis MS and Patterson C: Hold me tight: Role of the heat shock protein family of chaperones in cardiac disease. Circulation. 122:1740–1751. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Chen S, Huang H, Yao J, Pan L and Ma H: Heat shock protein B6 potently increases non-small cell lung cancer growth. Mol Med Rep. 10:677–682. 2014.PubMed/NCBI

42 

Wang X, Zhao T, Huang W, Wang T, Qian J, Xu M, Kranias EG, Wang Y and Fan GC: Hsp20-engineered mesenchymal stem cells are resistant to oxidative stress via enhanced activation of Akt and increased secretion of growth factors. Stem Cells. 27:3021–3031. 2009.PubMed/NCBI

43 

Zhang X, Wang X, Zhu H, Kranias EG, Tang Y, Peng T, Chang J and Fan GC: Hsp20 functions as a novel cardiokine in promoting angiogenesis via activation of VEGFR2. PLoS One. 7:e327652012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2017
Volume 37 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li W, Liu J, Zou D, Cai X, Wang J, Wang J, Zhu L, Zhao L, Ou R, Xu Y, Xu Y, et al: Exploration of bladder cancer molecular mechanisms based on miRNA-mRNA regulatory network. Oncol Rep 37: 1461-1468, 2017
APA
Li, W., Liu, J., Zou, D., Cai, X., Wang, J., Wang, J. ... Xu, Y. (2017). Exploration of bladder cancer molecular mechanisms based on miRNA-mRNA regulatory network. Oncology Reports, 37, 1461-1468. https://doi.org/10.3892/or.2017.5433
MLA
Li, W., Liu, J., Zou, D., Cai, X., Wang, J., Wang, J., Zhu, L., Zhao, L., Ou, R., Xu, Y."Exploration of bladder cancer molecular mechanisms based on miRNA-mRNA regulatory network". Oncology Reports 37.3 (2017): 1461-1468.
Chicago
Li, W., Liu, J., Zou, D., Cai, X., Wang, J., Wang, J., Zhu, L., Zhao, L., Ou, R., Xu, Y."Exploration of bladder cancer molecular mechanisms based on miRNA-mRNA regulatory network". Oncology Reports 37, no. 3 (2017): 1461-1468. https://doi.org/10.3892/or.2017.5433