Open Access

A novel tumor suppressing gene, ARHGAP9, is an independent prognostic biomarker for bladder cancer

  • Authors:
    • Xuan‑Mei Piao
    • Pildu Jeong
    • Chunri Yan
    • Ye‑Hwan Kim
    • Young Joon Byun
    • Yanjie Xu
    • Ho Won Kang
    • Sung Phil Seo
    • Won Tae Kim
    • Jong‑Young Lee
    • Isaac Y. Kim
    • Sung‑Kwon Moon
    • Yung Hyun Choi
    • Eun‑Jong Cha
    • Seok Joong Yun
    • Wun‑Jae Kim
  • View Affiliations

  • Published online on: November 19, 2019     https://doi.org/10.3892/ol.2019.11123
  • Pages: 476-486
  • Copyright: © Piao 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

Screening for genes or markers relevant to bladder cancer (BC) tumorigenesis and progression is of vital clinical significance. The present study used reverse‑transcription quantitative PCR reaction assays to examine the expression of mRNA encoding Rho GTPase‑activating protein 9 (ARHGAP9) in BC tissue samples and to determine whether ARHGAP9 is an independent prognostic biomarker for non‑muscle invasive BC (NMIBC) and muscle invasive BC (MIBC). The results revealed that the downregulation of ARHGAP9 expression in the tissue of patients with NMIBC or MIBC was significantly associated with a poor prognosis. In patients with NMIBC, a high expression of ARHGAP9 was significantly associated with prolonged recurrence‑free survival, whereas in MIBC patients, it was significantly associated with an increased progression‑free and cancer‑specific survival. The risk of cancer‑specific death was 2.923 times higher (95% confidence interval, 1.192‑7.163) when ARHGAP9 levels were decreased. In conclusion, lower expressions of ARHGAP9 correlated with BC prognosis, indicating that it may be a useful marker for guiding treatment application.

Introduction

Bladder cancer (BC), one of the most common malignancies worldwide, is classified into two subtypes based on cancer cell infiltration into the muscle layer of the bladder. Non-muscle invasive BC (NMIBC) is less aggressive but has a high recurrence rate, whereas muscle invasive BC (MIBC) tends to metastasize and has a relatively poor prognosis (13). High throughput techniques such as microarray analysis and next generation sequencing, which are used commonly in the fields of genetics and epigenetics, have identified several genes involved in cancer pathogenesis, and have led to identification of cancer biomarkers and to development of novel effective gene targeted therapies (4). In a previous study, we used next generation sequencing and miRNA microarray assays to identify several miRNAs and their target genes that are differentially expressed in BC (5). We found that a novel gene, Rho GTPase-activating protein 9 (ARHGAP9), is down-regulated in BC. In addition, hsa-miR-3620, which interacts with ARHGAP9, is up-regulated.

Rho GTPases are key regulators of the actin cytoskeleton, which plays an important role in cell adhesion and migration. The switch mechanism of Rho GTPases is controlled by binding to GTP or GDP (68). ARHGAP9 contains a diverse combination of functional protein domains, including the RhoGAP, SH3, WW, and PH domains (9). Binding of the RhoGAP domain to GTP-bound Rho proteins accelerates GTPase activity, and defective Rho GTPase signaling is implicated in tumorigenesis and metastasis (10,11). Silencing ARHGAP9 inhibits proliferation, migration, and invasion of breast cancer cells (12). Activated ARHGAP9 inhibits adhesion of a human leukemia cell line, KG-1, to fibronectin and collagen through activation of cdc42 and Rac1 but not RhoA (6).

Here, we asked whether ARHGAP9 is a novel prognostic biomarker for BC. We used real-time polymerase chain reaction (PCR) to compare expression of ARHGAP9 mRNA in human BC and control tissues (the latter comprised normal tissue surrounding BC and normal bladder mucosa); and analyzed its ability to predict prognosis of NMIBC and MIBC. ARHGAP9, known as a MAP kinase docking protein, was encoded by ARHGAP9 gene, which shares 16 bases with Gli1 in their 3′ ends (9,13). Accordingly, we asked whether ARHGAP9 plays a role in the MAPK and Hedgehog signaling pathways.

Materials and methods

Patients and tissue samples

The biospecimens used in the present study were provided by the Chungbuk National University Hospital, a member of the National Biobank of Korea, which is supported by the Ministry of Health, Welfare, and Family Affairs. The study was approved by the Institutional Review Board at Chungbuk National University (GR2010-12-010), and the experiments were undertaken with the informed written consents of all participants. The study methodologies conformed with the standards set by the Declaration of Helsinki. The baseline characteristics of the case subjects (n=237 bladder tissue samples) are shown in Table I. Among these, 140 samples were from primary BC patients and were histologically verified as transitional cell carcinomas; the remaining 97 samples used as the control set comprised normal bladder mucosa or normal tissues from the area surrounding BC. To reduce the chances of confounding factors affecting the analyses, patients diagnosed with concomitant carcinoma in situ or carcinoma in situ lesions alone were excluded. Voided urine cytology was tested before surgical treatment to assist BC diagnosis and/or prognosis. Fresh-frozen specimens were obtained during surgical resection of transitional cell carcinoma at Chungbuk National University Hospital. All tumors were macro-dissected, typically within 15 min of surgical resection. Each specimen was confirmed by pathological analysis of a part of fresh-frozen specimens obtained from radical cystectomy and transurethral resection of bladder tumor (TURBT). Tumors were staged (2002 TNM Classification) and graded (2004 WHO Classification), according to standard criteria (14). Clinically metastatic disease and non-cystectomy cases were not excluded from the study. Each patient was followed and managed suggested management according to standard recommendations (1517). Surveillance was performed by cystoscopic examination and upper urinary tract imaging in accordance with European Association of Urology guidelines (16). Recurrence was defined as relapse of primary NMIBC of the same pathologic stage, and progression of NMIBC and MIBC was defined as TNM stage progression after disease recurrence. The mean follow-up period for NMIBC patients was 72.95 months (range, 3.2–172.2). The mean follow-up period for MIBC patients was 36.18 months (range, 3.0–141.4).

Table I.

Clinicopathological features of primary BC patient and control tissues (surrounding normal tissues and normal bladder mucosae).

Table I.

Clinicopathological features of primary BC patient and control tissues (surrounding normal tissues and normal bladder mucosae).

BC (140)

VariablesNMIBCMIBCControlP-value
No.974397
Mean age ± SD63.45±13.7967.60±9.8461.98±14.320.083a
Sex (%) 0.975a
  Male80 (82.5%)36 (83.7%)81 (83.5%)
  Female17 (17.5%)7 (16.3%)16 (16.5%)
Operation (%) <0.001b
  TUR-BT97 (100.0%)17 (39.5%)
  Radical cystectomy026 (60.5%)
Tumor size (%) 0.003b
  ≤1 cm16 (16.5%)2 (4.7%)
  2–3 cm37 (38.1%)11 (25.6%)
  >3 cm37 (38.1%)28 (65.1%)
Multiplicity (%) 0.108b
  Single52 (53.6%)30 (69.8%)
  2–728 (28.9%)7 (16.3%)
  >711 (11.3%)4 (9.3%)
Grade, 2004 WHO grading system (%) <0.001b
  Low72 (74.2%)8 (18.6%)
  High25 (25.8%)35 (81.4%)
Stage (%) <0.001b
  TaN0M026 (26.8%)
  T1N0M071 (73.2%)
  T2N0M0 13 (30.2%)
  T3N0M0 6 (14.0%)
T≥4 or N≥1 or M1 24 (55.8%)
Chemotherapy (%) <0.001b
  No97 (100.0%)23 (53.5%)
  Yes020 (46.5%)
BCG therapy (%) <0.001b
  No56 (57.7%)38 (88.4%)
  Yes40 (41.2%)5 (11.6%)
Recurrence, no. of patients (%)
  No59 (60.8%)
  Yes38 (39.2%)
Progression, no. of patients (%) 0.126b
  No79 (81.4%)30 (69.8%)
  Yes18 (18.6%)13 (30.2%)
Survival, no. of patients (%) 0.009b
  Alive64 (66.0%)21 (48.8%)
  Non-cancer-specific death18 (18.6%)3 (7.0%)
  Cancer-specific death15 (15.5%)19 (44.2%)
  Mean follow-up, months (range)72.95 (3.20–172.20)36.18 (3.00–141.40)

a P-value obtained using Kruskal-Wallis H test (BC compared with control).

b P-value obtained using the Mann-Whitney U test (NMIBC compared with MIBC). BC, bladder cancer; BCG, Bacillus Calmette-Guerin; NMIBC, non-muscle invasive bladder cancer; MIBC, muscle invasive bladder cancer; SD, standard deviation.

RNA extraction

Total RNA was extracted from tissues using TRIzol reagent (Invitrogen), as described previously (18), and stored at −80°C. Next, cDNA was synthesized from 1 µg of total RNA using a First Strand cDNA Synthesis kit (Clontech, TAKARA), according to the manufacturer's protocol.

Microarray analysis

Five hundred nanograms of total RNA was used for labeling and hybridization prior to analysis, according to the manufacturer's protocols (Illumina). After the bead chips were scanned with an Illumina Bead Array Reader, the Robust Multiarray Average in R package was used to perform global correction, quantile normalization, and median polish summarization of the microarray data. P-values (t test) were calculated from bead mRNA signal intensities (1921). The full set of microarray data set are available online at http://www.ncbi.nlm.nih.gov/geo/under data series accession number GSE13507 (21).

mRNA sequencing

Total sequencing reads were subjected to preprocessing as follows: Adapter trimming was performed using cutadapt with default parameters, and quality trimming (Q30) was performed using FastQC with default parameters. Processed reads were mapped to the human reference genome [Ensembl 72 (GRCh37: hg19)] using tophat and cufflink with default parameters (22). Fragments Per Kilobase of exon per million fragments Mapped (FPKM) values were normalized and quantitated using R package Tag Count Comparison (TCC) (23) to determine statistical significance (e.g., P and Q values) and differential expression (e.g., -fold changes).

Quantitative PCR analysis

Tissue mRNAs were amplified by quantitative PCR performed using a Rotor Gene 6000 instrument (Qiagen) and quantified using the 2−∆∆cq method (24). QuantitativePCR reactions were carried out using the SYBR Premix Ex Taq II (Clontech, TAKARA). The following primers were used to amplify candidate genes: ARHGAP9 (Gene ID: ENSG00000123329), sense, 5′-CAGAGCAGTGCCTCTCTC-3′ (18 bp, Tm 58°C); antisense, 5′-CTGCTGGGTCAGATGTCTC-3′ (19 bp, Tm 58°C) and the amplicon size was 179 bp. The control GAPDH (Gene ID: ENSG00000111640) primers were as follows: sense, 5′-CATGTTCGTCATGGGTGTGA-3′ (20 bp, Tm 60°C); antisense, 5′-ATGGCATGGACTGTGGTCAT-3′ (20 bp, Tm 60°C) and the amplicon size was 156 bp. The PCR reaction was performed in a final volume of 10 µl, comprising 5 µl of 2× SYBR Premix EX Taq buffer, 0.5 µl of each 5′and 3′ primer (10 pM/µl), and 2 µl, of sample cDNA. A known concentration of the PCR product was then 10-fold serially diluted from 100 pg/µl to 0.1 pg/µl and used to establish a standard curve. The real-time PCR conditions were as follows: 1 cycle at 96°C for 20 sec, followed by 40 cycles of 3 sec at 96°C for denaturation, 15 sec at 60°C for annealing, and 15 sec at 72°C for extension. The melting program was performed at 72–95°C at a heating rate of 1°C per 45 sec. Rotor-Gene Q software 2.3.1.49 was used for capturing and analyzing spectral data. All samples were run in triplicate. Gene expression was normalized to the expression of GAPDH.

Statistical analysis

To reduce variation among microarrays, the intensity values for each microarray were rescaled using a quantile normalization method (19). Gene expression values were loge-transformed and median-centered across samples. The significance of various clinicopathological variables was evaluated using univariate and multivariate Cox proportional hazard regression models. Hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated to investigate relative risk. Survival curves to determine the prognostic value of the genetic biomarker were plotted using the Kaplan-Meier method and compared using the log-rank test. The Kruskal-Wallis H test and Mann-Whitney U test were used to examine expression of ARHGAP9 in BC tissues versus control tissues. Correlations between ARHGAP9 and genes involved in the MAPK and Hedgehog signaling pathways were examined by calculating non-parametric Spearman's correlation coefficients. Statistical analyses were performed using IBM SPSS Statistics ver. 20.0 (IBM) and GraphPad Prism 7 (GraphPad Software). P-values <0.05 were considered significant.

Results

Expression of ARHGAP9 mRNA in BC tissue

Microarray analysis revealed that expression of mRNA encoding ARHGAP9 in BC tissues was lower than that in control samples. The validation test showed that the real-time PCR results were identical to those of the microarray, i.e., expression of mRNA encoding ARHGAP9 was significantly lower in NMIBC and MIBC tissues than in normal control tissues (P<0.001; Fig. 1).

Expression of ARHGAP9 correlates with NMIBC prognosis

Univariate and multivariate Cox regression analyses revealed that expression of ARHGAP9 in NMIBC patients was an independent predictor of recurrence-free survival (RFS) (HR, 2.436; 95% CI, 1.132–5.243; P=0.023; Table II). Kaplan-Meier analysis demonstrated that NMIBC patients with ARHGAP9 expression levels in the upper 50th percentile experienced less recurrence than those with expression levels in the lower 50th percentile (log-rank test, P=0.043; Fig. 2A). Particularly, for T1 high grade(HG) BC patients, univariate and multivariate Cox regression analysis identified ARHGAP9 expression as an independent risk factor for T1HG BC recurrence (HR, 7.264; 95% CI, 1.291–45.091; P=0.025) and progression (HR, 14.987; 95% CI, 1.093–205.567; P=0.043; Table III). The RFS and progression-free survival (PFS) of T1HG BC patients with ARHGAP9 expression levels in the upper 50th percentile experienced less recurrence and progression than those with expression levels in the lower 50th percentile (log-rank test, P=0.013 and 0.026 respectively; Fig. 2B and C).

Table II.

Univariate and multivariate Cox regression analysis to predict NMIBC recurrence.

Table II.

Univariate and multivariate Cox regression analysis to predict NMIBC recurrence.

Univariate Cox analysisMultivariate Cox analysis


VariablesHR (95% CI)P-valueHR (95% CI)P-value
Age
≤70 (Ref.) vs. >702.994 (1.579–5.680)0.001a1.727 (0.820–3.637)0.151
Sex
Male (Ref.) vs. female1.314 (0.577–2.993)0.516
Tumor size
  ≤1 cmRef.0.028aRef.0.574
  2–3 cm1.700 (0.474–6.100)0.4161.251 (0.341–4.593)0.736
  >3 cm3.686 (1.093–12.425)0.035a1.779 (0.484–6.547)0.386
Multiplicity
  SingleRef.0.141
  2–71.071 (0.479–2.395)0.867
  >72.383 (0.985–5.767)0.054
2004 WHO Grade Low (Ref.) vs. high2.450 (1.275–4.708)0.007a1.823 (0.809–3.568)0.147
Stage
Ta (Ref.) vs. T12.938 (1.144–7.540)0.025a2.347 (0.803–6.857)0.119
BCG
No (Ref.) vs. yes1.918 (1.009–3.647)0.047a1.744 (0.852–3.568)0.128
ARHGAP9 expression High expression (Ref.) vs. Low expression1.939 (1.009–3.726)0.047a2.436 (1.132–5.243)0.023a

a P<0.05. NMIBC, non-muscle invasive bladder cancer; BCG, Bacillus Calmette-Guerin; CI, confidence interval; HR, hazard ratio; Ref., reference; ARHGAP9, Rho GTPase-activating protein 9.

Table III.

Univariate and multivariate Cox regression analysis to predict T1 high grade NMIBC recurrence and progression.

Table III.

Univariate and multivariate Cox regression analysis to predict T1 high grade NMIBC recurrence and progression.

RecurrenceProgression


Univariate Cox analysisMultivariate Cox analysisUnivariate Cox analysisMultivariate Cox analysis




VariablesHR (95% CI)P-valueHR (95% CI)P-valueHR (95% CI)P-valueHR (95% CI)P-value
Age
≤70 (Ref.) vs. >702.342 (0.625–8.776)0.207 1.567 (0.390–6.297)0.527
Sex
Male (Ref.) vs. female1.327 (0.342–5.154)0.682 2.748 (0.548–13.781)0.219
Tumor size
  ≤1 cmRef.0.976 Ref.0.468
  2–3 cm29604.1040.948 9687.8840.968
(0.000–2.839×10138) (0.000–3.269×10201)
  >3 cm25622.2700.949 36480.7410.964
(0.000–2.454×10138) (0.000–1.226×10202)
Multiplicity
  SingleRef.0.618 Ref.0.850
  2–71.450 (0.417–5.040)0.559 1.548 (0.345–6.943)0.568
  >72.933 (0.296–29.074)0.358 0.000 (0.000–0.000)0.991
BCG
No (Ref.) vs. yes1.247 (0.336–4.624)0.741 0.459 (0.119–1.766)0.257
ARHGAP9 expression High (Ref.) vs. low expression5.126 (1.247–21.066)0.023a7.2640.025a6.041 (1.026–35.571)0.047a14.9870.043a
(1.291–45.019) (1.093–205.567)

a P<0.05. NMIBC, non-muscle invasive bladder cancer; BCG, Bacillus Calmette-Guerin; CI, confidence interval; HR, hazard ratio; Ref., reference; ARHGAP9, Rho GTPase-activating protein 9.

Expression of ARHGAP9 correlates with MIBC prognosis

For MIBC patients, univariate and multivariate Cox regression analysis identified ARHGAP9 expression as an independent risk factor for disease progression (HR, 5.241; 95% CI, 1.456–18.870; P=0.011) and cancer-specific death (HR, 2.923; 95% CI, 1.192–7.163; P=0.019) (Tables IV and V). PFS and cancer specific survival (CSS) of patients with ARHGAP9 expression in the upper 50th percentile were significantly higher than those of patients in the lower 50th percentile (log-rank test, P=0.020 and 0.031, respectively; Fig. 3A and B).

Table IV.

Univariate and multivariate Cox regression analysis to predict MIBC progression.

Table IV.

Univariate and multivariate Cox regression analysis to predict MIBC progression.

Univariate Cox analysisMultivariate Cox analysis


VariablesHR (95% CI)P-valueHR (95% CI)P-value
Age
≤70 (Ref.) vs. >701.302 (0.432–3.926)0.639
Sex
Male (Ref.) vs. female5.625 (1.766–17.912)0.003a7.255 (2.062–25.528)0.002a
Operation
TURBT (Ref.) vs. Radical cystectomy0.948 (0.309–2.905)0.926
Tumor size
  ≤1 cmRef.0.406
  2–3 cm12417.036 (0.000–2.033×10143)0.954
  >3 cm35009.555 (0.000–5.718×10143)0.949
Multiplicity
  SingleRef.0.507
  2–70.358 (0.046–2.800)0.328
  >71.483 (0.324–6.787)0.611
2004 WHO Grade
Low (Ref.) vs. high31.010 (0.132–7298.224)0.218
Stage
T2Ref.0.851
T31.630 (0.297–8.958)0.574
T4 or N1 or M11.229 (0.358–4.222)0.744
Chemotherapy
No (Ref.) vs. yes3.912 (1.076–14.218)0.038a2.859 (0.752–10.868)0.123
ARHGAP9 expression High expression (Ref.) vs. Low expression3.818 (1.145–12.733)0.029a5.241 (1.456–18.870)0.011a

a P<0.05. MIBC, muscle invasive bladder cancer; CI, confidence interval; HR, hazard ratio; Ref., reference; ARHGAP9, Rho GTPase-activating protein 9.

Table V.

Univariate and multivariate Cox regression analysis for predicting the cancer-specific survival of patients with MIBC.

Table V.

Univariate and multivariate Cox regression analysis for predicting the cancer-specific survival of patients with MIBC.

Univariate Cox analysisMultivariate Cox analysis


VariablesHR (95% CI)P-valueHR (95% CI)P-value
Age
≤70 (Ref.) vs. >701.860 (0.791–4.371)0.155
Gender
Male (Ref.) vs. female3.379 (1.273–8.967)0.014a4.046 (1.491–10.976)0.006a
Operation
TURBT (Ref.) vs. Radical cystectomy1.026 (0.435–2.417)0.954
Tumor size
  ≤1 cmRef.0.386
  2–3 cm14923.217 (0.000–1.565E+115)0.941
  >3 cm32178.497 (0.000–3.369E+115)0.937
Multiplicity
  SingleRef.0.730
  2–70.709 (0.206–2.438)0.585
  >70.611 (0.137–2.725)0.519
2004 WHO Grade
Low (Ref.) vs. high3.009 (0.699–12.950)0.139
Stage
  T2Ref.0.480
  T30.909 (0.181–4.563)0.908
  T4 or N1 or M11.671 (0.641–4.358)0.294
Chemotherapy
No (Ref.) vs. yes1.482 (0.633–3.472)0.365
ARHGAP9 expression High expression (Ref.) vs. Low expression2.554 (1.058–6.163)0.037a2.923 (1.192–7.163)0.019a

a P<0.05. MIBC, muscle invasive bladder cancer; CI, confidence interval; HR, hazard ratio; Ref., reference; ARHGAP9, Rho GTPase-activating protein 9.

Relationship between ARHGAP9 and genes regulating the MAPK and Hedgehog signaling pathways in BC

To identify whether expression of ARHGAP9 correlates with that of genes regulating the MAPK and Hedgehog signaling pathways, we undertook gene network depiction and analysis using the GeneMANIA (http://www.genemania.org) web tool. We selected seven genes (ARHGAP9, epidermal growth factor receptor (EGFR), mitogen-activated protein kinase 1 (MAPK1, also known as ERK2), mitogen-activated protein kinase 14 (MAPK14, also known as p38α), mitogen-activated protein kinase kinase 3 (MKK3), mitogen-activated protein kinase kinase 6 (MKK6), and glioma-associated oncogene homolog 1 (Gli1)) showing potential inter-correlations (Supplementary Fig. S1). Non-parametric Spearman's correlation coefficients (based on microarray data) identified interactions among ARHGAP9, EGFR, MAPK1 (ERK2), MAPK14 (p38α), MKK3, MKK6, and Gli1. Table VI shows that expression of ARHGAP9 correlated positively with that of Gli1, which regulates the Hedgehog signaling pathway. In addition, ARHGAP9 interacted with MKK6 and MAPK1 (ERK2), both of which are essential components of the MAPK signal transduction pathway (P<0.05 for both).

Table VI.

Spearman correlation coefficients of Gli1, ARHGAP9, EGFR, MKK3, MKK6, MAPK1 (ERK2) and MAPK14 (p38α) in BC.

Table VI.

Spearman correlation coefficients of Gli1, ARHGAP9, EGFR, MKK3, MKK6, MAPK1 (ERK2) and MAPK14 (p38α) in BC.

Gli1ARHGAP9EGFRMKK3MKK6MAPK1 (ERK2)MAPK14 (p38α)
Gli1
  Spearman's Rho1.0000.518b−0.0090.099−0.0420.178a−0.202b
  P-value.0.0000.9110.2050.5890.0220.009
ARHGAP9
  Spearman's Rho0.518b1.0000.0840.125−0.168a0.233b−0.138
  P-value0.000.0.2830.1090.0310.0030.076
EGFR
  Spearman's Rho−0.0090.0841.0000.194a−0.1180.301b0.192b
  P-value0.9110.283.0.0120.1300.0000.013
MKK3
  Spearman's Rho0.0990.1250.194a1.0000.1010.327b0.315b
  P-value0.2050.1090.012.0.1950.0000.000
MKK6
  Spearman's Rho−0.042−0.168a−0.1180.1011.000−0.093−0.056
  P-value0.5890.0310.1300.195.0.2330.472
MAPK1(ERK2)
  Spearman's Rho0.178a0.233b0.301b0.327b−0.0931.0000.167a
  P-value0.0220.0030.0000.0000.233.0.032
MAPK14 (p38α)
  Spearman's Rho−0.202b−0.1380.192a0.315b−0.0560.167a1.000
  P-value0.0090.0760.0130.0000.4720.032.

a P<0.05.

b P<0.01. BC, bladder cancer.

Discussion

ARHGAP9 sits adjacent to Gli1 on human chromosome 12q13.3; two genes have overlapping 16 bases in their 3′-ends (13), suggesting that Gli1 and ARHGAP9 may regulate each other. Studies suggest that Gli1 is down-regulated in BC (25); indeed, Gli1 is considered to be the most reliable biomarker of Hedgehog pathway activity (2527). The microarray data presented herein shows that mRNA expression of Gli1 and ARHGAP9 were down-regulated in BC tissues, and that there was a positive correlation between the two (Table VI); this indicates that ARHGAP9, which lies adjacent to Gli1, might be a novel regulator of Gli1.

As a novel MAP kinase docking protein, ARHGAP9 associates specifically with ERK2 and p38α via complementarily charged residues within the WW domain of ARHGAP9 and the CD domains of ERK2 and p38α. This interaction suppresses MAP kinase activation; but does not affect that of RhoGAP (9). MAPK activation is a common event in tumor progression and metastasis. Inhibition of ERK1/2 and p38 MAP kinase pathways in BC could inhibit proliferation and growth (28). The key target in this signal transduction pathway is EGFR, a receptor tyrosine kinase (29). Binding of EGF to EGFR in BC activates EGFR, which is already overexpressed; furthermore, the Ras-MAPK pathway is activated through the MAPK/ERK pathway. This continuous ‘ON’ status of MAPK signaling results in overexpression of MEK2 and MKK3, 4, and 6, which lie upstream of MAP kinase (i.e., ERK2 and p38α) and activate ERK2 and p38α, leading to reduced interaction between ARHGAP9 and ERK2 or p38α in BC (this is probably attributable to competitive displacement by overexpressed docking proteins) (Fig. 4). The microarray data revealed a competitive correlation between expression of ARHGAP9 mRNA and that of MKK6, and a positive correlation between ARHGAP9 and ERK2 (Table VI). These findings suggest that ARHGAP9 acts as a tumor suppressor gene in BC. EGFR acts as a receptor molecule in the MAPK signaling pathway, and is a prognostic marker for many cancer types, including BC (30). Our previous study showed that EGFR is a progression-related gene in MIBC; increased expression of EGFR is associated with a poor prognosis (31). Here, we found that lower expression of ARHGAP9 was related to poor PFS and CSS (Fig. 3A and B), which is consistent with previous results. However, no definitive evidence has been demonstrated on the recurrence rate of MIBC after radical cystectomy, and the definition of local and distant recurrence is not standardized (32). In our preliminary study, twenty-six MIBC patients received radical cystectomy and only three of them were manifested recurrence, such result should be examined in further study with more samples for the statistically significant validation of the survival analysis.

Furthermore, the ARHGAP9 mRNA expression could predict the recurrence of NMIBC, that is, lower expression of ARHGAP9 was related to poor RFS (Fig. 2A). In particular, T1HG BC patients with higher expression of ARHGAP9 experienced less recurrence and progression (Fig. 2B and C). A more careful monitoring and optimal treatment recommendation should be implemented for T1HG BCs because of their highly recurrent nature and risk of progression to MIBC (33), which highlights the strategy for predicting prognosis. This study indicates that ARHGAP9 gene has a good performance in predicting prognosis of T1HG BC patients.

In addition, TCGA data from the Human Pathology Atlas (https://www.proteinatlas.org/ENSG00000123329-ARHGAP9/pathology/tissue/urothelial+cancer) show that BC patients with higher expression of ARHGAP9 mRNA tend to survive longer, though it is not statistically significant (P=0.069). On the basis of the results of this study, we can conclude that ARHGAP9 regulates growth and proliferation of BC by regulating the MAPK signaling pathway. Future studies should use real-time PCR assays to validate the results of microarray tests to confirm reliability of the data. For a better understanding of ARHGAP9, its protein levels in BC should be evaluated and the experimental samples should be increased to reduce the statistical limitations in the future. Moreover, the function of miR-3620, which interacted with ARHGAP9 mRNA, could be clarified by validating the function of ARHGAP9 in the future.

In conclusion, our findings provide a novel tumor suppressor gene in BC, which could be served as an independent prognostic marker for stratification of NMIBC and MIBC patients into favorable and poor prognosis. Moreover, a new paradigm in BC tumorigenesis and pathogenesis is estimated, since this novel gene seems to involve in the crucial tumorigenesis signaling pathways.

Supplementary Material

Supporting Data

Acknowledgements

The biospecimens used in the present study were provided by the Chungbuk National University Hospital, a member of the National Biobank of Korea, which is supported by the Ministry of Health, Welfare, and Family Affairs. All samples derived from the National Biobank of Korea were obtained with informed consent under institutional review board-approved protocols. The authors would like to thank Ms. Eun-Ju Shim from the National Biobank of Korea at Chungbuk National University Hospital for preparing samples and her excellent technical assistance.

Funding

The present study was supported by the International Science and Business Belt Program of the Ministry of Science, ICT and Future Planning (grant no. 2015-DD-RD-0070); the National Research Foundation of Korea funded by the Korean government (grant no. 2018R1A2B2005473); and the Basic Science Research Program of the National Research Foundation of Korea, funded by the Ministry of Education (grant no. 2017R1D1A1B03033629).

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

XMP, PJ, SJY and WJK designed the study and all experiments. XMP performed the experiments. YHK, YJB, YX, SPS and SKM collected patient samples. XMP, CY, HWK and WTK assisted with data collection. XMP, JYL, IYK, YHC, EJC and SJY analyzed the data. WJK provided funding. XMP, SJY and WJK wrote the manuscript.

Ethics approval and consent to participate

The collection and analysis of all samples were approved by the Institutional Review Board at Chungbuk National University (approval no. GR2010-12-010). The study methodologies conformed with the standards set by the Declaration of Helsinki. All samples derived from the National Biobank of Korea were obtained with informed consent under institutional review board-approved protocols.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

ARHGAP9

Rho GTPase-activating protein 9

BC

Bladder cancer

CI

confidence interval

CIS

carcinoma in situ

CSS

cancer-specific survival

EGFR

epidermal growth factor receptor

Gli1

glioma-associated oncogene homolog 1

HR

hazard ratio

MAPK1

mitogen-activated protein kinase 1 (also known as ERK2)

MAPK14

mitogen-activated protein kinase 14 (also known as p38α)

MIBC

muscle invasive BC

MKK3

mitogen-activated protein kinase kinase 3

MKK6

mitogen-activated protein kinase kinase 6

NGS

next generation sequencing

NMIBC

non-muscle invasive BC

PFS

progression-free survival

Real-time PCR

real-time polymerase chain reaction

RFS

recurrence-free survival

T1HG

T1 high grade

References

1 

Lotan Y, Black PC, Caba L, Chang SS, Cookson MS, Daneshmand S, Kamat AM, McKiernan JM, Pruthi RS, Ritch CR, et al: Optimal trial design for studying urinary markers in bladder cancer: A collaborative review. Eur Urol Oncol. 1:223–230. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Soukup V, Čapoun O, Cohen D, Hernandez V, Babjuk M, Burger M, Compérat E, Gontero P, Lam T, MacLennan S, et al: Prognostic performance and reproducibility of the 1973 and 2004/2016 World Health Organization grading classification systems in non-muscle-invasive bladder cancer: A European Association of Urology non-muscle invasive bladder cancer guidelines panel systematic review. Eur Urol Suppl. 72:801–813. 2017. View Article : Google Scholar

3 

Westhoff E, Witjes JA, Fleshner NE, Lerner SP, Shariat SF, Steineck G, Kampman E, Kiemeney LA and Vrieling A: Body mass index, diet-related factors, and bladder cancer prognosis: A systematic review and meta-analysis. Bladder Cancer. 4:91–112. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Sethi S, Kong D, Land S, Dyson G, Sakr WA and Sarkar FH: Comprehensive molecular oncogenomic profiling and miRNA analysis of prostate cancer. Am J Transl Res. 5:200–211. 2013.PubMed/NCBI

5 

Lee JY, Yun SJ, Jeong P, Piao XM, Kim YH, Kim J, Subramaniyam S, Byun YJ, Kang HW, Seo SP, et al: Identification of differentially expressed miRNAs and miRNA-targeted genes in bladder cancer. Oncotarget. 9:27656–27666. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Furukawa Y, Kawasoe T, Daigo Y, Nishiwaki T, Ishiguro H, Takahashi M, Kitayama J and Nakamura Y: Isolation of a novel human gene, ARHGAP9, encoding a rho-GTPase activating protein. Biochem Bioph Res Commun. 284:643–649. 2001. View Article : Google Scholar

7 

Hall A: Rho GTPases and the control of cell behaviour. Biochem Soc Trans. 33:891–895. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Etienne-Manneville S and Hall A: Rho GTPases in cell biology. Nature. 420:629–635. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Ang BK, Lim CY, Koh SS, Sivakumar N, Taib S, Lim KB, Ahmed S, Rajagopal G and Ong SH: ArhGAP9, a novel MAP kinase docking protein, inhibits Erk and p38 activation through WW domain binding. J Mol Signal. 2:12007. View Article : Google Scholar : PubMed/NCBI

10 

Sahai E and Marshall CJ: RHO-GTPases and cancer. Nat Rev Cancer. 2:133–142. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Jaffe AB and Hall A: Rho GTPases in transformation and metastasis. Adv Cancer Res. 84:57–80. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Wang T and Ha M: Silencing ARHGAP9 correlates with the risk of breast cancer and inhibits the proliferation, migration, and invasion of breast cancer. J Cell Biochem. 119:7747–7756. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Katoh Y and Katoh M: Integrative genomic analyses on GLI1: Positive regulation of GLI1 by Hedgehog-GLI, TGFβ-Smads, and RTK-PI3K-AKT signals, and negative regulation of GLI1 by Notch-CSL-HES/HEY, and GPCR-Gs-PKA signals. Int J Oncol. 35:187–192. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Sharma S, Ksheersagar P and Sharma P: Diagnosis and treatment of bladder cancer. Am Fam Physician. 80:717–723. 2009.PubMed/NCBI

15 

Hall MC, Chang SS, Dalbagni G, Pruthi RS, Seigne JD, Skinner EC, Wolf JS Jr and Schellhammer PF: Guideline for the management of nonmuscle invasive bladder cancer (stages Ta, T1, and Tis): 2007 update. J Urology. 178:2314–2330. 2007. View Article : Google Scholar

16 

Babjuk M, Böhle A, Burger M, Capoun O, Cohen D, Compérat EM, Hernández V, Kaasinen E, Palou J, Rouprêt M, et al: EAU guidelines on non-muscle-invasive urothelial carcinoma of the bladder: Update 2016. Eur Urol. 71:447–461. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Witjes JA, Compérat E, Cowan NC, De Santis M, Gakis G, Lebret T, Ribal MJ, Van der Heijden AG and Sherif A; European Association of Urology, : EAU guidelines on muscle-invasive and metastatic bladder cancer: Summary of the 2013 guidelines. Eur Urol. 65:778–792. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Kim WT, Kim J, Yan C, Jeong P, Choi SY, Lee OJ, Chae YB, Yun SJ, Lee SC and Kim WJ: S100A9 and EGFR gene signatures predict disease progression in muscle invasive bladder cancer patients after chemotherapy. Ann Oncol. 25:974–979. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Bolstad BM, Irizarry RA, Åstrand M and Speed TP: A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics. 19:185–193. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U and Speed TP: Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 4:249–264. 2003. View Article : Google Scholar : PubMed/NCBI

21 

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

22 

Trapnell C, Roberts A, Goff L, Pertea G, Kim D, Kelley DR, Pimentel H, Salzberg SL, Rinn JL and Pachter L: Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc. 7:562–578. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Sun J, Nishiyama T, Shimizu K and Kadota K: TCC: An R package for comparing tag count data with robust normalization strategies. BMC Bioinformatics. 14:2192013. View Article : Google Scholar : PubMed/NCBI

24 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Pignot G, Vieillefond A, Vacher S, Zerbib M, Debre B, Lidereau R, Amsellem-Ouazana D and Bieche I: Hedgehog pathway activation in human transitional cell carcinoma of the bladder. Br J Cancer. 106:1177–1186. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Lee J, Platt KA, Censullo P and Ruiz i Altaba A: Gli1 is a target of Sonic hedgehog that induces ventral neural tube development. Development. 124:2537–2552. 1997.PubMed/NCBI

27 

Kimura H, Stephen D, Joyner A and Curran T: Gli1 is important for medulloblastoma formation in Ptc1+/- mice. Oncogene. 24:4026–4036. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Kumar B, Sinclair J, Khandrika L, Koul S, Wilson S and Koul HK: Differential effects of MAPKs signaling on the growth of invasive bladder cancer cells. Int J Oncol. 34:1557–1564. 2009.PubMed/NCBI

29 

Spiess PE and Czerniak B: Dual-track pathway of bladder carcinogenesis: Practical implications. Arch Pathol Lab Med. 130:844–852. 2006.PubMed/NCBI

30 

Nicholson RI, Gee JM and Harper ME: EGFR and cancer prognosis. Eur J Cancer. 37 (Suppl 4):S9–S15. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Kim WJ, Kim SK, Jeong P, Yun SJ, Cho IC, Kim IY, Moon SK, Um HD and Choi YH: A four-gene signature predicts disease progression in muscle invasive bladder cancer. Mol Med. 17:478–485. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Mari A, Campi R, Tellini R, Gandaglia G, Albisinni S, Abufaraj M, Hatzichristodoulou G, Montorsi F, van Velthoven R, Carini M, et al: Patterns and predictors of recurrence after open radical cystectomy for bladder cancer: A comprehensive review of the literature. World J Urol. 36:157–170. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Yun SJ, Kim SK and Kim WJ: How do we manage high-grade T1 bladder cancer? Conservative or aggressive therapy? Investig Clin Urol. 57 (Suppl 1):S44–S51. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2020
Volume 19 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Piao XM, Jeong P, Yan C, Kim YH, Byun YJ, Xu Y, Kang HW, Seo SP, Kim WT, Lee JY, Lee JY, et al: A novel tumor suppressing gene, ARHGAP9, is an independent prognostic biomarker for bladder cancer. Oncol Lett 19: 476-486, 2020
APA
Piao, X., Jeong, P., Yan, C., Kim, Y., Byun, Y.J., Xu, Y. ... Kim, W. (2020). A novel tumor suppressing gene, ARHGAP9, is an independent prognostic biomarker for bladder cancer. Oncology Letters, 19, 476-486. https://doi.org/10.3892/ol.2019.11123
MLA
Piao, X., Jeong, P., Yan, C., Kim, Y., Byun, Y. J., Xu, Y., Kang, H. W., Seo, S. P., Kim, W. T., Lee, J., Kim, I. Y., Moon, S., Choi, Y. H., Cha, E., Yun, S. J., Kim, W."A novel tumor suppressing gene, ARHGAP9, is an independent prognostic biomarker for bladder cancer". Oncology Letters 19.1 (2020): 476-486.
Chicago
Piao, X., Jeong, P., Yan, C., Kim, Y., Byun, Y. J., Xu, Y., Kang, H. W., Seo, S. P., Kim, W. T., Lee, J., Kim, I. Y., Moon, S., Choi, Y. H., Cha, E., Yun, S. J., Kim, W."A novel tumor suppressing gene, ARHGAP9, is an independent prognostic biomarker for bladder cancer". Oncology Letters 19, no. 1 (2020): 476-486. https://doi.org/10.3892/ol.2019.11123