Overexpression of the YAP1 oncogene in clear cell renal cell carcinoma is associated with poor outcome

  • Authors:
    • Agnieszka Rybarczyk
    • Jakub Klacz
    • Agata Wronska
    • Marcin Matuszewski
    • Zbigniew Kmiec
    • Piotr M. Wierzbicki
  • View Affiliations

  • Published online on: May 15, 2017     https://doi.org/10.3892/or.2017.5642
  • Pages: 427-439
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Clear cell renal cell carcinoma (ccRCC) is the most common subtype of RCC (70-80%). Yes-associated protein 1 (YAP1) protein is a nuclear effector of the Hippo pathway and acts as a transcriptional co-activator of genes involved in the processes of growth and development of tissues. Hippo signaling, with its key kinases, MST2 and large tumor suppressor kinase 1 (LATS1), plays a significant role in the negative regulation of the amount and activity of YAP1 protein. Components of the Hippo pathway and YAP1 levels are frequently dysregulated in a variety of tumors, suggestive of their possible involvement in carcinogenesis. Our aim was to evaluate gene and protein expression profiles of YAP1, MST2 and LATS1 and the methylation status of MST2 and LATS1 promoters in ccRCC. mRNA levels of MST2, LATS1 and YAP1 genes were assessed in the tumor and matched normal kidney tissues of 86 patients, and in 12 samples of local metastases by quantitative PCR (qPCR). Proteins were semi-quantified in 58 patient samples by western blotting. Hypermethylation of LATS1 and MST2 promoters was measured by methylation‑specific high‑resolution-melting qPCR. We found that LATS1 promoter hypermethylation, decreased LATS1 mRNA/protein and increased YAP1 mRNA/protein levels in tumor samples were associated with higher TNM and Fuhrman's stages and patient survival. Higher YAP1 mRNA levels were associated with poor outcome (HR=4.03, p=0.036). No changes in MST2 (promoter/mRNA/protein) were found. We propose that deregulation of LATS1 and YAP1 expression is associated with ccRCC progression and poor patient survival. Measurement of YAP1 mRNA levels in paired tumor-normal kidney tissue samples may serve as a new prognostic factor in ccRCC.

Introduction

Clear cell renal cell carcinoma (ccRCC) is the most frequent RCC subtype and is characterized by a high mortality rate of 40% within 5 years, due to late diagnosis and distant metastases found in 30 (1) to 80% (2) of RCC patients at the time of examination or within the course of the disease. Among patients who undergo radical resection of the tumor, future metastatic disease develops in 20–40% of the ccRCC cases (3). The search for new molecular targets is continuing due to the high mortality rate of advanced RCC patients (4).

The Hippo pathway is an important regulator of cell proliferation, apoptosis, stem cell functions (5,6) as well as tissue growth and regeneration. Its deregulation is commonly observed in many human cancers, suggesting that alterations of Hippo signaling may be associated with tumor initiation and/or progression (79). The Hippo core cassette is formed by MST2 (serine/threonine kinase 3, STK3) and large tumor suppressor kinase 1 (LATS1) kinases (10). The phosphorylation of LATS1 by MST2 (with SAV1 and MOB1A/B co-activators) inhibits its transcriptional co-activator and downstream effector - Yes-associated protein 1 (YAP1) (11) via its phosphorylation, sequestration to the cytoplasm followed by YAP1 degradation (4,12). When YAP1 is located in the nucleus, it interacts with several transcriptional factors including TEA domain transcription factor 1–4 (TEAD1-4), OCT4, TP73 and ZEB1 (13). Increased expression of the YAP1 protein is associated with tissue regeneration or carcinogenesis (11,14,15). Moreover, the deregulation of the Hippo pathway components and/or YAP1 expression is frequently associated with the progression of various malignancies. Decreased expression of LATS1 gene and protein was observed in breast (16), colorectal (17) and non-small cell lung cancers (18), whereas lower MST2 mRNA and protein levels were reported in hepatocellular carcinoma (19) and malignant mesothelioma (20). Furthermore, the overexpression of YAP1 protein was observed in many cancer types, including lung (21), prostate (22), breast (23), and gallbladder cancers (24) and glioma (25). Since to date no quantitative analyses of the expression of the Hippo pathway effector, YAP1, and its key components, MST2 and LATS1 kinases, have been assessed in ccRCC, we decided to compare their mRNA and protein levels in tumor and normal kidney tissues, and in metastases of ccRCC. We also analyzed the methylation status of LATS1 and MST2 gene promoters by methylation-specific high-resolution-melting quantitative PCR (MS-HRM-qPCR), a novel quantitative technique.

Materials and methods

Patients and samples

Tissue samples were collected from 86 ccRCC patients who underwent radical nephrectomy at the Department of Urology, Medical University of Gdansk, Poland, between January 2011 and September 2013. The clinical data of patients are presented in Table I. The study was approved by the local Ethics Committee; written consent was obtained before surgery from each patient.

Table I.

Clinicopathological features of the ccRCC patients and the association between YAP1, LATS1 and MST2 mRNA levels and clinical data.

Table I.

Clinicopathological features of the ccRCC patients and the association between YAP1, LATS1 and MST2 mRNA levels and clinical data.

YAP1 qPCR results, n (%)LATS1 qPCR results, n (%)MST2 qPCR results n, (%)



Patient characteristics (n=86)Low (≤0.382)High (>0.382)P-value (low vs. high)aLow (≤1.982)High (>1.982)P-value (low vs. high)aLow (≤0.539)High (>0.539)P-value (low vs. high)a
Age (years)
  Mean: 62.16±11.24
  Range: 33–83
  ≤6215 (17)30 (35)0.6439 (45)6 (7)1.0024 (28)21 (24)0.66
  >6211 (13)30 (35) 36 (42)5 (6) 19 (22)22 (26)
Sex
  Female (n=38)10 (12)28 (32)0.6429 (31)9 (10)0.4316 (19)22 (26)0.28
  Male (n=48)16 (19)32 (37) 46 (49)2 (10) 27 (31)21 (24)
Tumor size (cm)
  ≤7 (n=5012 (14)24 (28)0.6439 (45)6 (7)1.0023 (27)13 (15)0.048
  >7 (n=36)14 (16)36 (42) 36 (52)5 (6) 20 (23)30 (35)
Fuhrmans histological grade
  1+2 (n=35)14 (16)21 (24)0.0827 (32)8 (9)0.0417 (20)18 (21)1.00
  3+4 (n=51)12 (14)39 (45) 48 (56)3 (3) 26 (30)25 (29)
TNM stage
  Non-metastatic
    T1-2N0M021 (24)16 (19) <0.000129 (34)8 (9)0.0418 (21)19 (22)1.00
  Metastatic
    T1-2N1M05 (6)44 (51) 46 (53)3 (3) 25 (29)24 (28)
    T3N0-1M0
    T4N0-2M0
    T1-4N2M0
    T1-4N0-2M1

a P-values were calculated by Fishers 2×2 test. ccRCC, clear cell renal cell carcinoma; YAP1, Yes-associated protein 1; LATS1, large tumor suppressor kinase 1; MST2, serine/threonine kinase 3. Bold indicates statistical significance.

Sample acquisition

Samples were obtained according to our previous reports (26,27). In short, dissected tissue samples of primary ccRCC tumors (n=86, named T), normal kidney (n=86, named C as control) and adrenal gland or the whole lymph node (n=12, named M), were collected in the operating theatre (by J.K.) and placed immediately in approximately five volumes of RNAlater (Ambion Inc., Austin, TX, USA).

Assessment of MST2, LATS1 and YAP1 mRNA expression

RNA isolation and cDNA synthesis were performed as previously described (26,27). Briefly, ExtractMe RNA kit (DNAGdansk, Gdansk, Poland) was used for RNA extraction. Two micrograms of total RNA was reverse transcribed with the use of RevertAid Reverse Transcriptase (Fermentas-Thermo Fischer Scientific, Fitchburg, WI, USA). qPCR details are presented in Table II. All reactions were run in duplicate. Based on the results of our previous study on the choice of suitable qPCR reference gene in ccRCC (27), we chose the assessment of GUSB gene expression to normalize the mRNA levels in the samples with the use of Schmittgen and Livaks ΔΔCt equation (28).

Table II.

Details of qPCR assays.

Table II.

Details of qPCR assays.

AssayPrimer sequencesAmplicon size/CpGs in productqPCR efficiencyqPCR reaction conditionsqPCR reaction content
LATS1 5′-AAGTATTTTTGGTGGGGTAGAG96 bp/9 nt93%95°C, 5 min; 42x (95°C, 5 sec; 56°C, 10 sec;5 µl SensiFast HRM
promoter methylation 5′-AAAAAAAACCAAATCCTCAC 72°C, 10 sec; 73 °C, 10 sec - sample reading).(with Eva-Green fluorophone)
MST2 5′-TTTGGAAAAAGATAAGGTTTTTAT118 bp/7 nt91%Melting curve: 95°C, 15 sec; 60°C, 1 min;(BioLine, London, UK),
promoter methylation 5′-CCTAAAATTAACTCTCAACCTCTC 60°C → 95°C reading every 0.1°C300 nM each primer, ∑ 10 µl
LATS1 mRNA level 5′-TGCACTGGCTTCAGATGGACAC177 bp93.4%95°C, 3 min; 37x (95°C, 5 sec; 58°C, 10 sec;5 µl SensiFast NoRox
measurment 5′-ATGTGCTAGACATCGCTGGTGC 72°C, 10 sec; 75 °C, 10 sec - sample reading).SYBR-Green
MST2 mRNA level 5′-CTATAACTGTGTGGCCGACATCTGG223 bp90.3%Melting curve: 95°C, 15 sec; 60°C, 1 min;
measurment 5′-TTGTGTTGCAGTAGCTCTCTGCTC 60°C → 95°C reading every 0.3°C(BioLine, London, UK),
YAP1 mRNA level 5′-TCAGACAACAACATGGCAGGACC235 bp101.3% 200 nM each primer, Σ 10 µl
measurment 5′-TCTCTGGTTCATGGCAAAACGAGG
GUSB mRNA level 5′-ATGCAGGTGATGGAAGAAGTGGTG177 bp99.6%95°C, 3 min; 35x (95°C, 5 sec; 57°C, 10 sec;
measurment for qPCR normalization 5′-AGAGTTGCTCACAAAGGTCACAGG 72°C, 10 sec; 75 °C, 10 sec - sample reading) Melting curve: 95°C, 15 sec; 60°C, 1 min; 60°C → 95°C reading every 0.3°C. Melting curve: 95°C, 15 sec; 60°C, 1 min; 60°C → 95°C reading every 0.3°
DNA extraction, bisulfite modification, acquisition of control DNA and MS-HRM-qPCR

The methodology has been previously described (26). In short, DNA was isolated to a total volume of 20 µl followed by bisulfide modification (DNA Methylation-Direct™ kit; Zymo Research, Irvine, CA, USA). For the generation of a dilution series of control DNA standards, fully methylated (named MD) and unmethylated (UMD) human genomic DNAs (Zymo) were used.

Methylation was assessed in the samples with the use of the MS-HRM-qPCR method (29). Reactions were set on the Step-One Plus apparatus, then post-PCR products were analyzed with the use of HRM software ver. 3.1 (both from Life Technologies, Grand Island, NY, USA). For each run, matched DNA from T, C and M samples were set; standard dilutions of control MD and UMD were made to 100, 50, 25, 10 and 0 of MD in UMD and used in the same PCR plate as well as the no template control.

Western blot analysis

Protein lysates were prepared with Mammalian Cell Extraction kit (BioVision, Milpitas, CA, USA). The lysates (10 µg) were loaded onto a 10% Mini-Protean TGX gel (Bio-Rad, Hercules, CA, USA), resolved by SDS-PAGE, and transferred to a PVDF membrane using the Trans-Blot Turbo system (Bio-Rad). Membranes were stained with 0.1% Ponceau S to ensure equal loading after transfer, and subsequently blocked with 5% albumin fraction V in TBS buffer with 0.1% Tween-20 (TBST) for 1 h at room temperature (RT). After washing with TBST, the membranes were incubated (overnight, 4°C) with specific primary antibodies in 2% albumin/TBS: rabbit anti-LATS1 (1:2,000, Bioss, Woburn, MA, USA), rabbit polyclonal anti-YAP1 (1:1,000), and rabbit monoclonal anti-MST2(STK3) (1:2,000) (both from Abcam, Cambridge, UK) and anti-GAPDH peroxidase-conjugated IgM (1:50,000; Sigma-Aldrich, St. Louis, MO, USA). After triple washing with TBST, the blots were incubated for 2 h at RT with horseradish peroxidase-conjugated secondary antibodies: anti-rabbit IgG or anti-mouse IgG (1:15,000; Sigma-Aldrich). Following triple washing with TBST, immunoreactive bands were detected on medical X-ray film (Agfa HealthCare, Mortsel, Belgium) using chemiluminescent peroxidase substrate (Sigma-Aldrich). Densitometric analysis of immunoreactive protein bands was performed with Quantity One software (Bio-Rad) and calculated as units = intensity/mm2. After normalization to GAPDH protein units for each sample, the semi-quantitative results for either tumor or metastasized samples were obtained as a ratio: mean unitsT/M/mean unitsC for MST2, LATS1 or YAP1 proteins.

Statistical analysis

Statistical analysis was performed with the use of the GraphPad Prism ver. 6.05 software (GraphPad Software, San Diego, CA, USA). The following statistical tests were used: non-parametric Mann-Whitney U, Kruskal-Wallis ANOVA, Fisher's 2×2 exact test, multivariate regression, and Cox-Mantel proportional hazard regression model. Survival relationships were presented as hazard ratios (HR) with their 95 confidence interval (CI) and p-values (30) using Cox and Kaplan-Meier estimations. Rates of overall survival (OS) and progression-free survival (PFS) were calculated separately. In all analyses, a two-sided p<0.05 was considered as statistically significant with a 95% CI.

Results

Clinicopathological characteristics of the patients

Of the 86 ccRCC patients (62.1±11.2 years, mean age ± SD) (Table I), 37 were diagnosed as stage I (T1-2N0M0), 8 as stage II (T2N0M0), 12 as stage III (T1-2N1M0 or T3N0-1M0) and 29 as stage IV (T4N0-2M0 or T1-4N2M0 or T1-4N0-2M1). TNM stages of the kidney cancer are as follows: stage I, tumor ≤7 cm and limited to the kidney; stage II, tumor 7–10 cm, limited to the kidney; stage III, tumor extends into major veins or perinephric tissues but not into the ipsilateral adrenal gland and not beyond Gerota fascia or T1-T3 with metastasis in a single regional lymph node; stage IV, metastasis in more than one regional lymph node or distant metastasis (31). At the time of surgery, 47.7% of the ccRCC patients were diagnosed with local or distant metastases. Histological nuclear staging in renal cancer is based on the Fuhrman grading; grade 1: small, round, uniform nuclei (10 microns), inconspicuous nucleoli; grade 2: slightly irregular nuclei, nuclear diameter 15 microns, open chromatin; grade 3: visible nucleoli, nuclei very irregular, diameter 20 microns, open chromatin (32). According to Fuhrman's division 4 patients were grade 1, 32 grade 2, 23 grade 3 and 26 were grade 4. None of the patients underwent chemotherapy or radiotherapy before surgery. The mean follow-up period was 21 months (range, 3–48). To date, 45 patients were alive (52%); all deaths (except for one patient) were related to ccRCC progression. Median OS rate was 12 months. During follow-up, metastases occurred in 38 (44%) patients while the median PFS rate was 6 months.

Expression of the YAP1, LATS1 and MST2 genes at the mRNA level

As shown in Fig. 1A, YAP1 mRNA levels in T (tumor) and M (metastatic) samples were ~5 and 4 times higher when compared to the C (control tissue) samples, respectively (p<0.01). When the samples were divided according to median mRNA values in the C samples, we found that 60 (70%) out of 86 tumor samples contained an increased YAP1 mRNA level (Table I). The mRNA levels of LATS1 were ~3 and 25 times lower in the T and M samples (p<0.05) (Fig. 1D) and lower LATS1 mRNA content was observed in 75/86 (87%) tumor samples (Table I). On the contrary, the expression of MST2 at the mRNA level was decreased only in M samples, showing a statistically not significantly increased ratio in 43/86 (50%) T samples as compared to the level in the control tissue (Fig. 1G and Table I). The comparison of the clinicopathological data with mRNA levels revealed that poorly developed ccRCC tumors (Fuhrman's grades 3 and 4) were characterized by decreased LATS1 and increased YAP1 mRNA levels (Table I and Fig. 1B and E) as compared to the control samples. In addition, we found either lower LATS1 mRNA level or higher YAP1 mRNA ratio in tumor ccRCC cases which were diagnosed with local (N1-2) or distant metastasis (M1) as shown in Table I and Fig. 1C and F. No statistically significant relationships between MST2 mRNA ratios and clinical data were observed except for the higher content of MST2 transcript in samples obtained from larger tumors (Table I).

Expression of YAP1, LATS1 and MST2 proteins

The semi-quantification of the studied proteins normalized to GAPDH protein was performed in paired samples of 58 ccRCC cases as well as in 8 M cases. As presented in Fig. 2A and Table III, the YAP1 protein level was ~2 times higher in 47 (81%) of the 58 analyzed T samples, whereas LATS1 protein ratio was ~4 times lower in 42 (72%) T samples when compared to the C samples (Fig. 2D). The relationship between clinicopathological data and LATS1 and YAP1 protein expression was noted. Poorly developed (high Fuhrman's grades) T cases were characterized by increased YAP1 and decreased LATS1 protein ratios (Table III and Fig. 2B and E). The difference in protein expression of either LATS1 or YAP1 between non-metastatic vs. metastatic tumor ccRCC cases was observed (Table III and Fig. 2C and F). Semi-quantification of MST2 protein did not show any differences either between tumor and normal kidney samples or between cancer samples classified according to clinicopathological status (Table III and Fig. 2G H and I).

Table III.

Comparison between YAP1, LATS1 and MST2 protein levels, LATS1 and MST2 methylation status and clinical data of the 58 ccRCC patients.

Table III.

Comparison between YAP1, LATS1 and MST2 protein levels, LATS1 and MST2 methylation status and clinical data of the 58 ccRCC patients.

YAP1 protein assessment (AU) (%)LATS1 methylationLATS1 protein assessment (AU) (%)MST2 methylationMST2 protein assessment (AU) (%)






Patients (n=86)Low (≤17.636)High (>17.636)P-value (low vs. high)a<25%25–100%P-value (low vs. high)aLow (≤12.667)High (>12.669)P-value (low vs. high)a<25%25–100%P-value (low vs. high)aLow (≤19.31)High (>19.31)P-value (low vs. high)a
Age (years)
  Median: 62.5
  Mean: 62.82±11.94
  Range: 33–83
  ≤627 (12)22 (38)0.5014 (24)15 (25)0.7919 (33)10 (17)0.3819 (33)10 (17)0.5919 (33)10 (17)0.78
  >624 (7)25 (43) 16 (28)13 (22) 23 (40)6 (10) 16 (28)13 (22) 17 (29)12 (21)
Sex
  Female (n=28)4 (7)24 (41)0.5118 (31)10 (16)0.0721 (36)7 (12)0.7714 (24)14 (24)0.1814 (24)14 (24)0.06
  Male (n=30)7 (12)23 (40) 12 (21)18 (31) 21 (36)9 (16) 21 (36)9 (16) 23 (40)7 (12)
Tumor size (cm)
  ≤7 (n=24)5 (9)19 (33)0.7512 (21)12 (21)1.0018 (31)6 (10)0.4714 (24)10 (17)1.0013 (22)11 (19)0.41
  >7 (n=34)6 (10)28 (48) 18 (31)16 (27) 24 (41)10 (17) 21 (36)13 (22) 23 (40)11 (19)
Fuhrmans histological grade
  1+2 (n=25)8 (14)17 (29)0.0421 (36)4 (7) <0.000115 (26)10 (17)0.0313 (22)12 (21)0.2916 (28)9 (16)1.00
  3+4 (n=33)3 (5)30 (52) 9 (15)24 (41) 28 (48)5 (9) 22 (38)11 (19) 20 (34)13 (22)
TNM stage
  Non-metastatic8 (11)16 (22)0.0222 (38)7 (12)0.000518 (31)11 (19)0.1417 (29)12 (21)1.0019 (33)10 (17)0.78
    T1-2N0M0
  Metastatic
    T1-2N1M05 (7)44 (60) 8 (14)21 (36) 24 (41)5 (9) 18 (31)11 (19) 17 (29)12 (21)
    T3N0-1M0
    T4N0-2M0
    T1-4N2M0
    T1-4N0-2M1

a P-values were calculated by Fishers 2×2 test. ccRCC, clear cell renal cell carcinoma; LATS1, large tumor suppressor kinase 1; YAP1, Yes-associated protein 1. Bold indicates statistical significance.

LATS1 and MST2 promoter methylation status

Methylation analysis was carried out in 58 tumor and 10 control samples. According to the analysis of MD/UMD standards, the results of MS-HRM-qPCR were qualified into four grades: 1, 0–10% methylation; 2, 10–25%; 3, 25–50%; 4, 50–100%. Based on the results of 10 control samples and our previous results (26), we set the value >25% methylation as the hypermethylation status for either LATS1 or MST2 promoters. We found that LATS1 or MST2 hypermethylation was observed in 28 (48%) or 22 (38%) of 58 tumor ccRCC samples, respectively (Table III). The hypermethylation of LATS1 promoter was associated with higher Fuhrman's grades (3 and 4 vs. 1 and 2) as well as the presence of local and/or distant metastasis (Table III). Since the same T samples were analyzed for either LATS1 or MST2 methylation and mRNA and protein content, we ascertained whether the hypermethylation of the gene promoter region was associated with its mRNA/protein content. As shown in Fig. 3, the mRNA and protein expression of LATS1 gene was related to the hypermethylation status of this gene; such observation was not proven for the MST2 gene.

Relationships between LATS1, MST2 and YAP1 proteins

We checked possible correlations between mRNA-mRNA, mRNA-protein, protein-mRNA and protein-protein levels of LATS1-YAP1, LATS1-MST2 and MST2-YAP1. We found a negative correlation between LATS1 protein and YAP1 protein levels when all paired samples of 58 patients were taken into consideration (rs=−0.51; p<0.05, Spearman's test; Fig. 4).

Association between molecular findings and clinicopathological parameters and patient outcome

As presented in Figs. 5A and B and 6A and B, OS as well as PFS were strongly associated with a higher TNM and Fuhrman's grading in the patients. The molecular data revealed that increased YAP1 expression levels either at the mRNA or protein levels as well as the hypermethylation of LATS1 promoter were related to both PFS and OS (Figs. 5C-E and 6C-E). The increased level of MST2 mRNA was associated with shorter OS (Fig. 5F).

Cox proportional hazard model with multivariate analyses revealed that the YAP1 mRNA level was an independent predictor of OS in ccRCC patients when assessed by Fuhrman's histological grade (Table IV). There was no association between molecular data and hazard ratio when the PFS rate was checked (Table V).

Table IV.

Univariable and multivariable Cox regression analysis of the overall survival rate of the ccRCC patients.

Table IV.

Univariable and multivariable Cox regression analysis of the overall survival rate of the ccRCC patients.

Univariable analysisMultivariable analysis


ParametersP-valueHR (95% CI)P-valueHR (95% CI)
Sex
  Female vs. Male0.0642.28 (0.95–5.46)
Age (years)
  >62 vs. ≤620.560.78 (0.34–1.786)
Tumor size (cm)
  >7 vs. ≤70.290.64 (0.285–1.47)
Tumor grade
  T3+4 vs. T1+20.0014.72 (1.85–12.06)0.910.93 (0.28–3.15)
Histological grade
  F3+4 vs. F1+20.00058.61 (2.54–29.18)0.0196.04 (1.34–27.26)
LATS1 mRNA levels
  ↓ (≤1.982) vs. ↑ (>1.982)0.581.35 (0.46–3.99)
LATS1 methylation
  ↑ (>25%) vs. ↓ (≤25%)0.013.06 (1.29–7.29)0.521.35 (0.52–3.51)
LATS1 protein levels
  ↓ (≤12.669) vs. ↑ (>12.669)0.330.58 (0.19–1.72)
YAP1 mRNA levels
  ↑ (>0.328) vs. ↓ (≤0.328)0.014.87 (1.43- 16.52)0.0364.03 (0.96–16.79)
YAP1 protein levels
  ↑ (>17.363) vs. ↓ (≤17.363)0.0476.11 (0.82–45.48)0.671.60 (0.17–14.43)
MST2 mRNA levels
  ↓ (≤0.539) vs. ↑ (>0.539)0.721.16 (0.49–2.71)
MST2 methylation
  ↑ (>25%) vs. ↓ (≤25%)0.520.75 (0.32–1.78)
MST2 protein levels
  ↓ (≤10.09) vs. ↑ (>10.09)0.820.91 (0.39–2.11)

[i] ccRCC, clear cell renal cell carcinoma; HR, hazard ratio; LATS1, large tumor suppressor kinase 1; YAP1, Yes-associated protein 1; CI, confidence interval. Bold indicates statistical significance.

Table V.

Univariable and multivariable Cox regression analysis of progression-free survival rate of ccRCC patients.

Table V.

Univariable and multivariable Cox regression analysis of progression-free survival rate of ccRCC patients.

Univariable analysisMultivariable analysis


ParametersP-valueHR (95% CI)P-valueHR (95% CI)
Sex
  Female vs. Male0.113.12 (1.29–7.59)
Age (years)
  >62 vs. ≤620.440.78 (0.34–1.786)
Tumor size (cm)
  >7 vs. ≤70.430.72 (0.32–1.62)
Tumor grade
  T3+4 vs. T1+20.0023.84 (1.58–9.31)0.770.84 (0.26–2.66)
Histological grade
  F3+4 vs. F1+20.000215.01 (3.51–64.19)0.00113.68 (2.73–68.34)
LATS1 mRNA levels
  ↓(≤ 1.982) vs. ↑ (>1.982)0.540.61 (0.12–2.95)
LATS1 methylation
  ↑(> 25%) vs. ↓ (≤25%)0.651.27 (0.43–3.77)
LATS1 protein levels
  ↓(≤ 12.669) vs. ↑ (>12.669)0.490.70 (0.26–1.89)
YAP1 mRNA levels
  ↑ (> 0.328) vs. ↓ (≤0.328)0.0082.39 (0.88- 6.45)0.091.84 (0.45–6.23)
YAP1 protein levels
  ↑ (>17.363)vs. ↓ (≤17.363)0.0076.21 (0.83–46.07)0.121.72 (0.19–14.91)
MST2 mRNA levels
  ↓ (≤ 0.539) vs. ↑ (>0.539)0.611.23 (0.54–2.79)
MST2 methylation
  ↑ (> 25%) vs. ↓ (≤25%)0.180.55 (0.22–1.32)
MST2 protein levels
  ↓ (≤ 10.09) vs. ↑ (>10.09)0.520.76 (0.34–1.72)

[i] ccRCC, clear-cell renal cell carcinoma; HR, hazard ratio; LATS1, large tumor suppressor kinase 1; YAP1, Yes-associated protein 1; CI, confidence interval. Bold indicates statistical significance.

Discussion

The Hippo pathway is an important regulator of cell proliferation, tissue homeostasis, organ size and stem cell functions (6). Its deregulation is frequently observed in many types of malignancies, suggesting that alterations of this signaling are connected with cancer progression and patient survival (79,21,33,34). The core components of this pathway include MST1/2, SAV1, LATS1/2 and MOB1 proteins (10,12,15). When the Hippo signaling is active, LATS1/2 kinases phosphorylate two major downstream effectors, YAP1 or its paralog, TAZ, resulting in their ubiquitination and proteolytic degradation (35,36). In contrast, deregulation of the pathway components, the consequent Hippo silencing, increases the YAP1 protein level in the cell as well as augments the nuclear localization of YAP1 (37). In turn, YAP1 nuclear accumulation triggers the upregulation of target genes (e.g., CTFG and CYR61), which are associated with processes such as cell migration, proliferation and angiogenesis (37).

The recent results of in vitro studies show that the inhibition of LATS1 kinase is strongly connected with the upregulation of YAP1 resulting in the increased metastatic potential of cancer cells (35,38). Mei et al showed that direct interaction between small ubiquitin-like modifier (SUMO) and LATS1 protein in L02 (normal human hepatic) and HepG2 (hepatocellular carcinoma) cells resulted in the attenuation of LATS1 kinase activity and inhibition of the Hippo pathway. As a consequence, the levels of YAP1, CTFG and CYR61 proteins were increased in SUMOtylated-LATS1 cells (35). Our results based on clinical samples of ccRCC showed a direct association between the presence of LATS1 and YAP1 in kidney tissues; a decreased LATS1 protein level was correlated with increased ratio of YAP1 protein in both tumor and matched normal kidney tissue samples. Another recent study on LATS1-YAP1 interaction in cancer (38) was performed in MDA-MB-231 and MCF7 breast cancer cell lines. Nokin et al found that methylglyoxal, a glycolysis side-product, indirectly targets inactivation of LATS1 in cells. As a result, increased levels of YAP1 protein and its co-effectors were observed which corresponded with the increased metastatic potential of cancer cells in a mouse xenograft model (38). The results of our study indicate that the decreased expression of LATS1 and increased YAP1 either at the mRNA or protein levels are highly associated with renal cancer progression.

Our data corroborate the findings of Chen et al (39) in an RCC cell line (786-O) as well as in tissue samples. In paired tumor and normal kidney samples of 30 ccRCC patients they observed decreased LATS1 mRNA and protein levels in tumor samples; ccRCC progression was associated with lower LATS1 content (39). Our data obtained on a much larger group of ccRCC patients extend these observations suggestive of the roles of LATS1 and YAP1 in ccRCC development since we found that the patients with deregulated LATS1 or YAP1 mRNA and protein levels share poorer clinical outcome. Thus, our and Chen et al (39) findings suggest that measurements of YAP1 mRNA content in ccRCC tumor samples could serve as a potential survival marker together with high Fuhrman's grades. In contrast to a previous study (37) we used quantitative techniques (qPCR vs. RT-PCR and MS-HRM-qPCR vs. bisulfide sequencing PCR) to assess a much larger group of ccRCC patients (86 vs. 30). Although we did not focus on the expression of Hippo pathway components in renal cancer cell lines, Chen et al showed in 786-O and HEK293 kidney cell lines that the decreased expression of LATS1 was associated with promoter hypermethylation (39) which was found by us in ccRCC clinical samples. Moreover, we observed that LATS1 hypermethylation in tumor samples was characteristic of ccRCC patients with earlier occurrence of either metastasis or death. Chen et al also found that the controlled decrease in LATS1 protein level resulted in an increased YAP1 protein level. Furthermore, they observed that overexpression of LATS1 downregulated the YAP1 protein level, inhibited cell proliferation, induced cell apoptosis and cell cycle arrest in 786-O cells (39). LATS1 downregulation and its contribution to cancer progression has been observed in other malignances such as glioma (40), nosopharyngeal carcinoma (41), astrocytoma (42), non-small cell lung cancer (18), breast cancer (16), colorectal cancer (17) and renal carcinoma (39). Additionally, association between LATS1 hypermethylation and tumor progression has been noted in lung cancer (43), schwannomas (44), oral squamous cell carcinoma (45), colorectal cancer (17) and astrocytoma (42), however, the authors did not observe the influence of LATS1 methylation status on patient outcome. Therefore, we believe that our observations may promote studies of LATS1 gene/protein expression to assess the impact on ccRCC progression and prognosis.

Our results suggest that the second core part of Hippo signaling, MST2 protein, is neither involved in ccRCC progression nor in YAP1 regulation. Although MST1/2 kinases have been acknowledged as tumor-suppressor proteins since loss of function of MST1/2 was observed in prostate (46) and breast cancer (47), and a decreased MST1 mRNA level was associated with node metastasis in colorectal cancer (48), however, in hepatocellular carcinoma HepG2 cells increased MST1/2 levels were reported (49). Decreased MST1 expression was associated with promoter methylation of this gene in soft tissue sarcomas (50). Since we did not find an association between MST1 promoter methylation and gene expression, we suggest that such a regulation of MST1 gene expression does not occur in ccRCC. MST1 protein is the upstream regulator of YAP1 protein (10,12,13,15,48,51,52), therefore the lack of an MST1/YAP1 association as observed by us in ccRCC should be discussed. The relationship between MST1/2 protein and YAP1 level in intestinal epithelium was observed by Zhou et al during an in vivo study (53). Their study using an Mst1/2-deficient mouse model showed that MST1 and MST2 proteins are crucial in the regulation of the Yap1 protein level in normal colonic epithelium (53). On the contrary, they found that the antiproliferative role of MST1 or MST2 was overcome in colon cancer by the abundance of Yap1 protein (53). Such an observation is in line with our results, since we did not find alterations in the expression of MST1 mRNA or protein levels in the studied samples of ccRCC. Another in vivo study using mouse models showed different results in regards to the Mst2/Yap1 association in cancer development (19). Zhou et al found that tumorigenesis of hepatocellular carcinoma was associated with loss of Mst2 and a decreased level of phosphorylated Yap1 protein (19). Such an observation could be contrary to our results, however, they observed that the deregulation of Mst1/2 protein did not change the level of Lats1/2 proteins. Based on that, we suppose that such independent regulation of MST2 and LATS1 may occur in ccRCC. However, such a conclusion should be supported by further studies. Moreover, since our study is the first to use complex MST2 quantification in ccRCC, we propose that lack of contribution of this gene in renal cancer progression must be confirmed by independent studies.

The most significant observation revealed in our study was, in our opinion, the possibility of YAP1 mRNA measurement as a potential prognostic factor in ccRCC. Our previous study showed a similar correlation between Hippo upstream regulator, RASSF1A gene, and patient outcome (26). Therefore, in this study we aimed to assess the possible role of YAP1 in ccRCC. Although our report is not the first study of YAP1 expression in ccRCC since Cao et al published a similar study in 2014 (54), there are some significant differences: a larger group of patients (86 vs. 30 persons), study on metastasized samples, modern quantitative techniques (qPCR vs. classical PCR) and survival data. Despite the mentioned differences, Cao et al obtained comparable results since the increased YAP1 protein level was associated with higher Fuhrman's and clinical stages (54). They also performed in vitro studies on 786-O and HEK293 kidney cells and found that knockdown of YAP1 inhibited expression of the TEAD1 gene as well as suppressed cell proliferation (54). Most studies on the role of YAP1 in other cancer types such as RCC (39), oral squamous cell carcinoma (55), ovarian cancer (56), head and neck cancer (57), colorectal cancer (58), melanoma (59), lung (18) and breast cancer (23), revealed an association between YAP1 overexpression (mostly at the protein level) and tumor progression. Furthermore, we found that increased YAP1 levels of either mRNA or protein in tumor samples were associated with poorer patient outcome (survival and occurrence of metastasis). Other authors found a similar correlation between higher YAP1 levels and patient outcome in esophageal cancer (60), gastric adenocarcinoma (61) and papillary thyroid cancer (62).

Another important aspect is the mechanism of YAP1 mRNA regulation. Notably, we observed that only ccRCC patients with increased YAP1 mRNA levels in tumor samples were characterized by a higher risk of death (Cox test). Recent data indicate that some microRNA molecules directly regulate the YAP1 mRNA level. Pan et al found that miR-509-3p targeted YAP1 mRNA in a large group (293 cases from TCGA cohort) of ovarian cancer (63). Moreover, miR-138 was found to be a strong suppressor of YAP1 mRNA in oral squamous cell carcinoma (64) and in non-small cell lung cancer (65). The reported associations between decreased levels of either miR-509-3p or miR-138 in the studied types of cancer and poorer patient outcome (6365), consolidating the influence of YAP1 in tumor progression. In fact, the contribution of YAP1 protein in tumor progression is so important that it was acknowledged as a pivotal molecular target in modern cancer treatment (5,34,38,51,52,66,67). Some authors found an association between YAP1 overexpression and chemoresistance of cancer cells, e.g., in head and neck cancer cases resistant to cetuximab (57), resistance to RAF- and MEK-targeted therapy (33), 5-FU chemotherapy-resistant colon cancer (68) and osteosarcoma resistance (69).

In conclusion, we suggest that dysregulation of LATS1 and YAP1 levels, but not MST2, is associated with ccRCC progression and patient survival. We propose that the assessment of YAP1 mRNA levels in paired tumor-normal kidney tissue samples could serve as a new prognostic factor in ccRCC.

Acknowledgements

The authors wish to thank Dr Marcin Stanislawowski and Dr Tomasz Slebioda from the Department of Histology for the laboratory support. The study was supported by National Science Centre (Poland) grant 2012/05/B/NZ4/02735 and ST-12 internal funds of the Medical University of Gdansk, Poland.

References

1 

Novara G, Martignoni G, Artibani W and Ficarra V: Grading systems in renal cell carcinoma. J Urol. 177:430–436. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Massari F, Bria E, Maines F, Milella M, Giannarelli D, Cognetti F, Pappagallo G, Tortora G and Porta C: Adjuvant treatment for resected renal cell carcinoma: Are all strategies equally negative? Potential implications for trial design with targeted agents. Clin Genitourin Cancer. 11:471–476. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Keizman D, Rouvinov K, Sella A, Gottfried M, Maimon N, Kim JJ, Eisenberger MA, Sinibaldi V, Peer A, Carducci MA, et al: Is there a ‘Trial Effect’ on outcome of patients with metastatic renal cell carcinoma treated with sunitinib? Cancer Res Treat. 48:281–287. 2016. View Article : Google Scholar : PubMed/NCBI

4 

van der Mijn JC, Mier JW, Broxterman HJ and Verheul HM: Predictive biomarkers in renal cell cancer: Insights in drug resistance mechanisms. Drug Resist Updat. 17:77–88. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Zhao B, Lei QY and Guan KL: The Hippo-YAP pathway: New connections between regulation of organ size and cancer. Curr Opin Cell Biol. 20:638–646. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Zhao B, Li L, Lei Q and Guan KL: The Hippo-YAP pathway in organ size control and tumorigenesis: An updated version. Genes Dev. 24:862–874. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Liu H, Jiang D, Chi F and Zhao B: The Hippo pathway regulates stem cell proliferation, self-renewal, and differentiation. Protein Cell. 3:291–304. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Harvey KF, Zhang X and Thomas DM: The Hippo pathway and human cancer. Nat Rev Cancer. 13:246–257. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Zhao B, Tumaneng K and Guan KL: The Hippo pathway in organ size control, tissue regeneration and stem cell self-renewal. Nat Cell Biol. 13:877–883. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Halder G and Johnson RL: Hippo signaling: Growth control and beyond. Development. 138:9–22. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Moya IM and Halder G: The Hippo pathway in cellular reprogramming and regeneration of different organs. Curr Opin Cell Biol. 43:62–68. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Wierzbicki PM and Rybarczyk A: The Hippo pathway in colorectal cancer. Folia Histochem Cytobiol. 53:105–119. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Janse van Rensburg HJ and Yang X: The roles of the Hippo pathway in cancer metastasis. Cell Signal. 28:1761–1772. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Johnson R and Halder G: The two faces of Hippo: Targeting the Hippo pathway for regenerative medicine and cancer treatment. Nat Rev Drug Discov. 13:63–79. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Cai J, Zhang N, Zheng Y, De Wilde RF, Maitra A and Pan D: The Hippo signaling pathway restricts the oncogenic potential of an intestinal regeneration program. Genes Dev. 24:2383–2388. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Morinaga N, Shitara Y, Yanagita Y, Koida T, Kimura M, Asao T, Kimijima I, Takenoshita S, Hirota T, Saya H, et al: Molecular analysis of the h-warts/LATS1 gene in human breast cancer. Int J Oncol. 17:1125–1129. 2000.PubMed/NCBI

17 

Wierzbicki PM, Adrych K, Kartanowicz D, Stanislawowski M, Kowalczyk A, Godlewski J, Skwierz-Bogdanska I, Celinski K, Gach T, Kulig J, et al: Underexpression of LATS1 TSG in colorectal cancer is associated with promoter hypermethylation. World J Gastroenterol. 19:4363–4373. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Lin XY, Zhang XP, Wu JH, Qiu XS and Wang EH: Expression of LATS1 contributes to good prognosis and can negatively regulate YAP oncoprotein in non-small-cell lung cancer. Tumour Biol. 35:6435–6443. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Zhou D, Conrad C, Xia F, Park JS, Payer B, Yin Y, Lauwers GY, Thasler W, Lee JT, Avruch J, et al: Mst1 and Mst2 maintain hepatocyte quiescence and suppress hepatocellular carcinoma development through inactivation of the Yap1 oncogene. Cancer Cell. 16:425–438. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Miyanaga A, Masuda M, Tsuta K, Kawasaki K, Nakamura Y, Sakuma T, Asamura H, Gemma A and Yamada T: Hippo pathway gene mutations in malignant mesothelioma: Revealed by RNA and targeted exon sequencing. J Thorac Oncol. 10:844–851. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Bora-Singhal N, Nguyen J, Schaal C, Perumal D, Singh S, Coppola D and Chellappan S: YAP1 regulates OCT4 activity and SOX2 expression to facilitate self-renewal and vascular mimicry of stem-like cells. Stem Cells. 33:1705–1718. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Machado-Neto JA, Lazarini M, Favaro P, Franchi GC Jr, Nowill AE, Saad ST and Traina F: ANKHD1, a novel component of the Hippo signaling pathway, promotes YAP1 activation and cell cycle progression in prostate cancer cells. Exp Cell Res. 324:137–145. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Vlug EJ, van de Ven RA, Vermeulen JF, Bult P, van Diest PJ and Derksen PW: Nuclear localization of the transcriptional coactivator YAP is associated with invasive lobular breast cancer. Cell Oncol (Dordr). 36:375–384. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Li M, Lu J, Zhang F, Li H, Zhang B, Wu X, Tan Z, Zhang L, Gao G, Mu J, et al: Yes-associated protein 1 (YAP1) promotes human gallbladder tumor growth via activation of the AXL/MAPK pathway. Cancer Lett. 355:201–209. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Liu YC and Wang YZ: Role of Yes-associated protein 1 in gliomas: Pathologic and therapeutic aspects. Tumour Biol. 36:2223–2227. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Klacz J, Wierzbicki PM, Wronska A, Rybarczyk A, Stanislawowski M, Slebioda T, Olejniczak A, Matuszewski M and Kmiec Z: Decreased expression of RASSF1A tumor suppressor gene is associated with worse prognosis in clear cell renal cell carcinoma. Int J Oncol. 48:55–66. 2016.PubMed/NCBI

27 

Wierzbicki PM, Klacz J, Rybarczyk A, Slebioda T, Stanislawowski M, Wronska A, Kowalczyk A, Matuszewski M and Kmiec Z: Identification of a suitable qPCR reference gene in metastatic clear cell renal cell carcinoma. Tumour Biol. 35:12473–12487. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Schmittgen TD and Livak KJ: Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 3:1101–1108. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Wojdacz TK and Dobrovic A: Methylation-sensitive high resolution melting (MS-HRM): A new approach for sensitive and high-throughput assessment of methylation. Nucleic Acids Res. 35:e412007. View Article : Google Scholar : PubMed/NCBI

30 

Avădănei ER, Wierzbicki PM, Giuşcă SE, Grigoraş A, Amălinei C and Căruntu ID: Macrophage profile in primary versus secondary liver tumors. Folia Histochem Cytobiol. 52:112–123. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Sobin LH, Gospodarowicz MK and Wittekind C: International Union against Cancer: TNM classification of malignant tumours. Wiley-Blackwell; Chichester, West Sussex, UK; Hoboken, NJ: 2010

32 

Delahunt B, Sika-Paotonu D, Bethwaite PB, Jordan T William, Magi-Galluzzi C, Zhou M, Samaratunga H and Srigley JR: Grading of clear cell renal cell carcinoma should be based on nucleolar prominence. Am J Surg Pathol. 35:1134–1139. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Lin L, Sabnis AJ, Chan E, Olivas V, Cade L, Pazarentzos E, Asthana S, Neel D, Yan JJ, Lu X, et al: The Hippo effector YAP promotes resistance to RAF- and MEK-targeted cancer therapies. Nat Genet. 47:250–256. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Zhang L, Yang S, Chen X, Stauffer S, Yu F, Lele SM, Fu K, Datta K, Palermo N, Chen Y, et al: The hippo pathway effector YAP regulates motility, invasion, and castration-resistant growth of prostate cancer cells. Mol Cell Biol. 35:1350–1362. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Mei L, Yuan L, Shi W, Fan S, Tang C, Fan X, Yang W, Qian Y, Hussain M and Wu X: SUMOylation of large tumor suppressor 1 at Lys751 attenuates its kinase activity and tumor-suppressor functions. Cancer Lett. 386:1–11. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Saucedo LJ and Edgar BA: Filling out the Hippo pathway. Nat Rev Mol Cell Biol. 8:613–621. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Yu FX, Zhao B and Guan KL: Hippo pathway in organ size control, tissue homeostasis, and cancer. Cell. 163:811–828. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Nokin MJ, Durieux F, Peixoto P, Chiavarina B, Peulen O, Blomme A, Turtoi A, Costanza B, Smargiasso N, Baiwir D, et al: Methylglyoxal, a glycolysis side-product, induces Hsp90 glycation and YAP-mediated tumor growth and metastasis. eLife. 5:52016. View Article : Google Scholar

39 

Chen KH, He J, Wang DL, Cao JJ, Li MC, Zhao XM, Sheng X, Li WB and Liu WJ: Methylation associated inactivation of LATS1 and its effect on demethylation or overexpression on YAP and cell biological function in human renal cell carcinoma. Int J Oncol. 45:2511–2521. 2014.PubMed/NCBI

40 

Zhang H, Geng D, Gao J, Qi Y, Shi Y, Wang Y, Jiang Y, Zhang Y, Fu J, Dong Y, et al: Expression and significance of Hippo/YAP signaling in glioma progression. Tumour Biol. 37:15665–15676. 2016. View Article : Google Scholar

41 

He J, Tang F, Liu L, Chen L, Li J, Ou D, Zhang L, Li Z, Feng D, Li W, et al: Positive regulation of TAZ expression by EBV-LMP1 contributes to cell proliferation and epithelial-mesenchymal transition in nasopharyngeal carcinoma. Oncotarget. Dec 2–2016.(Epub ahead of print). doi.org/10.18632/oncotarget.13775. View Article : Google Scholar

42 

Jiang Z, Li X, Hu J, Zhou W, Jiang Y, Li G and Lu D: Promoter hypermethylation-mediated down-regulation of LATS1 and LATS2 in human astrocytoma. Neurosci Res. 56:450–458. 2006. View Article : Google Scholar : PubMed/NCBI

43 

Sasaki H, Hikosaka Y, Kawano O, Yano M and Fujii Y: Hypermethylation of the large tumor suppressor genes in Japanese lung cancer. Oncol Lett. 1:303–307. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Oh JE, Ohta T, Satomi K, Foll M, Durand G, McKay J, Le Calvez-Kelm F, Mittelbronn M, Brokinkel B, Paulus W, et al: Alterations in the NF2/LATS1/LATS2/YAP pathway in schwannomas. J Neuropathol Exp Neurol. 74:952–959. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Reddy VR, Annamalai T, Narayanan V and Ramanathan A: Hypermethylation of promoter region of LATS1 - a CDK interacting protein in oral squamous cell carcinomas - a pilot study in India. Asian Pac J Cancer Prev. 16:1599–1603. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Karamitopoulou E, Zlobec I, Patsouris E, Peros G and Lugli A: Loss of E-cadherin independently predicts the lymph node status in colorectal cancer. Pathology. 43:133–137. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Li L, Fang R, Liu B, Shi H, Wang Y, Zhang W, Zhang X and Ye L: Deacetylation of tumor-suppressor MST1 in Hippo pathway induces its degradation through HBXIP-elevated HDAC6 in promotion of breast cancer growth. Oncogene. 35:4048–4057. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Rawat SJ and Chernoff J: Regulation of mammalian Ste20 (Mst) kinases. Trends Biochem Sci. 40:149–156. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Liang K, Zhou G, Zhang Q, Li J and Zhang C: Expression of hippo pathway in colorectal cancer. Saudi J Gastroenterol. 20:188–194. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Seidel C, Schagdarsurengin U, Blümke K, Würl P, Pfeifer GP, Hauptmann S, Taubert H and Dammann R: Frequent hypermethylation of MST1 and MST2 in soft tissue sarcoma. Mol Carcinog. 46:865–871. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Piccolo S, Dupont S and Cordenonsi M: The biology of YAP/TAZ: Hippo signaling and beyond. Physiol Rev. 94:1287–1312. 2014. View Article : Google Scholar : PubMed/NCBI

52 

Plouffe SW, Hong AW and Guan KL: Disease implications of the Hippo/YAP pathway. Trends Mol Med. 21:212–222. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Zhou D, Zhang Y, Wu H, Barry E, Yin Y, Lawrence E, Dawson D, Willis JE, Markowitz SD, Camargo FD, et al: Mst1 and Mst2 protein kinases restrain intestinal stem cell proliferation and colonic tumorigenesis by inhibition of Yes-associated protein (Yap) overabundance. Proc Natl Acad Sci USA. 108:E1312–E1320. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Cao JJ, Zhao XM, Wang DL, Chen KH, Sheng X, Li WB, Li MC, Liu WJ and He J: YAP is overexpressed in clear cell renal cell carcinoma and its knockdown reduces cell proliferation and induces cell cycle arrest and apoptosis. Oncol Rep. 32:1594–1600. 2014.PubMed/NCBI

55 

Dong C, Wei KJ, Zhang WB, Sun H, Pan HY and Zhang L: LATS2 induced by TNF-alpha and inhibited cell proliferation and invasion by phosphorylating YAP in oral squamous cell carcinoma. J Oral Pathol Med. 44:475–481. 2015. View Article : Google Scholar : PubMed/NCBI

56 

He C, Lv X, Hua G, Lele SM, Remmenga S, Dong J, Davis JS and Wang C: YAP forms autocrine loops with the ERBB pathway to regulate ovarian cancer initiation and progression. Oncogene. 34:6040–6054. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Jerhammar F, Johansson AC, Ceder R, Welander J, Jansson A, Grafström RC, Söderkvist P and Roberg K: YAP1 is a potential biomarker for cetuximab resistance in head and neck cancer. Oral Oncol. 50:832–839. 2014. View Article : Google Scholar : PubMed/NCBI

58 

Konsavage WM Jr, Kyler SL, Rennoll SA, Jin G and Yochum GS: Wnt/β-catenin signaling regulates Yes-associated protein (YAP) gene expression in colorectal carcinoma cells. J Biol Chem. 287:11730–11739. 2012. View Article : Google Scholar : PubMed/NCBI

59 

Yuan H, Liu H, Liu Z, Zhu D, Amos CI, Fang S, Lee JE and Wei Q: Genetic variants in Hippo pathway genes YAP1TEAD1TEAD4 are associated with melanoma-specific survival. Int J Cancer. 137:638–645. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Zhao J, Li X, Yang Y, Zhu D, Zhang C, Liu D, Wu K and Zhao S: Effect of YAP1 silencing on esophageal cancer. Onco Targets Ther. 9:3137–3146. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Li P, Sun D, Li X, He Y, Li W, Zhao J, Wang Y, Wang H and Xin Y: Elevated expression of Nodal and YAP1 is associated with poor prognosis of gastric adenocarcinoma. J Cancer Res Clin Oncol. 142:1765–1773. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Liao T, Wen D, Ma B, Hu JQ, Qu N, Shi RL, Liu L, Guan Q, Li DS and Ji QH: Yes-associated protein 1 promotes papillary thyroid cancer cell proliferation by activating the ERK/MAPK signaling pathway. Oncotarget. Feb 14–2017.(Epub ahead of print). doi.org/10.18632/oncotarget.14319.

63 

Pan Y, Robertson G, Pedersen L, Lim E, Hernandez-Herrera A, Rowat AC, Patil SL, Chan CK, Wen Y, Zhang X, et al: miR-509-3p is clinically significant and strongly attenuates cellular migration and multi-cellular spheroids in ovarian cancer. Oncotarget. 7:25930–25948. 2016.PubMed/NCBI

64 

Xu R, Zeng G, Gao J, Ren Y, Zhang Z, Zhang Q, Zhao J, Tao H and Li D: miR-138 suppresses the proliferation of oral squamous cell carcinoma cells by targeting Yes-associated protein 1. Oncol Rep. 34:2171–2178. 2015.PubMed/NCBI

65 

Xiao L, Zhou H, Li XP, Chen J, Fang C, Mao CX, Cui JJ, Zhang W, Zhou HH, Yin JY, et al: MicroRNA-138 acts as a tumor suppressor in non small cell lung cancer via targeting YAP1. Oncotarget. 7:40038–40046. 2016.PubMed/NCBI

66 

Lee KW, Lee SS, Kim SB, Sohn BH, Lee HS, Jang HJ, Park YY, Kopetz S, Kim SS, Oh SC, et al: Significant association of oncogene YAP1 with poor prognosis and cetuximab resistance in colorectal cancer patients. Clin Cancer Res. 21:357–364. 2015. View Article : Google Scholar : PubMed/NCBI

67 

Song S, Honjo S, Jin J, Chang SS, Scott AW, Chen Q, Kalhor N, Correa AM, Hofstetter WL, Albarracin CT, et al: The Hippo coactivator YAP1 mediates EGFR overexpression and confers chemoresistance in esophageal cancer. Clin Cancer Res. 21:2580–2590. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Touil Y, Igoudjil W, Corvaisier M, Dessein AF, Vandomme J, Monté D, Stechly L, Skrypek N, Langlois C, Grard G, et al: Colon cancer cells escape 5FU chemotherapy-induced cell death by entering stemness and quiescence associated with the c-Yes/YAP axis. Clin Cancer Res. 20:837–846. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Wang DY, Wu YN, Huang JQ, Wang W, Xu M, Jia JP, Han G, Mao BB and Bi WZ: Hippo/YAP signaling pathway is involved in osteosarcoma chemoresistance. Chin J Cancer. 35:472016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2017
Volume 38 Issue 1

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
Rybarczyk A, Klacz J, Wronska A, Matuszewski M, Kmiec Z and Wierzbicki PM: Overexpression of the YAP1 oncogene in clear cell renal cell carcinoma is associated with poor outcome. Oncol Rep 38: 427-439, 2017
APA
Rybarczyk, A., Klacz, J., Wronska, A., Matuszewski, M., Kmiec, Z., & Wierzbicki, P.M. (2017). Overexpression of the YAP1 oncogene in clear cell renal cell carcinoma is associated with poor outcome. Oncology Reports, 38, 427-439. https://doi.org/10.3892/or.2017.5642
MLA
Rybarczyk, A., Klacz, J., Wronska, A., Matuszewski, M., Kmiec, Z., Wierzbicki, P. M."Overexpression of the YAP1 oncogene in clear cell renal cell carcinoma is associated with poor outcome". Oncology Reports 38.1 (2017): 427-439.
Chicago
Rybarczyk, A., Klacz, J., Wronska, A., Matuszewski, M., Kmiec, Z., Wierzbicki, P. M."Overexpression of the YAP1 oncogene in clear cell renal cell carcinoma is associated with poor outcome". Oncology Reports 38, no. 1 (2017): 427-439. https://doi.org/10.3892/or.2017.5642