Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Int J Mol Sci ; 24(22)2023 Nov 18.
Article in English | MEDLINE | ID: mdl-38003666

ABSTRACT

Cadherins (calcium-dependent adhesion proteins) are important in cellular adhesion and may play a role in the development and progression of renal cell carcinoma (RCC). This study investigated changes in cadherin 3 (CDH3; P-cadherin) mRNA expression, DNA methylation, and protein expression in RCC and compared the results with the histopathological and clinical characteristics of patients. The possible contribution of CDH3 to tumor cell invasiveness was tested in a functional assay using siRNA-based suppression of CDH3 expression and subsequent real-time impedance analysis using a Matrigel invasion model. Our analyses revealed a tumor-specific loss of CDH3 mRNA expression, CDH3 DNA hypermethylation, and loss of distal tubular and collecting duct CDH3 protein expression in RCC. A relatively higher methylation level in tumors was associated with a loss of cell differentiation and higher clinical stage. siRNA-induced suppression of CDH3 expression modulated the invasion characteristics of tumor cells in the impedance-based real-time cellular analysis. Our results indicate that loss of CDH3 expression is common in RCC and may contribute to the pathogenesis of a subset of RCC. Further studies to reveal the mechanisms of loss of expression and its effects on the invasive behavior of renal tumor cells are required.


Subject(s)
Cadherins , Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Cadherins/metabolism , Carcinoma, Renal Cell/genetics , DNA Methylation , Kidney Neoplasms/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
2.
Int J Mol Sci ; 23(19)2022 Sep 23.
Article in English | MEDLINE | ID: mdl-36232491

ABSTRACT

Approximately 21% of patients with renal cell cancer (RCC) present with synchronous metastatic disease at the time of diagnosis, and metachronous metastatic disease occurs in 20-50% of cases within 5 years. Recent advances in adjuvant treatment of aggressive RCC following surgery suggest that biomarker-based prediction of risk for distant metastasis could improve patient selection. Biometrical analysis of TCGA-KIRC data identified candidate loci in the NK6 homeobox 2 gene (NKX6-2) that are hypermethylated in primary metastatic RCC. Analyses of NKX6-2 DNA methylation in three gene regions including a total of 16 CpG sites in 154 tumor-adjacent normal tissue, 189 RCC, and 194 metastatic tissue samples from 95 metastasized RCC patients revealed highly significant tumor-specific, primary metastatic-specific, and metastatic tissue-specific hypermethylation of NKX6-2. Combined CpG site methylation data for NKX6-2 and metastasis-associated genes (INA, NHLH2, and THBS4) demonstrated similarity between metastatic tissues and metastatic primary RCC tissues. The random forest method and evaluation of an unknown test cohort of tissues using receiver operator characteristic curve analysis revealed that metastatic tissues can be differentiated by a median area under the curve of 0.86 (p = 1.7 × 10-8-7.5 × 10-3) in 1000 random runs. Analysis of variable importance demonstrated an above median contribution for decision-making of at least one CpG site in each of the genes, suggesting superior informativity for sites annotated to NHLH2 and NKX6-2. Thus, DNA methylation of NKX6-2 is associated with the metastatic state of RCC tissues and contributes to a four-gene-based statistical predictor of tumoral and metastatic renal tissues.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Biomarkers , Carcinoma, Renal Cell/pathology , CpG Islands/genetics , DNA Methylation/genetics , Homeodomain Proteins/genetics , Humans , Kidney Neoplasms/pathology
3.
Int J Mol Sci ; 23(10)2022 May 10.
Article in English | MEDLINE | ID: mdl-35628134

ABSTRACT

Both age-dependent and age-independent alteration of DNA methylation in human tissues are functionally associated with the development of many malignant and non-malignant human diseases. TCGA-KIRC data were biometrically analyzed to identify new loci with age-dependent DNA methylation that may contribute to tumor risk in normal kidney tissue. ANKRD34B and ZIC1 were evaluated as candidate genes by pyrosequencing of 539 tissues, including 239 normal autopsy, 157 histopathologically tumor-adjacent normal, and 143 paired tumor kidney samples. All candidate CpG loci demonstrated a strong correlation between relative methylation levels and age (R = 0.70−0.88, p < 2 × 10−16) and seven out of 10 loci were capable of predicting chronological age in normal kidney tissues, explaining 84% of the variance (R = 0.92). Moreover, significantly increased age-independent methylation was found for 9 out of 10 CpG loci in tumor-adjacent tissues, compared to normal autopsy tissues (p = 0.001−0.028). Comparing tumor and paired tumor-adjacent tissues revealed two patient clusters showing hypermethylation, one cluster without significant changes in methylation, and a smaller cluster demonstrating hypomethylation in the tumors (p < 1 × 10−10). Taken together, our results show the presence of additional methylation risk factors besides age for renal cancer in normal kidney tissue. Concurrent tumor-specific hypermethylation suggests a subset of these loci are candidates for epigenetic renal cancer susceptibility.


Subject(s)
DNA Methylation , Kidney Neoplasms , Kidney , Repressor Proteins , Transcription Factors , Age Factors , CpG Islands , Epigenesis, Genetic , Genetic Predisposition to Disease , Humans , Kidney/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
BMC Cancer ; 21(1): 444, 2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33882870

ABSTRACT

BACKGROUND: DNA methylation is frequently observed in the development and progression of many human tumors as well as renal cell cancer (RCC). Tumor Associated Calcium Signal Transducer 2 (TACSTD2) participates in cell cycle progression through MAPK signalling pathway activation. Moreover, tumor-specific hypermethylation and association with aggressive cancer characteristics has been found for lung adenocarcinoma, hepatocellular carcinoma and cholangiocarcinoma. Whether TACSTD2 is tumor specifically hypermethylated in RCC or shows association of methylation with adverse clinicopathological parameters and survival of patients has not been investigated at yet. METHODS: Quantitative methylation-specific PCR (qMSP) analysis of a locus in the intron 1 region of TACSTD2 gene was carried out in a cross-sectional study of 127 paired RCC and normal samples. In silico analysis of TACSTD2 methylation in the TCGA Kidney Renal Clear Cell Carcinoma (KIRC) dataset of 280 patients served as validation cohort. Statistical analyses were carried out using the two-sided paired t-test for matched tumor and normal sample comparisons, logistic regression for subgroup comparisons, Cox regression for analysis of recurrence free survival (RFS) and Pearson correlation analysis for correlation of TACSTD2 methylation and TACSTD2 mRNA in KIRC data. RESULTS: Higher methylation levels in RCC were significantly associated with advanced disease (p < 0.001), high tumor stage (p = 0.003), tumor differentiation (p = 0.033) and presence of lymph node (p = 0.021) or distant metastases (p = 0.008). TACSTD2 hypermethylation was associated with a shorter RFS of patients and demonstrate statistical independency from clinical parameters as state of metastasis, tumor stage, grade and state of advanced disease. In silico validation using TCGA KIRC data also demonstrated association of TACSTD2 loci with adverse clinicopathology and shortened RFS of patients. In addition, in silico analyses of TCGA KIRC data showed an inverse correlation between DNA methylation levels of TACSTD2 and mRNA expression. CONCLUSIONS: Our results suggest an association between TACSTD2 methylation and disease progression and clinical course of RCC.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Calcium Signaling , Calcium/metabolism , Carcinoma, Renal Cell/etiology , Carcinoma, Renal Cell/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , DNA Methylation , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , CpG Islands , Disease Progression , Disease Susceptibility , Female , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Prognosis
5.
Cancers (Basel) ; 14(1)2021 Dec 22.
Article in English | MEDLINE | ID: mdl-35008203

ABSTRACT

The detection of DNA methylation in primary tumor tissues could be relevant for early stratification of aggressive renal cell carcinomas (RCCs) as a basis for future personalized adjuvant therapy. Methylated TCGA KIRC based candidate CpG loci in INA, NHLH2, and THBS4 that are possibly associated with RCC metastasis were evaluated by pyrosequencing in 154 paired normal adjacent and primary tumor tissues, as well as in 202 metastatic tissues. Statistical analysis was carried out by bivariate logistic regression for group comparisons, log rank survival analysis, and unsupervised and supervised analysis for the classification of tumors. Increased methylation of INA, NHLH2, and THBS4 loci were significantly associated with distant metastasis in primary tumors (p < 0.05), tissue-specific hypermethylation in metastatic (p = 7.88 × 10-8, 5.57 × 10-10, 2.06 × 10-7) and tumor tissues (p = 3.72 × 10-24, 3.17 × 10-13, 1.58 × 10-19), and shortened progression free survival in patients (p = 0.03). Combined use of CpG site-specific methylation permits the discrimination of tissues with metastatic disease and reveals a significant contribution of CpG sites in all genes to the statistical classification model. Thus, metastasis in RCC is significantly associated with methylation alterations in INA, NHLH2, and THBS4 loci, providing independent information for the potential early detection of aggressive renal cancers as a rationale for stratifying patients to adjuvant therapies.

6.
Clin Epigenetics ; 12(1): 33, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32070431

ABSTRACT

BACKGROUND: While a considerable number of tumor-specific hypermethylated loci have been identified in renal cell cancer (RCC), DNA methylation of loci showing successive increases in normal, tumoral, and metastatic tissues could point to genes with high relevance both for the process of tumor development and progression. Here, we report that DNA methylation of a locus in a genomic region corresponding to the 3'UTR of the transcription factor T-box brain 1 (TBR1) mRNA accumulates in normal renal tissues with age and possibly increased body mass index. Moreover, a further tissue-specific increase of methylation was observed for tumor and metastatic tissue samples. RESULTS: Biometric analyses of the TCGA KIRC methylation data revealed candidate loci for age-dependent and tumor-specific DNA methylation within the last exon and in a genomic region corresponding to the 3'UTR TBR1 mRNA. To evaluate whether methylation of TBR1 shows association with RCC carcinogenesis, we measured 15 tumor cell lines and 907 renal tissue samples including 355 normal tissues, 175 tissue pairs of normal tumor adjacent and corresponding tumor tissue as well 202 metastatic tissues samples of lung, bone, and brain metastases by the use of pyrosequencing. Statistical evaluation demonstrated age-dependent methylation in normal tissue (R = 0.72, p < 2 × 10-16), association with adiposity (P = 0.019) and tumor-specific hypermethylation (P = 6.1 × 10-19) for RCC tissues. Comparison of tumor and metastatic tissues revealed higher methylation in renal cancer metastases (P = 2.65 × 10-6). CONCLUSIONS: Our analyses provide statistical evidence of association between methylation of TBR1 and RCC development and disease progression.


Subject(s)
Carcinoma, Renal Cell/genetics , DNA Methylation/genetics , DNA, Neoplasm/genetics , Kidney Neoplasms/pathology , T-Box Domain Proteins/genetics , Adiposity/genetics , Adult , Aged , Aged, 80 and over , Brain/metabolism , Brain Neoplasms/genetics , Carcinogenesis/genetics , Carcinoma, Renal Cell/secondary , Cell Line, Tumor/metabolism , CpG Islands/genetics , Disease Progression , Epigenesis, Genetic/genetics , Female , Humans , Kidney/metabolism , Kidney/pathology , Male , Middle Aged , RNA, Messenger/genetics , Risk Factors
7.
Adv Ther ; 37(1): 288-299, 2020 01.
Article in English | MEDLINE | ID: mdl-31721113

ABSTRACT

INTRODUCTION: The corticotropin-releasing hormone (CRH) system, its receptors corticotropin-releasing hormone receptor 1 (CRHR1) and 2 (CRHR2), and its corresponding binding protein corticotropin-releasing hormone-binding protein (CRHBP) as well as the urocortin proteins-structural homologues to CRH, which are included in this peptide family-have become interesting oncological targets recently. Carcinogenesis of various human tumors has been reported with an altered presence of members of this system. The aim of the present study was to examine the role of urocortin 3 (UCN3) in renal cell carcinoma (RCC). METHODS: Therefore, tumoral tissues of 106 patients with RCC and available corresponding normal tissues were analyzed using qPCR for quantitative mRNA expression analysis. Tissue localization and protein signals of UCN3 in normal and tumoral renal specimens were evaluated using western blot and immunohistochemistry. In addition, correlation studies of UCN3 mRNA expression with clinicopathological parameters of patients with RCC and different histological subtypes were evaluated. RESULTS: UCN3 mRNA was significantly downregulated in nearly all tumoral tissues (p = 7.92 × 10-13). The same effect was observed at protein level using immunohistochemistry. Level of UCN3 mRNA expression was not directly correlated with clinicopathological parameters. CONCLUSION: We report for the first time the significant downregulation of UCN3 in RCC. These results demonstrate a possible involvement of the CRH system and its significance in carcinogenesis of RCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Corticotropin-Releasing Hormone/metabolism , Kidney Neoplasms/metabolism , Urocortins/metabolism , Blotting, Western , Carrier Proteins/metabolism , Female , Humans , Immunohistochemistry , Male , RNA, Messenger/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism
8.
Oncol Rep ; 42(5): 2159-2168, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31545450

ABSTRACT

DNA methylation plays an important role in the genesis and progression of tumor diseases. To identify new DNA methylation markers possibly associated with the clinical characteristics of renal cell carcinoma (RCC), we investigated loci in the sarcosine dehydrogenase (SARDH) gene. SARDH is involved in the metabolism of the glycine­derivative sarcosine and is closely linked through a functional control loop. Statistical evaluation of methylation data and clinical characteristics of patients showed that kidney tumors with clinically aggressive features such as a high tumor stage, positive lymph nodes, distant metastases or a previously advanced tumor status exhibited significantly lower methylation of a locus in the SARDH gene. Moreover, SARDH methylation was found to be a significant prognostic factor for recurrence­free survival in RCC patients showing statistical independence from the clinical prognosticators, grade, stage and state of metastasis. In conclusion, the methylation status of the SARDH­CGI was identified as an independent prognostic candidate marker for RCC.


Subject(s)
Carcinoma, Renal Cell/pathology , DNA Methylation , Kidney Neoplasms/pathology , Sarcosine Dehydrogenase/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Female , Humans , Kidney Neoplasms/genetics , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Prognosis , Survival Analysis
9.
Oncol Rep ; 40(6): 3861-3868, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30272321

ABSTRACT

Recent studies have shown that NELL1 expression is silenced epigenetically in human renal cell cancer (RCC) tissues and in RCC cell lines. However, it remains unknown whether NELL1 promoter methylation observed in clinical specimens might be associated with the clinicopathology or survival of patients with RCC. We analyzed NELL1 DNA methylation in tissues from patients with RCC and in adjacent normal renal tissues. In addition, we evaluated NELL1 methylation in cell lines derived from different urogenital tumors (prostate cancer, urothelial cancer and RCC). We performed regression analyses to determine whether NELL1 methylation is associated with clinicopathological parameters and recurrence­free survival (RFS). This cross­sectional study included 98 patients with RCC and 63 paired tumor and adjacent normal tissue samples. We analyzed a locus in the intron 1 region of NELL1 with pyrosequencing. We performed in silico analysis of NELL1 methylation in the TCGA Kidney Renal Clear Cell Carcinoma (KIRC) data set (n=284 patients), which served as a validation study. Statistical analyses were performed with the two­sided paired t­test for paired tumor and adjacent normal samples. We used logistic regression for subgroup comparisons and Cox regression for RFS comparisons. The mean methylation level was 6.8% higher in RCC tissues compared to paired adjacent normal tissues (paired t­test, P<0.001). Methylation levels in RCC were associated with advanced disease (P=0.002), the presence of distant metastases (P=0.004), and shorter RFS (P=0.035, HR: 4.15). In silico validation with TCGA KIRC data for adjacent loci also demonstrated that high relative methylation levels were associated with adverse clinicopathology and shortened RFS. Our results suggest that NELL1 methylation contributes to RCC disease progression. This finding could provide a clinical marker to complement recent functional analyses in tumor models.


Subject(s)
Carcinoma, Renal Cell/genetics , DNA Methylation , Kidney Neoplasms/genetics , Nerve Tissue Proteins/genetics , Sequence Analysis, DNA/methods , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Calcium-Binding Proteins , CpG Islands , Cross-Sectional Studies , Epigenesis, Genetic , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Promoter Regions, Genetic , Regression Analysis , Survival Analysis , Young Adult
10.
Adv Ther ; 35(11): 2054-2068, 2018 11.
Article in English | MEDLINE | ID: mdl-30232708

ABSTRACT

INTRODUCTION: There is an urgent need to identify patients with bladder cancer (BC) who are at high risk of recurrence or progression. Calgranulin A is a strong marker for muscle-invasive or advanced BC and recent studies have shown its potential for identifying patients at risk even in non-muscle-invasive bladder cancer (NMIBC). The present study examines risks of recurrence and progression dependent on immunostaining with calgranulin A in NMIBC. METHODS: Calgranulin A protein expression was evaluated through the immunohistochemistry of 158 randomly selected, transurethrally resected BC specimens of separate patients (pTa 89, pT1 69) using tissue microarrays. Kaplan-Meier survival analysis and Cox regression were performed to determine whether calgranulin A expression is associated with recurrence-free survival (RFS), progression-free survival (PFS), or cancer-specific survival (CSS). RESULTS: Calgranulin A expression is significantly different between pTa and pT1 tumors (p = 0.000, Mann-Whitney U test) and between tumor grades (p = 0.015, Kruskal-Wallis test). Kaplan-Meier estimates produced significant results for low and high calgranulin A expression concerning RFS [5y-RFS 70.4 ± 4.0% vs. 35.9 ± 12.5%, median RFS not reached (NR) vs. 12.0 ± 4.4 month, p = 0.029, log-rank test], PFS (5y-PFS 90.3 ± 2.7% vs. 51.5 ± 14.0%, median PFS NR in both groups, p = 0.000, log-rank test), and CSS (5y-CSS 92.9 ± 2.6% vs. 70.7 ± 12.4%, median CSS NR in both groups, p = 0.005, log-rank test). Calgranulin A remained an independent factor for RFS (p = 0.024, HR 2.43) and PFS (p = 0.002, HR 5.92) according to the multivariate Cox regression model. CONCLUSIONS: Calgranulin A expression in NMIBC, detected through immunohistochemistry, is a promising marker for the identification of NMIBC patients at high risk of recurrence and progression.


Subject(s)
Calgranulin A , Neoplasm Invasiveness/diagnosis , Neoplasm Recurrence, Local/diagnosis , Risk Assessment/methods , Urinary Bladder Neoplasms , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Calcium-Binding Proteins/analysis , Calcium-Binding Proteins/metabolism , Calgranulin A/analysis , Calgranulin A/metabolism , Disease Progression , Female , Humans , Immunohistochemistry , Male , Neoplasm Grading , Prognosis , Reproducibility of Results , Retrospective Studies , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
11.
Oncol Lett ; 14(6): 8190-8196, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29250194

ABSTRACT

Rho GDP dissociation inhibitor-ß (ARHGDIB) is an important mediator of cell signaling. The expression of ARHGDIB is associated with tumor growth and metastasis in a variety of non-genitourinary cancers; however, the role of ARHGDIB in renal cell carcinoma (RCC) has not yet been evaluated. In the present study, tissue samples from 105 patients undergoing surgery for RCC were obtained. The expression levels of ARHGDIB mRNA in normal kidney tissues and in corresponding cancer tissues were analyzed by reverse transcription-quantitative polymerase chain reaction. Differences in relative mRNA expression levels were assessed using paired two-sample t-tests. Expression levels were analyzed with respect to various clinical parameters, and associations were tested using a bivariate logistic regression model. Relative mRNA expression levels in healthy renal tissues compared with cancerous tissues from the same kidney were assessed using paired t-tests. Expression data were compared with respect to survival data by the Kaplan-Meier method/Cox regression analysis. The results revealed that the relative mRNA expression level of ARHGDIB was significantly higher in the lysates of RCC tumor tissues (P<0.001) when compared with healthy renal tissues in a paired analysis of 74 samples; this finding was consistent with the analysis of ARHGDIB mRNA expression levels in all RCC samples, as well as in the subset of clear cell RCC (ccRCC) samples. The relative mRNA expression level of ARHGDIB was also increased in ccRCC tissues compared with papillary RCC tissues (P<0.001). On univariate Cox regression analysis, recurrence-free survival (RFS) was significantly associated with metastasis, locally advanced disease and tumor grade (P=0.018, P=0.002 and P<0.001, respectively). Furthermore, in the subgroup of patients with ccRCC, increased ARHGDIB mRNA expression was significantly associated with a longer RFS time (P=0.001). In summary, the results indicate that ARHGDIB mRNA is highly expressed in RCC tissues in general and is positively associated with RFS in ccRCC. As ARHGDIB has a known effect on angiogenesis and immune modulation, the present study suggests that the functional analysis of ARHGDIB should be performed in the future.

12.
PLoS One ; 11(10): e0163873, 2016.
Article in English | MEDLINE | ID: mdl-27695045

ABSTRACT

The relevance of Corticotropin Releasing Hormone (CRH)-system in human malignancies is a question of growing interest. Here we investigated hypermethylation and epigenetic silencing of the CRH-Binding Protein (CRHBP) gene in clear cell renal cell cancer (ccRCC). Relative methylation of the CRHBP CpG island (CGI) was determined in 17 tumor cell lines as well as 86 ccRCC samples and 66 paired normal tissues using pyrosequencing and quantitative methylation specific PCR of bisulfite converted DNA. Results were statistically compared with relative mRNA expression levels of CRHBP and clinicopathological parameters of patients. Re-expression of CRHBP following 5-aza-2´-deoxycytidine treatment was investigated by quantitative mRNA expression analysis. Real-time impedance analysis was applied for analysis of invasiveness of renal tumor cells following si-RNA knockdown of CRHBP expression or ectopic expression of CRHBP. We found the CRHBP CGI to be frequently methylated in tumor cell lines of renal, prostatic, and bladder cancer. Comparison of methylation in normal and paired renal cancer tissue specimens revealed hypermethylation of the CRHBP CGI in tumors (p<1*10-12). DNA methylation and decreased mRNA expression were correlated (R = 0.83, p<1*10-12). Tumor cell lines showed 5-aza-2´-deoxycytidine dependent reduction of methylation and re-expression of CRHBP was associated with altered cellular invasiveness of renal cancer cells in real-time impedance invasion assays. Hypermethylation and inverse relationship with mRNA expression were validated in silico using the TCGA network data. We describe for the first time tumor specific epigenetic silencing of CRHBP and statistical association with aggressive tumors thus suggesting the CRH system to contribute to the development of kidney cancer.


Subject(s)
Carcinoma, Renal Cell/genetics , Carrier Proteins/genetics , DNA Methylation/genetics , Gene Silencing , Adult , Aged , Aged, 80 and over , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoplasm Metastasis , RNA, Messenger/genetics
13.
Anticancer Res ; 35(10): 5443-51, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26408707

ABSTRACT

BACKGROUND/AIM: The genetic characterization of prostate tumors is important for personalized therapy. The aim of the present study was to investigate the role of previously described prostate cancer-related genes in the genetic characterization of prostate tumors. MATERIALS AND METHODS: Forty-two genes were selected for expression analysis (real time-quantitative polymerase chain reaction). One normal prostatic epithelial cell line and three standardized prostate cancer cell lines were used. Twenty-eight patients treated with radical prostatectomy were included in the study. RESULTS: The following genes appeared to be possibly related to the metastatic potential of the tumor: ELOVL fatty acid elongase 7 (ELOVL7), enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2), gastrulation brain homeobox 2 (GBX2), golgi membrane protein 1 (GOLM1), homeobox C6 (HOXC6), minichromosome maintenance complex component 6 (MCM6), marker of proliferation Ki-67 (MKI67), mucin 1, cell surface associated (MUC1), MYC binding protein 2, E3 ubiquitin protein ligase (MYCBP2), somatostatin receptor 1 (SSTR1), topoisomerase (DNA) II alpha 170 kDa (TOP2A) and exportin 6 (XPO6). Six genes were differentially expressed in patients with localized and locally advanced cancer (GOLM1, GBX2, XPO6, SSTR1, TOP2A and cell division cycle associated 5, CDCA5) and three genes (HOXC6, Cyclin-dependent kinase inhibitor 2A (CDKN2A) and MYC binding protein 2, E3 ubiquitin protein ligase, MYCBP2) in patients with a low vs. high Gleason grade/sum. CONCLUSION: Some of the investigated genes show promising prognostic and classification features, which might be useful in a clinical setting, warranting for further validation.


Subject(s)
Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Aged , Cell Line, Tumor , Gene Expression Profiling/methods , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Pilot Projects , Prospective Studies , Prostatectomy , Prostatic Neoplasms/surgery
14.
Target Oncol ; 10(2): 267-75, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25230694

ABSTRACT

GATA-binding proteins 1 (GATA1) and 2 (GATA2) are zinc-finger transcription factors and belong to the GATA family proteins 1-6. GATA1 interacts with the TP53 tumor suppressor gene, and both GATAs have been shown to be involved in cell growth, apoptosis, and tumorigenesis of several solid tumors. GATA1 and GATA2 expression alterations are associated with poor survival and adverse clinicopathology in prostate and colorectal cancer, while the significance and prognostic value in clear cell renal cell carcinoma (ccRCC) has not been investigated as yet. We investigated relative messenger RNA (mRNA) expression levels of GATA1 and GATA2 in 77 ccRCC and 58 paired adjacent noncancerous renal tissues by quantitative real-time reverse-transcribed PCR. Relative mRNA expression levels were determined using the ΔΔCt method. GATA1 and GATA2 expression levels were significantly decreased in tumor tissues compared with normal tissues (p < 0.001, paired t test). In univariate logistic regression analysis, decreased GATA1 and GATA2 expression levels were associated with advanced tumor disease (p = 0.005 and 0.008), positive distant metastasis (p = 0.03 and 0.001), and lymph node metastasis status (p = 0.011 and 0.038). Reduced expression levels of GATA1 and GATA2 were associated with an increased risk of disease recurrence (p = 0.005 and 0.006; hazard ratio = 0.05 and 0.21). Pairwise bivariate analysis after adjusting for clinicopathological parameters revealed relative mRNA expression of GATA1, but not GATA2, as an independent candidate prognosticator for ccRCC. Our results support that GATA1 and GATA2 are involved in ccRCC tumor biology possibly affecting tumor development and aggressiveness.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , GATA1 Transcription Factor/genetics , GATA2 Transcription Factor/genetics , Kidney Neoplasms/genetics , RNA, Messenger/genetics , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/secondary , Carcinoma, Renal Cell/therapy , Cross-Sectional Studies , Disease-Free Survival , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Lymphatic Metastasis , Male , Middle Aged , Proportional Hazards Models , Real-Time Polymerase Chain Reaction , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors , Time Factors
15.
PLoS One ; 9(3): e91440, 2014.
Article in English | MEDLINE | ID: mdl-24633192

ABSTRACT

VEGF-targeted therapy increases both the progression-free (PFS) and overall survival (OS) of patients with metastasized renal cell cancer (mRCC). Identification of molecular phenotypes of RCC could improve risk-stratification and the prediction of the clinical disease course. We investigated whether gene-specific DNA hypermethylation can predict PFS and OS among patients undergoing anti-VEGF-based therapy. Primary tumor tissues from 18 patients receiving targeted therapy were examined retrospectively using quantitative methylation-specific PCR analysis of CST6, LAD1, hsa-miR-124-3, and hsa-miR-9-1 CpG islands. PFS and OS were analyzed for first-line and sequential antiangiogenic therapies using the log rank statistics. Sensitivity and specificity were determined for predicting first-line therapy failure. Hypermethylation of CST6 and LAD1 was associated with both a shortened PFS (log rank p = 0.009 and p = 0.004) and OS (p = 0.011 and p = 0.043). The median PFS observed for the high and low methylation groups of CST6 and LAD1 was 2.0 vs.11.4 months. LAD1 methylation had a specificity of 1.0 (95% CI 0.65-1.0) and a sensitivity of 0.73 (95% CI 0.43-0.90) for the prediction of first-line therapy. CST6 and LAD1 methylation are candidate epigenetic biomarkers showing unprecedented association with PFS and OS as well as specificity for the prediction of the response to therapy. DNA methylation markers should be considered for the prospective evaluation of larger patient cohorts in future studies.


Subject(s)
Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/mortality , DNA Methylation , Aged , Aged, 80 and over , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Cluster Analysis , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Prognosis , ROC Curve , Treatment Outcome
16.
BMC Cancer ; 14: 101, 2014 Feb 17.
Article in English | MEDLINE | ID: mdl-24533449

ABSTRACT

BACKGROUND: GATA-5, a zinc-finger transcription factor and member of the GATA family proteins 1-6, is known to be involved in cellular differentiation. We recently found that tumor-specific hypermethylation of the GATA5 CpG island (CGI) occurs in renal cell carcinoma (RCC) and is associated with an adverse clinical outcome. In this study, we investigated whether epigenetic GATA5 alterations may result in changes in GATA5 mRNA expression levels and correlate with the observed prognostic impact of epigenetic changes in GATA5 in RCC. METHODS: Quantitative real-time reverse-transcribed polymerase chain reaction was applied to measure relative GATA5 mRNA expression levels in 135 kidney tissue samples, including 77 clear cell RCC (ccRCC) tissues and 58 paired adjacent normal renal tissue samples. Relative GATA5 expression levels were determined using the ΔΔCt method and detection of three endogenous control genes then compared to previously measured values of relative methylation. RESULTS: The mean relative GATA5 mRNA expression level exhibited an approximately 31-fold reduction in tumor specimens compared with corresponding normal tissues (p < 0.001, paired t-test). Decreased GATA5 mRNA expression was inversely correlated with increased GATA5 CGI methylation (p < 0.001) and was associated with shortened recurrence-free survival in ccRCC patients (p = 0.023, hazard ratio = 0.25). CONCLUSION: GATA5 mRNA expression is decreased in ccRCC, likely due to gene silencing by methylation of the GATA5 CGI. Moreover, reduced GATA5 mRNA levels were associated with a poor clinical outcome, indicating a possible role of GATA5 for the development of aggressive ccRCC phenotypes.


Subject(s)
Carcinoma, Renal Cell/metabolism , CpG Islands/genetics , GATA5 Transcription Factor/biosynthesis , Neoplasm Recurrence, Local/metabolism , RNA, Messenger/biosynthesis , Aged , Biomarkers, Tumor/biosynthesis , Carcinoma, Renal Cell/mortality , DNA Methylation/genetics , Female , GATA5 Transcription Factor/antagonists & inhibitors , GATA5 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/mortality , RNA, Messenger/antagonists & inhibitors , Survival Rate/trends
17.
Cancer Med ; 3(2): 300-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24464810

ABSTRACT

Neurofilament Heavy polypeptid (NEFH) belongs to the group of type IV intermediate filament proteins. DNA methylation of the NEFH promoter and loss of expression have previously been shown to activate the AKT/ß-catenin pathway in tumor cells. When identifying hypermethylation of the NEFH CpG island (CGI) in renal cell cancer (RCC) we asked whether methylation could provide clinical or prognostic information for RCC and/or predict therapy response in patients with metastatic RCC (mRCC) undergoing antiangiogenic therapy. Relative methylation of the NEFH CGI was analyzed in 132 RCC samples and 83 paired normal tissues using quantitative methylation-specific PCR. Results were statistically compared with tumor histology, clinicopathological parameters, progression-free survival (PFS) as well as with overall survival (OS) in a subset of 18 mRCC patients following antiangiogenic therapy regimens. The NEFH CGI methylation demonstrated a tumor-specific increase (P < 0.001), association with advanced disease (P < 0.001), and distant metastasis (P = 0.005). Higher relative methylation was also significantly associated with a poor PFS (HR = 8.6, P < 0.001) independent from the covariates age, gender, diameter of tumors, state of advanced disease, and local and distant metastasis. Median OS following targeted therapy was 29.8 months for patients with low methylation versus 9.8 months for the group with high methylation (P = 0.028). We identified NEFH methylation as a candidate epigenetic marker for prognosis of RCC patients as well as prediction of anti-vascular endothelial growth factor-based therapy response.


Subject(s)
Carcinoma, Renal Cell/genetics , CpG Islands , DNA Methylation , Endothelial Growth Factors/antagonists & inhibitors , Kidney Neoplasms/genetics , Neurofilament Proteins/genetics , Aged , Angiogenesis Inhibitors/therapeutic use , Carcinoma, Renal Cell/blood , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cohort Studies , Cross-Sectional Studies , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Female , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Male , Prognosis , Promoter Regions, Genetic , Risk Assessment
18.
BMC Cancer ; 13: 199, 2013 Apr 22.
Article in English | MEDLINE | ID: mdl-23607589

ABSTRACT

BACKGROUND: Significance of Urocortin (Ucn or UcnI), Ucn2, Ucn3 and their receptors, Corticotropin Releasing Factor Receptor 1 and 2 (CRFR1 and CRFR2), and the binding protein, Corticotropin-Releasing Hormone-Binding Protein (CRHBP) in oncology is growing rapidly. The objective of our study was to assess the expression of the CRHBP mRNA and protein in renal cancer. METHODS: Tumoral tissues of 78 patients with clear cell renal cell cancer and their corresponding normal tissues were analyzed using quantitative mRNA expression analysis for detection of mRNA expression level. Protein expression and tissue localization of CRHBP protein in renal specimens was evaluated using western blotting, immunohistochemistry and double immunofluorescence, respectively. RESULTS: We found an approx. 33 fold decrease of average CRHBP mRNA level in tumoral tissues compared to paired normal tissues (p<0.001). Diminished CRHBP mRNA expression was positively correlated with advanced, metastasized and higher stage of disease (p<0.001, p=0.026, p=0.028 respectively). CRHBP protein was detected in glomeruli and proximal tubules of normal kidney while none or weak immunopositivity was found in cc-RCC (p<0.001). CONCLUSIONS: The expression analysis of CRHBP shows that cc-RCC is characterized by a significant loss of CRHBP mRNA expression that furthermore is associated with a more aggressive state of tumors. Depletion of CRHBP proteins also indicate that the protein as part of the UCN system may be involved in renal carcinogenesis.


Subject(s)
Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/secondary , Carrier Proteins/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , RNA, Messenger/metabolism , Aged , Carcinoma, Renal Cell/metabolism , Carrier Proteins/metabolism , Female , Humans , Kidney Glomerulus/metabolism , Kidney Neoplasms/metabolism , Kidney Tubules, Proximal/metabolism , Logistic Models , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging
19.
Transplantation ; 95(7): 928-32, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23388737

ABSTRACT

BACKGROUND: Recipient death is a leading cause for renal allograft loss. Cardiovascular mortality is the most important cause of death among this patient group. Single nucleotide polymorphisms (SNPs) in a noncoding region close to the CDKN2a/b senescence genes have been associated with higher cardiovascular morbidity and mortality in nontransplant populations. METHODS: We selected 2064 renal transplant recipients: 688 with a known cardiovascular cause of death and 1376 matched controls. DNA specimens were genotyped for the three SNPs with known risk allele (rs10757274, rs2383206, and rs10757278) and one SNP without risk allele (rs518394). Genotyping results were analyzed according to the frequency of risk alleles in the two groups. RESULTS: The risk allele for three SNPs was detected significantly more often in patients with a known cardiovascular cause of death than in matched controls (all P<0.05). Diabetes and time on dialysis were modifiers of this effect with the presence of high-risk alleles having a stronger impact in diabetic patients and those with longer dialysis time. There was no difference between groups for the investigated SNP without risk allele. CONCLUSIONS: Our results support data from large cohort studies in normal nontransplant populations, which suggested a higher risk for cardiovascular events in individuals carrying certain SNPs in senescence-associated genes. Notably, this finding was obtained in a population known to be at increased risk of cardiovascular death.


Subject(s)
Cardiovascular Diseases/genetics , Cardiovascular Diseases/mortality , Chromosomes, Human, Pair 9 , Kidney Transplantation/mortality , Polymorphism, Single Nucleotide , Adolescent , Adult , Aged , Case-Control Studies , Cause of Death , Chi-Square Distribution , Diabetes Mellitus/mortality , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Odds Ratio , Phenotype , Renal Dialysis/mortality , Risk Assessment , Risk Factors , Time Factors , Young Adult
20.
Urol Oncol ; 31(1): 74-81, 2013 Jan.
Article in English | MEDLINE | ID: mdl-21396839

ABSTRACT

BACKGROUND AND OBJECTIVES: Prostate cancer has a genetic component, and single nucleotide polymorphisms (SNPs) can contribute to the risk. We aimed to investigate the role of polymorphisms in 10 candidate genes with a key function in apoptosis. METHODS AND MATERIALS: Eight coding SNPs were chosen in ATM (Ser49Cys), BID (Ser56Cys), CASP8 (Asp302His), CASP10 (Val410Ile), LGALS3 (Pro64His), RASSF1 (Ser133Ala), TP53 (Arg72Pro), and TP53AIP1 (Ala7Val), and two non-coding SNPs were selected in BCL2 (-938C/A) and HDM2 (SNP309). A hospital-based case-control series of 510 prostate cancer patients and 490 healthy males from Northern Germany were genotyped for these polymorphisms. RESULTS: SNP rs4644 in LGALS3 showed evidence for a protective effect of the minor allele, encoding the His64 variant (OR 0.82, 95% CI 0.69;0.99, P = 0.04). Carriers were underrepresented among cases under a dominant model (OR 0.71; 95% CI 0.54;0.92; P = 0.01), and the effect appeared more pronounced in patients diagnosed before the age of 60 years (OR 0.52; 95% CI 0.31;0.85, P = 0.01). The other nine polymorphisms did not vary significantly between cases and controls, though subtle trends were noted for BCL2 (P = 0.07) and CASP10 (P = 0.08). The Asp302His variant of CASP8 tended to associate with a protective effect in the group with higher Gleason score under a dominant model (P = 0.03). Carriers of either the CASP8 or the CASP10 variants were underrepresented in the prostate cancer series (P = 0.02). CONCLUSIONS: These results provide first evidence to implicate the functional Pro64His variant of galectin-3 (LGALS3) in the genetic susceptibility towards prostate cancer. The potential role of polymorphisms in BCL2, CASP8, and CASP10 merits further investigation.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , Biomarkers, Tumor/genetics , Brachytherapy , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide/genetics , Prostatic Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Case-Control Studies , Follow-Up Studies , Germany , Hospitals, University , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , Prostatic Neoplasms/radiotherapy
SELECTION OF CITATIONS
SEARCH DETAIL
...