Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Int J Mol Sci ; 21(7)2020 Mar 30.
Article in English | MEDLINE | ID: mdl-32235585

ABSTRACT

The authors wish to make the following corrections to this paper [1]: in Figure 4 the same gelscans were mistakenly pasted to illustrate splicing changes of: i) BIM in KIJ-265T and KIJ308T cells,and ii) MCL-1 in UOK171 and KIJ-265T [...].

3.
Pol Przegl Chir ; 91(5): 12-20, 2019 Aug 31.
Article in English | MEDLINE | ID: mdl-31702571

ABSTRACT

The aim of our study was to evaluate of short-term outcomes of 4210 patients who underwent open abdominal surgery with used either single-layer, absorbable, or continuous sutures. Seromuscular anastomosis were presented (without mucosa). Fluid therapy: GDT and zero-balance GDT were also discussed. Anastomosis leakage developed in 6 patients. They were treated by creating a stoma. Simultaneously, septic shock was treated. Re-anastomoses were performed after some time and after sepsis was suppressed. Results of treatment with use of double-layer sutures, which was is use before 1978, were presented. Analysis of 536 patients treated at the same time was conducted and 53 patients with anastomosis leakage was identified. Out of this group, 2 patients survived. Based on the literature review, the methods for performing anastomosis. Attention was paid to the advantages of single layer over multi-layer sutures: effectiveness, simplicity and lower cost of treatment. The occurrence of mechanical ileus was less frequently observed since ceasing to employ peritoneoplasty, and preoperational preparation of gastrointestinal tract in patients had a beneficial influence on their postoperative course. Postoperative complications were discussed mainly based on additional examinations such as TC and MRI. The attention was focused on the importance of medical observation and clinical examination by an experienced surgeon in order to identify postoperative complications. Antibiotic treatment in cases of postoperative complications was presented, as well as fluid therapy: GDT, zero-balance GDT and the procedures employed in cases of complications such as: anastomosis leakage, mechanical ileus, inter-peritoneal abscess. Anastomotic stenosis was not observed in this group of patients.


Subject(s)
Absorbable Implants/adverse effects , Anastomosis, Surgical/adverse effects , Anastomotic Leak/etiology , Digestive System Surgical Procedures/methods , Postoperative Complications/etiology , Anastomosis, Surgical/methods , Digestive System Surgical Procedures/adverse effects , Female , Humans , Male , Middle Aged , Suture Techniques/instrumentation
4.
Cancers (Basel) ; 11(12)2019 Nov 20.
Article in English | MEDLINE | ID: mdl-31756931

ABSTRACT

Metabolic reprogramming is one of the hallmarks of renal cell cancer (RCC). We hypothesized that altered metabolism of RCC cells results from dysregulation of microRNAs targeting metabolically relevant genes. Combined large-scale transcriptomic and metabolic analysis of RCC patients tissue samples revealed a group of microRNAs that contribute to metabolic reprogramming in RCC. miRNAs expressions correlated with their predicted target genes and with gas chromatography-mass spectrometry (GC-MS) metabolome profiles of RCC tumors. Assays performed in RCC-derived cell lines showed that miR-146a-5p and miR-155-5p targeted genes of PPP (the pentose phosphate pathway) (G6PD and TKT), the TCA (tricarboxylic acid cycle) cycle (SUCLG2), and arginine metabolism (GATM), respectively. miR-106b-5p and miR-122-5p regulated the NFAT5 osmoregulatory transcription factor. Altered expressions of G6PD, TKT, SUCLG2, GATM, miR-106b-5p, miR-155-5p, and miR-342-3p correlated with poor survival of RCC patients. miR-106b-5p, miR-146a-5p, and miR-342-3p stimulated proliferation of RCC cells. The analysis involving >6000 patients revealed that miR-34a-5p, miR-106b-5p, miR-146a-5p, and miR-155-5p are PanCancer metabomiRs possibly involved in global regulation of cancer metabolism. In conclusion, we found that microRNAs upregulated in renal cancer contribute to disturbed expression of key genes involved in the regulation of RCC metabolome. miR-146a-5p and miR-155-5p emerge as a key "metabomiRs" that target genes of crucial metabolic pathways (PPP (the pentose phosphate pathway), TCA cycle, and arginine metabolism).

5.
Exp Cell Res ; 363(2): 208-217, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29331391

ABSTRACT

SRSF1, SRSF2 and hnRNP A1 are splicing factors that regulate the expression of oncogenes and tumor suppressors. SRSF1 and SRSF2 contribute to the carcinogenesis in the kidney. Despite their importance, the mechanisms regulating their expression in cancer are not entirely understood. Here, we investigated the microRNA-mediated regulation of SRSF1, SRSF2 and hnRNP A1 in renal cancer. The expression of microRNAs predicted to target SRSF1, SRSF2 and hnRNP A1 was disturbed in renal tumors compared with controls. Using qPCR, Western blot/ICC and luciferase reporter system assays we identified microRNAs that contribute to the regulation of expression of SRSF1 (miR-10b-5p, miR-203a-3p), SRSF2 (miR-183-5p, miR-200c-3p), and hnRNP A1 (miR-135a-5p, miR-149-5p). Silencing of SRSF1 and SRSF2 enhanced the expression of their targeting microRNAs. miR-183-5p and miR-200c-3p affected the expression of SRSF2-target genes, TNFRSF1B, TNFRSF9, CRADD and TP53. 3'UTR variants of SRSF1 and SRSF2 differed by the presence of miRNA-binding sites. In conclusion, we identified a group of microRNAs that contribute to the regulation of expression of SRSF1, SRSF2 and hnRNP A1. The microRNAs targeting SRSF1 and SRSF2 are involved in a regulatory feedback loop. microRNAs miR-183-5p and miR-200c-3p that target SRSF2, affect the expression of genes involved in apoptotic regulation.


Subject(s)
3' Untranslated Regions/genetics , Gene Expression Regulation, Neoplastic , Heterogeneous Nuclear Ribonucleoprotein A1/genetics , MicroRNAs/genetics , Serine-Arginine Splicing Factors/genetics , Cell Line, Tumor , Gene Expression Profiling , Humans , RNA Splicing/genetics
6.
Cancer Lett ; 412: 155-169, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29079415

ABSTRACT

In our previous study we found altered expression of 19 adhesion-related genes in renal tumors. In this study we hypothesized that disturbed expression of adhesion-related genes could be caused by microRNAs: short, non-coding RNAs that regulate gene expression. Here, we found that expression of 24 microRNAs predicted to target adhesion-related genes was disturbed in renal tumors and correlated with expression of their predicted targets. miR-25-3p, miR-30a-5p, miR-328 and miR-363-3p directly targeted adhesion-related genes, including COL5A1, COL11A1, ITGA5, MMP16 and THBS2. miR-363-3p and miR-328 inhibited proliferation of renal cancer cells, while miR-25-3p inhibited adhesion, promoted proliferation and migration of renal cancer cells. TGF-ß1 influenced the expression of miR-25-3p, miR-30a-5p, and miR-328. The analyzed microRNAs, their target genes and TGF-ß1 formed a network of strong correlations in tissue samples from renal cancer patients. The expression signature of microRNAs linked with TGF-ß1 levels correlated with poor survival of renal cancer patients. The results of our study suggest that TGF-ß1 coordinates the expression of microRNA network that regulates cellular adhesion in cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Kidney Neoplasms/pathology , MicroRNAs/physiology , Transforming Growth Factor beta1/physiology , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Collagen Type V/genetics , Computational Biology , Extracellular Matrix/physiology , Gene Regulatory Networks , Humans , Integrin alphaV/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/mortality
7.
Biochim Biophys Acta Mol Basis Dis ; 1863(3): 744-752, 2017 03.
Article in English | MEDLINE | ID: mdl-28012969

ABSTRACT

PURPOSE: Cellular metabolism of renal cell carcinoma (RCC) tumors is disturbed. The clinical significance of these alterations is weakly understood. We aimed to find if changes in metabolic pathways contribute to survival of RCC patients. MATERIAL AND METHODS: 35 RCC tumors and matched controls were used for metabolite profiling using gas chromatography-mass spectrometry and transcriptomic analysis with qPCR-arrays targeting the expression of 93 metabolic genes. The clinical significance of obtained data was validated on independent cohort of 468 RCC patients with median follow-up of 43.22months. RESULTS: The levels of 31 metabolites were statistically significantly changed in RCC tumors compared with controls. The top altered metabolites included beta-alanine (+4.2-fold), glucose (+3.4-fold), succinate (-11.0-fold), myo-inositol (-4.6-fold), adenine (-4.2-fold), uracil (-3.7-fold), and hypoxanthine (-3.0-fold). These disturbances were associated with altered expression of 53 metabolic genes. ROC curve analysis revealed that the top metabolites discriminating between tumor and control samples included succinate (AUC=0.91), adenine (AUC=0.89), myo-inositol (AUC=0.87), hypoxanthine (AUC=0.85), urea (AUC=0.85), and beta-alanine (AUC=0.85). Poor survival of RCC patients correlated (p<0.0001) with altered expression of genes involved in metabolism of succinate (HR=2.7), purines (HR=2.4), glucose (HR=2.4), beta-alanine (HR=2.5), and myo-inositol (HR=1.9). CONCLUSIONS: We found that changes in metabolism of succinate, beta-alanine, purines, glucose and myo-inositol correlate with poor survival of RCC patients.


Subject(s)
Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Metabolome , Transcriptome , Carcinoma, Renal Cell/epidemiology , Carcinoma, Renal Cell/metabolism , Female , Gas Chromatography-Mass Spectrometry , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genomics , Humans , Inositol/genetics , Inositol/metabolism , Kidney Neoplasms/epidemiology , Kidney Neoplasms/metabolism , Male , Metabolic Networks and Pathways , Metabolomics , Survival Analysis , beta-Alanine/genetics , beta-Alanine/metabolism
8.
Int J Mol Sci ; 17(10)2016 Sep 28.
Article in English | MEDLINE | ID: mdl-27690003

ABSTRACT

Serine and arginine rich splicing factor 2(SRSF2) belongs to the serine/arginine (SR)-rich family of proteins that regulate alternative splicing. Previous studies suggested that SRSF2 can contribute to carcinogenic processes. Clear cell renal cell carcinoma (ccRCC) is the most common subtype of kidney cancer, highly aggressive and difficult to treat, mainly due to resistance to apoptosis. In this study we hypothesized that SRSF2 contributes to the regulation of apoptosis in ccRCC. Using tissue samples obtained from ccRCC patients, as well as independent validation on The Cancer Genome Atlas (TCGA) data, we demonstrate for the first time that expression of SRSF2 is decreased in ccRCC tumours when compared to non-tumorous control tissues. Furthermore, by employing a panel of ccRCC-derived cell lines with silenced SRSF2 expression and qPCR arrays we show that SRSF2 contributes not only to splicing patterns but also to expression of multiple apoptotic genes, including new SRSF2 targets: DIABLO, BIRC5/survivin, TRAIL, BIM, MCL1, TNFRSF9, TNFRSF1B, CRADD, BCL2L2, BCL2A1, and TP53. We also identified a new splice variant of CFLAR, an inhibitor of caspase activity. These changes culminate in diminished caspase-9 activity and inhibition of apoptosis. In summary, we show for the first time that decreased expression of SRSF2 in ccRCC contributes to protection of cancer cells viability.

9.
J Urol ; 195(6): 1892-902, 2016 06.
Article in English | MEDLINE | ID: mdl-26631499

ABSTRACT

PURPOSE: Renal cell carcinoma is the most common highly metastatic kidney malignancy. Adhesion has a crucial role in the metastatic process. TGF (transforming growth factor)-ß1 is a pleiotropic cytokine that influences cancerous transformation. We hypothesized that 1) changes in the expression of adhesion related genes may influence survival rate of patients with renal cell carcinoma and 2) TGF-ß1 may contribute to changed expression of adhesion related genes. MATERIALS AND METHODS: Two-step quantitative real-time polymerase chain reaction arrays were used to analyze the expression of adhesion related genes in 77 tumors and matched pair controls. The prognostic significance of genes was evaluated in TCGA (The Cancer Genome Atlas) data on 468 patients with renal cell carcinoma. Quantitative real-time polymerase chain reaction and Western blot were applied for TGF-ß1 analysis. TGF-ß1 mediated regulation of gene expression was analyzed by TGF-ß1 supplementation of Caki-2 cells and quantitative real-time polymerase chain reaction. RESULTS: The expression of 19 genes related to adhesion and extracellular matrix remodeling was statistically significantly disturbed in renal cell carcinoma compared with controls. The 10-gene expression signature (COL1A1, COL5A1, COL11A1, FN1, ICAM1, ITGAL, ITGAM, ITGB2, THBS2 and TIMP1) correlated with poor survival (HR 2.85, p = 5.7e-10). TGF-ß1 expression was 22 times higher in renal cell carcinoma than in controls (p <0.0001). TGF-ß1 induced expression of TGFBI, COL1A1, COL5A1, COL8A1, FN1, ITGA5, ITGAM and TIMP1 in a renal cell carcinoma derived cell line. CONCLUSIONS: Disturbed expression of genes involved in adhesion and extracellular matrix remodeling develops early during renal cell carcinoma carcinogenesis and correlates with poor survival. TGF-ß1 contributes to changed expression of extracellular matrix and adhesion related genes. Bioinformatic analysis performed on a broad panel of cancers of nonkidney origin suggests that disturbed expression of genes related to extracellular matrix and adhesion may be a universal feature of cancerous progression.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Cell Adhesion/genetics , Extracellular Matrix/genetics , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/genetics , Transforming Growth Factor beta1/metabolism , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/mortality , Case-Control Studies , Cell Line, Tumor , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/mortality , Prognosis , Proportional Hazards Models , Real-Time Polymerase Chain Reaction , Survival Rate
10.
PLoS One ; 5(10): e13690, 2010 Oct 27.
Article in English | MEDLINE | ID: mdl-21082031

ABSTRACT

BACKGROUND: Clear cell renal cell carcinoma (ccRCC) is the most common type of renal cancer. One of the processes disturbed in this cancer type is alternative splicing, although phenomena underlying these disturbances remain unknown. Alternative splicing consists of selective removal of introns and joining of residual exons of the primary transcript, to produce mRNA molecules of different sequence. Splicing aberrations may lead to tumoral transformation due to synthesis of impaired splice variants with oncogenic potential. In this paper we hypothesized that disturbed alternative splicing in ccRCC may result from improper expression of splicing factors, mediators of splicing reactions. METHODOLOGY/PRINCIPAL FINDINGS: Using real-time PCR and Western-blot analysis we analyzed expression of seven splicing factors belonging to SR proteins family (SF2/ASF, SC35, SRp20, SRp75, SRp40, SRp55 and 9G8), and one non-SR factor, hnRNP A1 (heterogeneous nuclear ribonucleoprotein A1) in 38 pairs of tumor-control ccRCC samples. Moreover, we analyzed splicing patterns of five genes involved in carcinogenesis and partially regulated by analyzed splicing factors: RON, CEACAM1, Rac1, Caspase-9, and GLI1. CONCLUSIONS/SIGNIFICANCE: We found that the mRNA expression of splicing factors was disturbed in tumors when compared to paired controls, similarly as levels of SF2/ASF and hnRNP A1 proteins. The correlation coefficients between expression levels of specific splicing factors were increased in tumor samples. Moreover, alternative splicing of five analyzed genes was also disturbed in ccRCC samples and splicing pattern of two of them, Caspase-9 and CEACAM1 correlated with expression of SF2/ASF in tumors. We conclude that disturbed expression of splicing factors in ccRCC may possibly lead to impaired alternative splicing of genes regulating tumor growth and this way contribute to the process of carcinogenesis.


Subject(s)
Alternative Splicing , Apoptosis/physiology , Carcinoma, Renal Cell/genetics , Genes, Tumor Suppressor , Kidney Neoplasms/genetics , Oncogenes , Blotting, Western , Carcinoma, Renal Cell/pathology , Humans , Kidney Neoplasms/pathology , Polymerase Chain Reaction
11.
Biochim Biophys Acta ; 1802(11): 995-1005, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20691260

ABSTRACT

Thyroid hormone receptor ß1 (TRß1) is a hormone-dependent transcription factor activated by 3,5,3'-l-triiodothyronine (T3). TRß1 functions as a tumor suppressor and disturbances of the THRB gene are frequent findings in cancer. Translational control mediated by untranslated regions (UTRs) regulates cell proliferation, metabolism and responses to cellular stress, processes that are involved in carcinogenesis. We hypothesized that reduced TRß1 expression in clear cell renal cell cancer (ccRCC) results from regulatory effects of TRß1 5' and 3'UTRs on protein translation. We determined TRß1 expression and alternative splicing of TRß1 5' and 3'UTRs in ccRCC and control tissue together with expression of the type 1 deiodinase enzyme (coded by DIO1, a TRß1 target gene). Tissue concentrations of T3 (which are generated in part by D1) and expression of miRNA-204 (an mRNA inhibitor for which a putative interaction site was identified in the TRß1 3'UTR) were also determined. TRß1 mRNA and protein levels were reduced by 70% and 91% in ccRCC and accompanied by absent D1 protein, a 58% reduction in tissue T3 concentration and 2-fold increase in miRNA-204. Structural analysis of TRß1 UTR variants indicated that reduced TRß1 expression may be maintained in ccRCC by posttranscriptional mechanisms involving 5'UTRs and miRNA-204. The tumor suppressor activity of TRß1 indicates that reduced TRß1 expression and tissue hypothyroidism in ccRCC tumors is likely to be involved in the process of carcinogenesis or in maintaining a proliferative advantage to malignant cells.


Subject(s)
Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Thyroid Hormone Receptors beta/genetics , Untranslated Regions/genetics , 5' Untranslated Regions/genetics , Alternative Splicing , Base Sequence , Blotting, Western , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Genetic , Molecular Sequence Data , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Triiodothyronine/metabolism
12.
Thyroid ; 19(10): 1105-13, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19534619

ABSTRACT

BACKGROUND: Alternative splicing, one of the sources of protein diversity, is often disturbed in cancer. Type 1 iodothyronine deiodinase (DIO1) catalyzes deiodination of thyroxine generating triiodothyronine, an important regulator of cell proliferation and differentiation. The expression of DIO1 is disturbed in different types of cancer. The aim of the study was to analyze the alternative splicing of DIO1 and its possible disturbance in renal cancer. METHODS: Using real-time PCR, we analyzed 19 tissue samples (T) of renal cancer and 19 matched control samples (C) of the opposite pole of the kidney, not infiltrated by tumor, and 6 control samples (N) (nonneoplastic kidney abnormalities). RESULTS: Cloning of DIO1 mRNA isoforms revealed 11 different transcripts, among them 7 new splice variants, not previously reported. The expression of all variants of DIO1 was dramatically (>90%) and significantly (p < or = 0.0003) lowered in samples T compared to control samples C. The ratio of mRNA isoforms encoding DIO1 protein variants possessing or lacking the active center was lowered in samples T compared with control samples C, suggesting disturbed alternative splicing of DIO1. The expression of mRNA of splicing factors SF2/ASF (splicing factor-2/alternative-splicing factor) and hnRNPA1 (heterogeneous ribonucleoprotein A1), regulating 5'-splice site selection, was significantly but not proportionally lowered in samples T compared to samples C. The mRNA ratio of splicing factors SF2/ASF and hnRNPA1 correlated with the ratio of mRNA isoforms encoding DIO1 protein variants possessing or lacking the active center in controls C but not in samples T. CONCLUSIONS: Our results show that the expression and alternative splicing of DIO1 mRNA is disturbed in renal cancer, possibly due to changes in expression of splicing factors SF2/ASF and hnRNPA1.


Subject(s)
Gene Expression Regulation, Enzymologic/genetics , Iodide Peroxidase/genetics , Kidney Neoplasms/genetics , Protein Isoforms/genetics , Cloning, Molecular , DNA Primers , Databases, Genetic , Humans , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
Thyroid ; 17(11): 1039-48, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17910524

ABSTRACT

We show here that the promoter of E2F1 gene, encoding one of the key regulators of cell proliferation, is overly active in the presence of low amounts of triiodothyronine (T3) and in the presence of mutant thyroid hormone receptor. We also show that T3-thyroid hormone receptor pathway of regulation of molecular processes is disturbed in clear cell renal cell carcinoma (ccRCC) on several levels, including overexpression of thyroid hormone receptors and the disturbance of their binding to DNA and to the hormone. In comparison to the cancer-free kidneys and peritumoral respective control tissues, E2F1 mRNA and protein levels are significantly increased in cancer tissues. A significant correlation between E2F1 mRNA and protein levels has been found in both control types and ccRCCs. No correlation was observed between the amount of E2F1 mRNA and the amount of thyroid hormone receptors or their DNA or T3 binding activity, suggesting that the function of thyroid hormone receptors could be markedly disturbed in both tumor and peritumoral cells. In summary, we show that ccRCC is characterized by the overexpression of E2F1, which is likely a result of a deregulated control of T3-dependent molecular processes.


Subject(s)
Carcinoma, Renal Cell/metabolism , E2F1 Transcription Factor/biosynthesis , Receptors, Thyroid Hormone/physiology , Adult , Aged , Aged, 80 and over , DNA/metabolism , Female , Gene Expression Regulation, Neoplastic/physiology , Humans , Male , Middle Aged , Promoter Regions, Genetic/physiology , RNA, Messenger/metabolism , Thyroid Hormone Receptors alpha/biosynthesis , Thyroid Hormone Receptors beta/biosynthesis , Triiodothyronine/metabolism
14.
Acta Biochim Pol ; 54(3): 595-602, 2007.
Article in English | MEDLINE | ID: mdl-17726548

ABSTRACT

The expression of cyclin E gene (CCNE) in relation to the expression of its major regulatory protein, E2F1, was examined in clear cell renal cell carcinomas (ccRCC). We show that the overexpression of E2F1 is accompanied by the significant increase of the mean amounts of cyclin E mRNA, as well as of total cyclin E protein and its low molecular weight forms in cancer tissues as compared to peritumoral controls. A significant increase of the mean amount of total cyclin E was found in peritumoral tissues compared to cancer-free kidneys, suggesting that cancer cells might secrete factors having a profound influence on the metabolism of neighbouring tissues. A significant, positive correlations between E2F1 protein and total cyclin E mRNA, as well as between E2F1 protein and full length cyclin E protein were found in cancer-free kidneys and in peritumoral tissues, but not in ccRCCs. The overexpression of cyclin E positively correlated with the decreasing degree of tumor differentiation, implicating a role for cyclin E in the promotion of tumorigenesis.


Subject(s)
Carcinoma, Renal Cell/genetics , Cyclin E/genetics , Liver Neoplasms/genetics , Oncogene Proteins/genetics , Blotting, Western , Carcinoma, Renal Cell/metabolism , Cyclin E/metabolism , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Electrophoretic Mobility Shift Assay , Humans , Kidney/metabolism , Kidney/pathology , Liver Neoplasms/metabolism , Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
Nephron Exp Nephrol ; 93(4): e150-7, 2003.
Article in English | MEDLINE | ID: mdl-12759576

ABSTRACT

Vitamin D co-regulates cell proliferation, differentiation and apoptosis, the processes that are disturbed in cancer tissues. It acts through the vitamin D nuclear receptor (VDR) that binds to DNA in the regulatory sequences of the target genes. As the kidney is one of the key organs for vitamin D metabolism and action, we analyzed VDR expression and its DNA binding activity in human renal clear cell cancer. 24 tumors, 24 controls that were excised from the opposite pole of the same kidney and 7 controls originating from kidneys without cancer were examined. Independently of tumor grading neither Northern blots nor immunoblotting demonstrated statistically significant differences of the mean VDR mRNA and protein amounts, respectively, in the cancer as compared to both control types. In contrast, the amount of VDR-DNA complexes was lower in 52.2% of the tumors in comparison to their corresponding controls. After normalization against VDR receptor protein amount in 34.8% of the tumors VDR-DNA binding was at least 3-4 times weaker than in the controls. However, the expression of vitamin D-dependent P21 gene on the mRNA level was not decreased in these cancers. It remains to be elucidated if altered VDR function due to its impaired binding to DNA contributes to the process of tumorigenesis, and what potential vitamin D-dependent mechanisms are involved in this process.


Subject(s)
Adenocarcinoma, Clear Cell/metabolism , Carcinoma, Renal Cell/metabolism , DNA, Neoplasm/metabolism , Kidney Neoplasms/metabolism , Receptors, Calcitriol/biosynthesis , Receptors, Calcitriol/metabolism , Adenocarcinoma, Clear Cell/genetics , Blotting, Northern , Blotting, Western , Carcinoma, Renal Cell/genetics , Cell Nucleus/chemistry , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Neoplastic/genetics , Humans , Kidney Neoplasms/genetics , Nuclear Proteins/analysis , RNA, Messenger/analysis , RNA, Messenger/genetics , Receptors, Calcitriol/genetics , Vitamin D Response Element/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...