Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Biomolecules ; 10(10)2020 09 23.
Article in English | MEDLINE | ID: mdl-32977498

ABSTRACT

Syndecan-1 is a transmembrane heparan sulfate proteoglycan which is indispensable in the structural and functional integrity of epithelia. Normal hepatocytes display strong cell surface expression of syndecan-1; however, upon malignant transformation, they may lose it from their cell surfaces. In this study, we demonstrate that re-expression of full-length or ectodomain-deleted syndecan-1 in hepatocellular carcinoma cells downregulates phosphorylation of ERK1/2 and p38, with the truncated form exerting an even stronger effect than the full-length protein. Furthermore, overexpression of syndecan-1 in hepatoma cells is associated with a shift of heparan sulfate structure toward a highly sulfated type specific for normal liver. As a result, cell proliferation and proteolytic shedding of syndecan-1 from the cell surface are restrained, which facilitates redifferentiation of hepatoma cells to a more hepatocyte-like phenotype. Our results highlight the importance of syndecan-1 in the formation and maintenance of differentiated epithelial characteristics in hepatocytes partly via the HGF/ERK/Ets-1 signal transduction pathway. Downregulation of Ets-1 expression alone, however, was not sufficient to replicate the phenotype of syndecan-1 overexpressing cells, indicating the need for additional molecular mechanisms. Accordingly, a reporter gene assay revealed the inhibition of Ets-1 as well as AP-1 transcription factor-induced promoter activation, presumably an effect of the heparan sulfate switch.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Proto-Oncogene Protein c-ets-1/genetics , Syndecan-1/genetics , Transcription Factor AP-1/genetics , Carcinoma, Hepatocellular/pathology , Cell Differentiation/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Heparitin Sulfate/pharmacology , Hepatocyte Growth Factor/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver Neoplasms/pathology , MAP Kinase Signaling System/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
2.
PLoS One ; 15(6): e0234873, 2020.
Article in English | MEDLINE | ID: mdl-32559232

ABSTRACT

OBJECTIVE: Comparison of human mRNA microarray results from tumor-associated and normal cervical fibroblasts revealed significant TFPI2 downregulation in tumor-associated fibroblasts isolated from cervical cancer, indicating that TFPI2 downregulation may play an important role in the pathogenesis of the disease. In the present work, we investigated the mechanism of TFPI2 downregulation in tumor-associated fibroblasts and tumor cells. METHODS: In vitro models of monocultures and co-cultures were established with tumor cells and fibroblasts to explore the changes of TFPI-2 expression and epigenetic modifications of the TFPI2 gene. RESULTS: The TFPI2 gene was hypermethylated only in tumor cells. Reduction of TFPI-2 protein levels in tumor-associated fibroblasts, although the gene was not methylated, suggested alternative regulatory mechanisms of gene expression, such as inhibition by microRNAs. The expression pattern of miR-23a, a gene thought to inhibit TFPI2 translation, showed changes strongly correlated to detected TFPI-2 protein alterations. Transfections with miR-23a mimics resulted in a decrease of TFPI-2 protein expression whereas miR-23a inhibitors increased the TFPI-2 amount. Due to downregulation of miR-23a expression by HPV in cancer cells, TFPI2 was silenced by promoter methylation. In contrary, miR-23a was active in HPV-free fibroblasts and inactivated TFPI2. CONCLUSION: These results indicate dual epigenetic inhibition of TFPI2 on the transcription level by promoter methylation in cancer cells and on the translation level by miR-23a in tumor-associated fibroblasts. As a consequence, inactivation of the TFPI2 gene plays a strategic role in the progression of cervical cancer.


Subject(s)
Gene Silencing , Glycoproteins/genetics , Uterine Cervical Neoplasms/genetics , Adult , Cancer-Associated Fibroblasts/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Glycoproteins/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Tumor Cells, Cultured
3.
Pathol Oncol Res ; 26(4): 2209-2223, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32307642

ABSTRACT

Up-regulation of the long non-coding RNA LINC00152 can contribute to cancer development, proliferation and invasion, including colorectal cancer, however, its mechanism of action in colorectal carcinogenesis and progression is only insufficiently understood. In this work we correlated LINC00152 expression with promoter DNA methylation changes in colorectal tissues along the normal-adenoma-carcinoma sequence and studied the effects of LINC00152 silencing on the cell cycle regulation and on the whole transcriptome in colon carcinoma cells using cell and molecular biology techniques. LINC00152 was significantly up-regulated in adenoma and colorectal cancer (p < 0.001) compared to normal samples, which was confirmed by real-time PCR and in situ hybridization. LINC00152 promoter hypomethylation detected in colorectal cancer (p < 0.01) was strongly correlated with increased LINC00152 expression (r=-0.90). Silencing of LINC00152 significantly suppressed cell growth, induced apoptosis and decreased cyclin D1 expression (p < 0.05). Whole transcriptome analysis of LINC00152-silenced cells revealed significant down-regulation of oncogenic and metastasis promoting genes (e.g. YES proto-oncogene 1, PORCN porcupine O-acyltransferase), and up-regulation of tumour suppressor genes (e.g. DKK1 dickkopf WNT signalling pathway inhibitor 1, PERP p53 apoptosis effector) (adjusted p < 0.05). Pathway analysis confirmed the LINC00152-related activation of oncogenic molecular pathways including those driven by PI3K/Akt, Ras, WNT, TP53, Notch and ErbB. Our results suggest that promoter hypomethylation related overexpression of LINC00152 can contribute to the pathogenesis of colorectal cancer by facilitating cell progression through the up-regulation of several oncogenic and metastasis promoting pathway elements.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/pathology , DNA Methylation , Promoter Regions, Genetic , RNA, Long Noncoding/genetics , Aged , Carcinogenesis , Case-Control Studies , Colorectal Neoplasms/genetics , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Prognosis , Proto-Oncogene Mas , Transcriptome
4.
Orv Hetil ; 159(2): 64-69, 2018 Jan.
Article in Hungarian | MEDLINE | ID: mdl-29307221

ABSTRACT

INTRODUCTION: According to the international literature, DNA methylation analysis of the promoter region of SNRPN locus is the most efficient way to start genetic investigation in patients with suspected Prader-Willi syndrome. AIM: Our aim was to develop a simple, reliable first-tier diagnosis to confirm Prader-Willi syndrome, therefore to compare our self-designed simple, cost-efficient high-resolution melting analysis and the most commonly used methylation-specific multiplex ligation-dependent probe amplification to confirm Prader-Willi syndrome. METHOD: We studied 17 clinically suspected Prader-Willi syndrome children and their DNA samples. With self-designed primers, bisulfite-sensitive polymerase chain reaction, high-resolution melting analysis and, as a control, methylation-specific multiplex ligation-dependent probe amplification were performed. RESULTS: Prader-Willi syndrome was genetically confirmed in 6 out of 17 clinically suspected Prader-Willi syndrome patients. The results of high-resolution melting analysis and methylation-specific multiplex ligation-dependent probe amplification were equivalent in each case. CONCLUSION: Using our self-designed primers and altered bisulfite-specific PCR conditions, high-resolution melting analysis appears to be a simple, fast, reliable and effective method for primarily proving or excluding clinically suspected Prade-Willi syndrome cases. Orv Hetil. 2018; 159(2): 64-69.


Subject(s)
Nucleic Acid Amplification Techniques/methods , Polymerase Chain Reaction/methods , Prader-Willi Syndrome/diagnosis , Child , Child, Preschool , Chromosomes, Human, Pair 15/genetics , Female , Genotype , Humans , Male , Prader-Willi Syndrome/genetics
5.
Obes Facts ; 10(4): 353-362, 2017.
Article in English | MEDLINE | ID: mdl-28787727

ABSTRACT

OBJECTIVE: Worldwide increasing childhood obesity is due to interactions between environmental and genetic factors, linked together by epigenetic mechanisms such as DNA methylation. METHODS: 82 obese children (>95th BMI percentile , age: 3-18 years) were included. Anthropometric data, metabolic parameters, 25-OH vitamin D (25OHD), and pubertal status were recorded, 24-hour blood pressure monitoring was performed. BMI standard deviation score (SDS) was calculated. Using candidate gene approach, obesity- (insulin-like growth factor 2 (IGF2), proopiomelanocortin (POMC)) and vitamin D metabolism-related genes (1-alfa-hydroxylase (CYP27B1), VDR) regulated by DNA methylation were selected. After isolating DNA from peripheral blood, bisulfite conversion, bisulfite specific polymerase chain reaction (BS-PCR), and pyrosequencing were carried out. RESULTS: No significant correlation between 25-OHD and metabolic parameters and DNA methylation status, but a tendency of positive correlation between VDR methylation status and 25-OHD (r = 0.2053,p = 0.066) were observed. Significant positive correlations between BMI SDS and CYP27B1 hypermethylation (r = 0.2371,p = 0.0342) and a significant negative correlation between IGF2 hypomethylation and BMI SDS (r = -0.305,p = 0.0059) were found. Conclusions Rate of obesity shows correlation with DNA methylation. Hypomethylation of IGF2 and hypermethylation of CYP27B1 genes might positively influence the rate of BMI observed in obese children.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , DNA Methylation , Epigenesis, Genetic/genetics , Insulin-Like Growth Factor II/genetics , Pediatric Obesity/genetics , Adolescent , Body Mass Index , Child , Child, Preschool , Female , Humans , Male , Receptors, Calcitriol/genetics
6.
J Cancer ; 8(2): 162-173, 2017.
Article in English | MEDLINE | ID: mdl-28243320

ABSTRACT

Background: To support cancer therapy, development of low cost library preparation techniques for targeted next generation sequencing (NGS) is needed. In this study we designed and tested a PCR-based library preparation panel with limited target area for sequencing the top 12 somatic mutation hot spots in colorectal cancer on the GS Junior instrument. Materials and Methods: A multiplex PCR panel was designed to amplify regions of mutation hot spots in 12 selected genes (APC, BRAF, CTNNB1, EGFR, FBXW7, KRAS, NRAS, MSH6, PIK3CA, SMAD2, SMAD4, TP53). Amplicons were sequenced on a GS Junior instrument using ligated and barcoded adaptors. Eight samples were sequenced in a single run. Colonic DNA samples (8 normal mucosa; 33 adenomas; 17 adenocarcinomas) as well as HT-29 and Caco-2 cell lines with known mutation profiles were analyzed. Variants found by the panel on APC, BRAF, KRAS and NRAS genes were validated by conventional sequencing. Results: In total, 34 kinds of mutations were detected including two novel mutations (FBXW7 c.1740:C>G and SMAD4 c.413C>G) that have not been recorded in mutation databases, and one potential germline mutation (APC). The most frequently mutated genes were APC, TP53 and KRAS with 30%, 15% and 21% frequencies in adenomas and 29%, 53% and 29% frequencies in carcinomas, respectively. In cell lines, all the expected mutations were detected except for one located in a homopolymer region. According to re-sequencing results sensitivity and specificity was 100% and 92% respectively. Conclusions: Our NGS-based screening panel denotes a promising step towards low cost colorectal cancer genotyping on the GS Junior instrument. Despite the relatively low coverage, we discovered two novel mutations and obtained mutation frequencies comparable to literature data. Additionally, as an advantage, this panel requires less template DNA than sequence capture colon cancer panels currently available for the GS Junior instrument.

7.
Oncotarget ; 7(26): 39293-39301, 2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27283768

ABSTRACT

PURPOSE: Targeted therapy represents an attractive alternative for rare tumors such as urachal carcinoma (UrC). The aim of this study was to assess the mutations of the most commonly affected 5 genes in the targetable EGFR-pathway in UrC and comapre their frequencies to those of found in urothelial and colorectal cancer. MATERIALS AND METHODS: Mutational hot-spots of selected genes were tested in 22 UrC samples by pyrosequencing. Mutational patterns were compared to those published for colorectal and urothelial cancers. Furthermore, we sought correlations between mutations and clinicopathological and follow-up data. RESULTS: We found 11 mutations in 10 of 22 (45%) patients. The most frequently mutated gene was KRAS (27%) followed by BRAF (18%) and NRAS (5%), while no mutations were detected in the EGFR and PIK3CA genes. No correlation was found between the mutation status and clinicopathological parameters (Sheldon/Mayo stage, tumor grade, metastases). Furthermore, none of the mutations correlated with progression-free or overall survival. CONCLUSIONS: The mutation pattern of UrC is more similar to colorectal than to urothelial cancer. However, the mutation characteristics of UrC seems to be unique suggesting that clinical decision-making for UrC cannot be simply adopted from urothelial or colorectal carcinoma. The high occurence of EGFR-pathway mutations warrants the testing for KRAS and BRAF mutations when considering anti-EGFR therapy in UrC.


Subject(s)
Adenocarcinoma/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , ErbB Receptors/genetics , GTP Phosphohydrolases/genetics , Membrane Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Urinary Bladder Neoplasms/genetics , Adult , Aged , Carcinoma/genetics , Colorectal Neoplasms/genetics , DNA Mutational Analysis , Disease Progression , Disease-Free Survival , Female , Follow-Up Studies , Humans , Male , Middle Aged , Mutation , Neoplasm Metastasis , Prognosis , Recurrence , Retrospective Studies , Treatment Outcome , Urothelium/pathology
8.
BMC Cancer ; 15: 736, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26482433

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) development is accompanied by changes in expression for several genes; but the details of the underlying regulatory procesess remain unknown. Our aims were to assess the role of epigenetic processes in tumour formation and to identify characteristic DNA methylation and miRNA alterations in the colorectal adenoma-carcinoma sequence. METHODS: Whole genome expression profiling was performed on colonic biopsy samples (49 healthy normal, 49 colorectal adenoma (AD), 49 CRC); on laser capture microdissected (LCM) epithelial and stromal cells from 6 CRC-normal adjacent tissue (NAT) samples pairs, and on demethylated human CRC cell lines using HGU133 Plus 2.0 microarrays (Affymetrix). Methylation status of genes with gradually altering expression along the AD-CRC sequence was further analysed on 10-10 macrodissected and 5-5 LCM samples from healthy colon, from adenoma and from CRC biopsy samples using bisulfite-sequencing PCR (BS-PCR) followed by pyrosequencing. In silico miRNA prediction for the selected genes was performed with miRWALK algorithm, miRNA expression was analysed on 3 CRC-NAT sample pairs and 3 adenoma tissue samples using the Human Panel I + II (Exiqon). SFRP1 immunohistochemistry experiments were performed. RESULTS: A set of transcripts (18 genes including MAL, SFRP1, SULT1A1, PRIMA1, PTGDR) showed decreasing expression (p < 0.01) in the biopsy samples along the adenoma-carcinoma sequence. Three of those (COL1A2, SFRP2, SOCS3) showed hypermethylation and THBS2 showed hypomethylation both in AD and in CRC samples compared to NAT, while BCL2, PRIMA1 and PTGDR showed hypermethylation only in the CRC group. miR-21 was found to be significantly (p < 0.01) upregulated in adenoma and tumour samples compared to the healthy colonic tissue controls and could explain the altered expression of genes for which DNA methylation changes do not appear to play role (e.g. BCL2, MAL, PTGS2). Demethylation treatment could upregulate gene expression of genes that were found to be hypermethylated in human CRC tissue samples. Decreasing protein levels of SFRP1 was also observed along the adenoma-carcinoma sequence. CONCLUSION: Hypermethylation of the selected markers (MAL, PRIMA1, PTGDR and SFRP1) can result in reduced gene expression and may contribute to the formation of colorectal cancer.


Subject(s)
Adenoma/genetics , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Myelin and Lymphocyte-Associated Proteolipid Proteins/genetics , Nerve Tissue Proteins/genetics , Receptors, Immunologic/genetics , Receptors, Prostaglandin/genetics , Adenoma/metabolism , Adenoma/pathology , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Methylation , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/biosynthesis , Membrane Proteins/biosynthesis , Myelin and Lymphocyte-Associated Proteolipid Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Polymerase Chain Reaction , Promoter Regions, Genetic , RNA, Messenger/genetics , Receptors, Immunologic/biosynthesis , Receptors, Prostaglandin/biosynthesis
9.
PLoS One ; 10(8): e0133836, 2015.
Article in English | MEDLINE | ID: mdl-26291085

ABSTRACT

Microarray analysis of promoter hypermethylation provides insight into the role and extent of DNA methylation in the development of colorectal cancer (CRC) and may be co-monitored with the appearance of driver mutations. Colonic biopsy samples were obtained endoscopically from 10 normal, 23 adenoma (17 low-grade (LGD) and 6 high-grade dysplasia (HGD)), and 8 ulcerative colitis (UC) patients (4 active and 4 inactive). CRC samples were obtained from 24 patients (17 primary, 7 metastatic (MCRC)), 7 of them with synchronous LGD. Field effects were analyzed in tissues 1 cm (n = 5) and 10 cm (n = 5) from the margin of CRC. Tissue materials were studied for DNA methylation status using a 96 gene panel and for KRAS and BRAF mutations. Expression levels were assayed using whole genomic mRNA arrays. SFRP1 was further examined by immunohistochemistry. HT29 cells were treated with 5-aza-2' deoxycytidine to analyze the reversal possibility of DNA methylation. More than 85% of tumor samples showed hypermethylation in 10 genes (SFRP1, SST, BNC1, MAL, SLIT2, SFRP2, SLIT3, ALDH1A3, TMEFF2, WIF1), whereas the frequency of examined mutations were below 25%. These genes distinguished precancerous and cancerous lesions from inflamed and healthy tissue. The mRNA alterations that might be caused by systematic methylation could be partly reversed by demethylation treatment. Systematic changes in methylation patterns were observed early in CRC carcinogenesis, occuring in precursor lesions and CRC. Thus we conclude that DNA hypermethylation is an early and systematic event in colorectal carcinogenesis, and it could be potentially reversed by systematic demethylation therapy, but it would need more in vitro and in vivo experiments to support this theory.


Subject(s)
Adenoma/genetics , Carcinoma/genetics , Colorectal Neoplasms/genetics , DNA Methylation/genetics , Transcriptome/genetics , Adolescent , Cell Line, Tumor , Colitis, Ulcerative/genetics , Gene Expression Regulation, Neoplastic/genetics , HT29 Cells , Humans , Mutation/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics
10.
Pathol Oncol Res ; 21(4): 1149-56, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25991403

ABSTRACT

We aimed to test the applicability of formalin-fixed and paraffin-embedded (FFPE) tissue samples for gene specific DNA methylation analysis after using two commercially available DNA isolation kits. Genomic DNA was isolated from 5 colorectal adenocarcinomas and 5 normal adjacent tissues from "recent", collected within 6 months, and "archived", collected more than 5 years ago, FFPE tissues using either High Pure FFPET DNA Isolation kit or QIAamp DNA FFPE Tissue kit. DNA methylation analysis of MAL, SFRP1 and SFRP2 genes, known to be hypermethylated in CRC, was performed using methylation-sensitive high resolution melting (MS-HRM) analysis and sequencing. QIAamp (Q) method resulted in slightly higher recovery in archived (HP: 1.22 ± 3.18 µg DNA; Q: 3.00 ± 4.04 µg DNA) and significantly (p < 0.05) higher recovery in recent samples compared to High Pure method (HP) (HP: 4.10 ± 2.91 µg DNA; Q: 11.51 ± 7.50 µg DNA). Both OD260/280 and OD260/230 ratios were lower, but still high in the High Pure isolated archived and recent samples compared to those isolated with QIAamp. Identical DNA methylation patterns were detected for all 3 genes tested by MS-HRM with both isolation kits in the recent group. However, despite of higher DNA recovery in QIAamp slightly more reproducible methylation results were obtained from High Pure isolated archived samples. Sequencing confirmed DNA hypermethylation in CRCs. In conclusion, reproducible DNA methylation patterns were obtained from recent samples using both isolation kits. However, long term storage may affect the reliability of the results leading to moderate differences between the efficiency of isolation kits.


Subject(s)
Colorectal Neoplasms/genetics , DNA Methylation/genetics , DNA/genetics , Formaldehyde/chemistry , Paraffin/chemistry , Sulfites/chemistry , Humans , Paraffin Embedding/methods , Reproducibility of Results , Sequence Analysis, DNA/methods
11.
BMC Cancer ; 15: 256, 2015 Apr 11.
Article in English | MEDLINE | ID: mdl-25885552

ABSTRACT

BACKGROUND: Comparison of tissue microarray results of 29 cervical cancer and 27 normal cervix tissue samples using immunohistochemistry revealed considerable reorganization of the fibrillar stroma of these tumors. Preliminary densitometry analysis of laminin-1, α-smooth muscle actin (SMA) and fibronectin immunostaining demonstrated 3.8-fold upregulation of laminin-1 and 5.2-fold increase of SMA in the interstitial stroma, indicating that these proteins and the activated fibroblasts play important role in the pathogenesis of cervical cancer. In the present work we investigated the role of normal and tumor-associated fibroblasts. METHODS: In vitro models were used to throw light on the multifactorial process of tumor-stroma interaction, by means of studying the cooperation between tumor cells and fibroblasts. Fibroblasts from normal cervix and cervical cancers were grown either separately or in co-culture with CSCC7 cervical cancer cell line. Changes manifest in secreted glycoproteins, integrins and matrix metallo-proteases (MMPs) were explored. RESULTS: While normal fibroblasts produced components of interstitial matrix and TGF-ß1 that promoted cell proliferation, cancer-associated fibroblasts (CAFs) synthesized ample amounts of laminin-1. The following results support the significance of laminin-1 in the invasion of CSCC7 cells: 1.) Tumor-associated fibroblasts produced more laminin-1 and less components of fibrillar ECM than normal cells; 2.) The production of laminin chains was further increased when CSCC7 cells were grown in co-culture with fibroblasts; 3.) CSCC7 cells were capable of increasing their laminin production; 4.) Tumor cells predominantly expressed integrin α6ß4 laminin receptors and migrated towards laminin. The integrin profile of both normal and tumor-associated fibroblasts was similar, expressing receptors for fibronectin, vitronectin and osteopontin. MMP-7 secreted by CSCC7 cells was upregulated by the presence of normal fibroblasts, whereas MMP-2 produced mainly by fibroblasts was activated in the presence of CSCC7 cells. CONCLUSIONS: Our results indicate that in addition to degradation of the basement membrane, invasion of cervical cancer is accomplished by the remodeling of the interstitial stroma, which process includes decrease and partial replacement of fibronectin and collagens by a laminin-rich matrix.


Subject(s)
Carcinogenesis/genetics , Extracellular Matrix/metabolism , Fibronectins/biosynthesis , Laminin/biosynthesis , Transforming Growth Factor beta1/biosynthesis , Uterine Cervical Neoplasms/genetics , Actins/biosynthesis , Actins/genetics , Adult , Aged , Basement Membrane/metabolism , Basement Membrane/pathology , Cell Proliferation/genetics , Coculture Techniques , Extracellular Matrix/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Fibronectins/genetics , Gene Expression Regulation, Neoplastic , Humans , Integrins/biosynthesis , Integrins/genetics , Laminin/genetics , Middle Aged , Tissue Array Analysis , Transforming Growth Factor beta1/genetics , Uterine Cervical Neoplasms/pathology
12.
Epigenomics ; 5(5): 569-81, 2013.
Article in English | MEDLINE | ID: mdl-24059802

ABSTRACT

DNA methylation analysis methods have undergone an impressive revolution over the past 15 years. Regarding colorectal cancer (CRC), the localization and distribution of several differently methylated genes have been determined by genome-wide DNA methylation assays. These genes do not just influence the pathogenesis of CRC, but can be used further as diagnostic or prognostic markers. Moreover, the identified four DNA methylation-based subgroups of CRC have important clinical and therapeutic merit. Since genome-wide DNA methylation analyzes result in a large amount of data, there is a need for complex bioinformatic and pathway analysis. Future challenges in epigenetic alterations of CRC include the demand for comprehensive identification and experimental validation of gene abnormalities. By introduction of genome-wide DNA methylation profiling into clinical practice not only the patients' risk stratification but development of targeted therapies will also be possible.


Subject(s)
Biomarkers, Tumor , DNA Methylation , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Epigenomics , Humans
13.
PLoS One ; 7(6): e39474, 2012.
Article in English | MEDLINE | ID: mdl-22745764

ABSTRACT

Syndecans are transmembrane heparan sulphate proteoglycans. Their role in the development of the malignant phenotype is ambiguous and depends upon the particular type of cancer. Nevertheless, syndecans are promising targets in cancer therapy, and it is important to elucidate the mechanisms controlling their various cellular effects. According to earlier studies, both syndecan-1 and syndecan-2 promote malignancy of HT-1080 human fibrosarcoma cells, by increasing the proliferation rate and the metastatic potential and migratory ability, respectively. To better understand their tumour promoter role in this cell line, syndecan expression levels were modulated in HT-1080 cells and the growth rate, chemotaxis and invasion capacity were studied. For in vivo testing, syndecan-1 overexpressing cells were also inoculated into mice. Overexpression of full length or truncated syndecan-1 lacking the entire ectodomain but containing the four juxtamembrane amino acids promoted proliferation and chemotaxis. These effects were accompanied by a marked increase in syndecan-2 protein expression. The pro-migratory and pro-proliferative effects of truncated syndecan-1 were not observable when syndecan-2 was silenced. Antisense silencing of syndecan-2, but not that of syndecan-1, inhibited cell migration. In vivo, both full length and truncated syndecan-1 increased tumour growth and metastatic rate. Based on our in vitro results, we conclude that the tumour promoter role of syndecan-1 observed in HT-1080 cells is independent of its ectodomain; however, in vivo the presence of the ectodomain further increases tumour proliferation. The enhanced migratory ability induced by syndecan-1 overexpression is mediated by syndecan-2. Overexpression of syndecan-1 also leads to activation of IGF1R and increased expression of Ets-1. These changes were not evident when syndecan-2 was overexpressed. These findings suggest the involvement of IGF1R and Ets-1 in the induction of syndecan-2 synthesis and stimulation of proliferation by syndecan-1. This is the first report demonstrating that syndecan-1 enhances malignancy of a mesenchymal tumour cell line, via induction of syndecan-2 expression.


Subject(s)
Cell Movement/physiology , Syndecan-1/metabolism , Syndecan-2/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Humans , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Syndecan-1/genetics , Syndecan-2/genetics
14.
PLoS One ; 4(10): e7346, 2009 Oct 05.
Article in English | MEDLINE | ID: mdl-19802384

ABSTRACT

Syndecan-1 forms complexes with growth factors and their cognate receptors in the cell membrane. We have previously reported a tubulin-mediated translocation of syndecan-1 to the nucleus. The transport route and functional significance of nuclear syndecan-1 is still incompletely understood. Here we investigate the sub-cellular distribution of syndecan-1, FGF-2, FGFR-1 and heparanase in malignant mesenchymal tumor cells, and explore the possibility of their coordinated translocation to the nucleus. To elucidate a structural requirement for this nuclear transport, we have transfected cells with a syndecan-1/EGFP construct or with a short truncated version containing only the tubulin binding RMKKK sequence. The sub-cellular distribution of the EGFP fusion proteins was monitored by fluorescence microscopy. Our data indicate that syndecan-1, FGF-2 and heparanase co-localize in the nucleus, whereas FGFR-1 is enriched mainly in the perinuclear area. Overexpression of syndecan-1 results in increased nuclear accumulation of FGF-2, demonstrating the functional importance of syndecan-1 for this nuclear transport. Interestingly, exogenously added FGF-2 does not follow the route taken by endogenous FGF-2. Furthermore, we prove that the RMKKK sequence of syndecan-1 is necessary and sufficient for nuclear translocation, acting as a nuclear localization signal, and the Arginine residue is vital for this localization. We conclude that syndecan-1 and FGF-2, but not FGFR-1 share a common transport route and co-localize with heparanase in the nucleus, and this transport is mediated by the RMKKK motif in syndecan-1. Our study opens a new perspective in the proteoglycan field and provides more evidence of nuclear interactions of syndecan-1.


Subject(s)
Cell Nucleus/metabolism , Fibroblast Growth Factor 2/biosynthesis , Gene Expression Regulation, Neoplastic , Glucuronidase/biosynthesis , Mesoderm/metabolism , Neoplasms/metabolism , Receptor, Fibroblast Growth Factor, Type 1/biosynthesis , Syndecan-1/biosynthesis , Syndecan-1/chemistry , Cell Line, Tumor , DNA Mutational Analysis , Fibroblast Growth Factor 2/physiology , Glucuronidase/physiology , Green Fluorescent Proteins/metabolism , Humans , Immunoprecipitation , Microscopy, Confocal , Models, Biological , Receptor, Fibroblast Growth Factor, Type 1/physiology , Syndecan-1/physiology
15.
Int J Cancer ; 125(2): 318-27, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19330836

ABSTRACT

A growing number of studies indicate the importance of the lysyl oxidase family in the promotion of epithelial neoplasms towards their more aggressive forms. However, the role of individual family members in carcinoma progression has yet to be ascertained. In this study, we analyzed LOXL2 expression in malignantly transformed MCF-7 and normal MCF-10A mammary epithelial cell line clones stably transduced with LOXL2 in vitro, and in normal and cancerous breast tissue samples in vivo. We found LOXL2 to be catalytically active in both MCF-7 and MCF-10 clones. LOXL2 overexpression promoted a more mesenchymal morphology in both cell types, but LOXL2-induced increase in migratory ability could only be established in MCF-7 clones. We demonstrated altered localization of the LOXL2 protein in breast cancer tissue compared to normal mammary tissue, and altered localization and processing of LOXL2 protein in breast cancer cell lines compared to normal cell lines, which may allow LOXL2 to interact with different intra and extracellular components during tumor progression. Results support the role of LOXL2 in selectively promoting a metastatic phenotype in breast tumor cells. Additional data suggest epigenetic molecular mechanisms in tumor specific regulation of LOXL2 expression that could be explored as a molecular target in the prevention of breast cancer progression.


Subject(s)
Amino Acid Oxidoreductases/physiology , Breast Neoplasms/pathology , Mammary Glands, Human/cytology , Neoplasm Metastasis , Amino Acid Oxidoreductases/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Blotting, Western , Breast Neoplasms/enzymology , Catalysis , Cell Line , Cell Line, Tumor , Decitabine , Gene Expression Regulation, Enzymologic/drug effects , Humans , Hydroxamic Acids/pharmacology , Immunohistochemistry , Mammary Glands, Human/enzymology , Reverse Transcriptase Polymerase Chain Reaction
16.
Genes Chromosomes Cancer ; 46(7): 644-55, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17394133

ABSTRACT

Lysyl oxidase-like 2 (LOXL2) belongs to an amine oxidase family whose members have been implicated in crosslink formation in stromal collagens and elastin, cell motility, and tumor development and progression. We previously demonstrated the association between increased LOXL2 expression and invasive/metastatic behavior in human breast cancer cells and mouse squamous and spindle cell carcinomas, interaction between LOXL2 and SNAIL in epithelial-mesenchymal transition, and localization of the LOXL2 gene to 8p21.2-21.3, within a minimally deleted region in several cancers, including colon and esophagus. In the present study, we analyzed LOXL2 expression in colon and esophageal tumors, and explored methylation as a regulator of LOXL2 expression. Immunohistochemistry using normal tissues demonstrated intracellular localization of LOXL2 in colonic enteroendocrine cells and esophageal squamous cells at the luminal surface, but not in mitotically active cells. Tissue array analysis of 52 colon adenocarcinomas and 50 esophageal squamous cell carcinomas revealed presence of LOXL2 expression in 83 and 92% of the samples, respectively, and a significant association between increased number of LOXL2-expressing cells and less-differentiated colon carcinomas. We determined that the methylation status of the 1150 bp 5' CpG island may contribute to the regulation of the gene. Loss of heterozygosity studies, using a microsatellite within intron 4 of the LOXL2 gene, revealed that loss of LOXL2 was unlikely to play a major role in either colon or esophageal tumors. These results suggest that increased LOXL2 expression in colon and esophageal cancer may contribute to tumor progression.


Subject(s)
Amino Acid Oxidoreductases/genetics , Cell Differentiation/genetics , Colonic Neoplasms/enzymology , Esophageal Neoplasms/enzymology , Adult , Aged , Aged, 80 and over , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Blotting, Western , Cell Line , Colonic Neoplasms/pathology , CpG Islands , DNA Primers , Decitabine , Esophageal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Loss of Heterozygosity , Male , Middle Aged , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
17.
Magy Onkol ; 50(2): 115-20, 2006.
Article in Hungarian | MEDLINE | ID: mdl-16888674

ABSTRACT

Syndecan-1 is a transmembrane heparan sulfate proteoglycan which plays pivotal role in cell-cell and cell-extracellular matrix interactions. However, its implication in the establishment of malignant phenotype is still controversial. Its expression indicates differentiated phenotype in certain tumors, while it confers invasive nature for others. For the better understanding of the role of syndecan-1 in cancer we transfected HT-1080 fibrosarcoma cell line with the full and a truncated construct of syndecan-1 and established stable cell lines with them. We studied the in vitro and in vivo growth capacity and metastatic potential of the transfectants in comparison with the cell line bearing only the EGFP expression vector. Our results showed that the growth rate of syndecan transfectants increased and they developed more lung metastases than the control cells. As local growth of the full transfectant was faster than that of the 78sig we presume that the full protein and maybe the shedding is needed for the local development of the tumor, but the intracellular and transmembrane domain is sufficient to promote metastasis formation.


Subject(s)
Fibrosarcoma/pathology , Membrane Glycoproteins/metabolism , Proteoglycans/metabolism , Sarcoma, Experimental/pathology , Animals , Cell Line, Tumor , Fibrosarcoma/metabolism , Immunohistochemistry , Lung Neoplasms/secondary , Mice , Neoplasm Invasiveness , Phenotype , Sarcoma, Experimental/metabolism , Syndecan-1 , Syndecans , Transfection
18.
Magy Onkol ; 48(3): 207-13, 2004.
Article in Hungarian | MEDLINE | ID: mdl-15520870

ABSTRACT

Proteoglycans are macromolecules formed by a protein core to which sugar chains are covalently attached. They are present on the cell surface and in the ECM of living things. In normal liver syndecan-1 is the dominant transmembrane proteoglycan, trace amounts of ECM proteoglycans are in the stromal components. The amounts of proteoglycans we studied increase in liver cirrhosis. In liver cancer abnormal localization of syndecan-1 and stroma rich in agrin was characteristic. The core proteins as well as the sugar chains of proteoglycans interact with and modulate the effect of regulatory factors. This implies that structural alterations of proteoglycans contribute to the development of malignant phenotype. Heparan sulfate chains of liver cancer are undersulfated with decreased or altered biological activity. Their binding capacity for transcription factor decreases, and they do not inhibit topoisomerase I enzyme. Truncated form of syndecan-1 lacking the extracellular domain of the molecule induces differentiation of hepatoma cell line and inhibits the shedding of syndecan-1. This phenomenon calls attention to the importance of syndecan-1 shedding in the regulation of cell behavior.


Subject(s)
Liver Diseases/metabolism , Liver Diseases/pathology , Liver/metabolism , Liver/pathology , Proteoglycans/metabolism , Animals , Cell Differentiation , Cell Transformation, Neoplastic , Heparitin Sulfate/metabolism , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Membrane Glycoproteins/metabolism , Signal Transduction , Syndecan-1 , Syndecans
SELECTION OF CITATIONS
SEARCH DETAIL
...