Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 50(12): 6702-6714, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35713523

ABSTRACT

The rapid transcriptional response to the transcription factor, glucocorticoid receptor (GR), including gene activation or repression, is mediated by the spatial association of genes with multiple GR binding sites (GBSs) over large genomic distances. However, only a minority of the GBSs have independent GR-mediated activating capacity, and GBSs with independent repressive activity were rarely reported. To understand the positive and negative effects of GR we mapped the regulatory environment of its gene targets. We show that the chromatin interaction networks of GR-activated and repressed genes are spatially separated and vary in the features and configuration of their GBS and other non-GBS regulatory elements. The convergence of the KLF4 pathway in GR-activated domains and the STAT6 pathway in GR-repressed domains, impose opposite transcriptional effects to GR, independent of hormone application. Moreover, the ROR and Rev-erb transcription factors serve as positive and negative regulators, respectively, of GR-mediated gene activation. We found that the spatial crosstalk between GBSs and non-GBSs provides a physical platform for sequestering the Ep300 co-activator from non-GR regulatory loci in both GR-activated and -repressed gene compartments. While this allows rapid gene repression, Ep300 recruitment to GBSs is productive specifically in the activated compartments, thus providing the basis for gene induction.


Subject(s)
E1A-Associated p300 Protein , Gene Expression Regulation , Receptors, Glucocorticoid , Receptors, Glucocorticoid/genetics , Transcriptional Activation/genetics , Cell Line, Tumor , Humans , Animals , Mice , E1A-Associated p300 Protein/metabolism
2.
Cell Rep ; 36(10): 109625, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34496233

ABSTRACT

The normal androgen receptor (AR) cistrome and transcriptional program are fundamentally altered in prostate cancer (PCa). Here, we profile the chromatin landscape and AR-directed transcriptional program in normal prostate cells and show the impact of SPOP mutations, an early event in prostate tumorigenesis. In genetically normal mouse prostate organoids, SPOP mutation results in accessibility and AR binding patterns similar to that of human PCa. Consistent with dependence on AR signaling, castration of SPOP mutant mouse models results in the loss of neoplastic phenotypes, and human SPOP mutant PCa shows a favorable response to AR-targeted therapies. Together, these data validate mouse prostate organoids as a robust model for studying epigenomic and transcriptional alterations in normal prostate, provide valuable datasets for further studies, and show that a single genomic alteration may be sufficient to reprogram the chromatin of normal prostate cells toward oncogenic phenotypes, with potential therapeutic implications for AR-targeting therapies.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation, Neoplastic/genetics , Prostate/metabolism , Prostatic Neoplasms/metabolism , Androgens/immunology , Animals , Carcinogenesis/genetics , Male , Mice, Transgenic , Prostatic Neoplasms/genetics , Receptors, Androgen/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology
3.
J Clin Invest ; 131(10)2021 05 17.
Article in English | MEDLINE | ID: mdl-33998599

ABSTRACT

BACKGROUNDMolecular characterization of prostate cancer (PCa) has revealed distinct subclasses based on underlying genomic alterations occurring early in the natural history of the disease. However, how these early alterations influence subsequent molecular events and the course of the disease over its long natural history remains unclear.METHODSWe explored the molecular and clinical progression of different genomic subtypes of PCa using distinct tumor lineage models based on human genomic and transcriptomic data. We developed transcriptional classifiers, and defined "early" and "late" categories of molecular subclasses from 8,158 PCa patients. Molecular subclasses were correlated with clinical outcomes and pathologic characteristics using Kaplan-Meier and logistic regression analyses.RESULTSWe identified PTEN and CHD1 alterations as subtype-specific late progression events specifically in ERG-overexpressing (ERG+) and SPOP-mutant tumors, respectively, and 2 distinct progression models consisting of ERG/PTEN (normal to ERG+ to PTEN-deleted) and SPOP/CHD1 (normal to SPOP-mutated to CHD1-deleted) with shared early tumorigenesis but distinct pathways toward progression. We found that within ERG+ and SPOP-mutant subtypes, late events were associated with worse prognosis. Importantly, the clinical and pathologic features associated with distinct late events at radical prostatectomy were strikingly different; PTEN deletions were associated with increased locoregional stage, while CHD1 deletions were only associated with increased grade, despite equivalent metastatic potential.CONCLUSIONThese findings suggest a paradigm in which specific subtypes of PCa follow distinct pathways of progression, at both the molecular and clinical levels. Therefore, the interpretation of common clinical parameters such as locoregional tumor stage may be influenced by the underlying tumor lineage, and potentially influence management decisions.FUNDINGProstate Cancer Foundation, National Cancer Institute, Urology Care Foundation, Damon Runyon Cancer Research Foundation, US Department of Defense, and the AIRC Foundation.


Subject(s)
Biomarkers, Tumor , Databases, Nucleic Acid , Neoplasm Proteins , Prostatic Neoplasms , RNA-Seq , Registries , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Disease-Free Survival , Humans , Male , Middle Aged , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Prostatic Neoplasms/classification , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/mortality , Retrospective Studies , Survival Rate
4.
Sci Rep ; 8(1): 14012, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30228286

ABSTRACT

Recently, functional connections between S-adenosylhomocysteine hydrolase (AHCY) activity and cancer have been reported. As the properties of AHCY include the hydrolysis of S-adenosylhomocysteine and maintenance of the cellular methylation potential, the connection between AHCY and cancer is not obvious. The mechanisms by which AHCY influences the cell cycle or cell proliferation have not yet been confirmed. To elucidate AHCY-driven cancer-specific mechanisms, we pursued a multi-omics approach to investigate the effect of AHCY-knockdown on hepatocellular carcinoma cells. Here, we show that reduced AHCY activity causes adenosine depletion with activation of the DNA damage response (DDR), leading to cell cycle arrest, a decreased proliferation rate and DNA damage. The underlying mechanism behind these effects might be applicable to cancer types that have either significant levels of endogenous AHCY and/or are dependent on high concentrations of adenosine in their microenvironments. Thus, adenosine monitoring might be used as a preventive measure in liver disease, whereas induced adenosine depletion might be the desired approach for provoking the DDR in diagnosed cancer, thus opening new avenues for targeted therapy. Additionally, including AHCY in mutational screens as a potential risk factor may be a beneficial preventive measure.


Subject(s)
Adenosine/deficiency , Adenosylhomocysteinase/antagonists & inhibitors , Biomarkers, Tumor/analysis , Carcinoma, Hepatocellular/pathology , Cell Cycle Checkpoints , DNA Damage , Liver Neoplasms/pathology , Adenosylhomocysteinase/genetics , Adenosylhomocysteinase/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mutation , Proteome , RNA, Small Interfering/genetics , Transcriptome , Tumor Cells, Cultured
5.
J Vis Exp ; (129)2017 11 13.
Article in English | MEDLINE | ID: mdl-29155775

ABSTRACT

Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC-seq) is a method used for the identification of open (accessible) regions of chromatin. These regions represent regulatory DNA elements (e.g., promoters, enhancers, locus control regions, insulators) to which transcription factors bind. Mapping the accessible chromatin landscape is a powerful approach for uncovering active regulatory elements across the genome. This information serves as an unbiased approach for discovering the network of relevant transcription factors and mechanisms of chromatin structure that govern gene expression programs. ATAC-seq is a robust and sensitive alternative to DNase I hypersensitivity analysis coupled with next-generation sequencing (DNase-seq) and formaldehyde-assisted isolation of regulatory elements (FAIRE-seq) for genome-wide analysis of chromatin accessibility and to the sequencing of micrococcal nuclease-sensitive sites (MNase-seq) to determine nucleosome positioning. We present a detailed ATAC-seq protocol optimized for human primary immune cells i.e. CD4+ lymphocytes (T helper 1 (Th1) and Th2 cells). This comprehensive protocol begins with cell harvest, then describes the molecular procedure of chromatin tagmentation, sample preparation for next-generation sequencing, and also includes methods and considerations for the computational analyses used to interpret the results. Moreover, to save time and money, we introduced quality control measures to assess the ATAC-seq library prior to sequencing. Importantly, the principles presented in this protocol allow its adaptation to other human immune and non-immune primary cells and cell lines. These guidelines will also be useful for laboratories which are not proficient with next-generation sequencing methods.


Subject(s)
Chromatin/metabolism , Chromosome Mapping/methods , DNA/genetics , Sequence Analysis, DNA/methods , T-Lymphocytes/physiology , High-Throughput Nucleotide Sequencing/methods , Humans
6.
Eur J Cell Biol ; 96(6): 579-590, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28647132

ABSTRACT

S-adenosylhomocysteine hydrolase (AHCY) is thought to be located at the sites of ongoing AdoMet-dependent methylation, presumably in the cell nucleus. Endogenous AHCY is located both in cytoplasm and the nucleus. Little is known regarding mechanisms that drive its subcellular distribution, and even less is known on how mutations causing AHCY deficiency affect its intracellular dynamics. Using fluorescence microscopy and GFP-tagged AHCY constructs we show significant differences in the intensity ratio between nuclei and cytoplasm for mutant proteins when compared with wild type AHCY. Interestingly, nuclear export of AHCY is not affected by leptomycin B. Systematic deletions showed that AHCY has two regions, located at both sides of the protein, that contribute to its nuclear localization, implying the interaction with various proteins. In order to evaluate protein interactions in vivo we engaged in bimolecular fluorescence complementation (BiFC) based studies. We investigated previously assumed interaction with AHCY-like-1 protein (AHCYL1), a paralog of AHCY. Indeed, significant interaction between both proteins exists. Additionally, silencing AHCYL1 leads to moderate inhibition of nuclear export of endogenous AHCY.


Subject(s)
Adenosylhomocysteinase/genetics , Adenosylhomocysteinase/metabolism , Protein Interaction Maps/genetics , Active Transport, Cell Nucleus/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , Fatty Acids, Unsaturated/pharmacology , Gene Deletion , Humans , Microscopy, Fluorescence , Mutation , Protein Binding
7.
Protoplasma ; 254(3): 1175-1185, 2017 May.
Article in English | MEDLINE | ID: mdl-28013411

ABSTRACT

Glucocorticoids and their receptor (GR) have been an important area of research because of their pleiotropic physiological functions and extensive use in the clinic. In addition, the association between GR and glucocorticoids, which is highly specific, leads to rapid nuclear translocation where GR associates with chromatin to regulate gene transcription. This simplified model system has been instrumental for studying the complexity of transcription regulation processes occurring at chromatin. In this review we discuss our current understanding of GR action that has been enhanced by recent developments in genome wide measurements of chromatin accessibility, histone marks, chromatin remodeling and 3D chromatin structure in various cell types responding to glucocorticoids.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Chromatin/physiology , Gene Expression Regulation/physiology , Glucocorticoids/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Binding Sites , Humans , Mice , Promoter Regions, Genetic , Protein Binding , Transcription, Genetic
8.
BioData Min ; 9: 26, 2016.
Article in English | MEDLINE | ID: mdl-27547241

ABSTRACT

BACKGROUND: Mass spectrometry (MS) are a group of a high-throughput techniques used to increase knowledge about biomolecules. They produce a large amount of data which is presented as a list of hundreds or thousands of proteins. Filtering those data efficiently is the first step for extracting biologically relevant information. The filtering may increase interest by merging previous data with the data obtained from public databases, resulting in an accurate list of proteins which meet the predetermined conditions. RESULTS: In this article we present msBiodat Analysis Tool, a web-based application thought to approach proteomics to the big data analysis. With this tool, researchers can easily select the most relevant information from their MS experiments using an easy-to-use web interface. An interesting feature of msBiodat analysis tool is the possibility of selecting proteins by its annotation on Gene Ontology using its Gene Id, ensembl or UniProt codes. CONCLUSION: The msBiodat analysis tool is a web-based application that allows researchers with any programming experience to deal with efficient database querying advantages. Its versatility and user-friendly interface makes easy to perform fast and accurate data screening by using complex queries. Once the analysis is finished, the result is delivered by e-mail. msBiodat analysis tool is freely available at http://msbiodata.irb.hr.

9.
Mar Drugs ; 13(7): 4179-96, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26198235

ABSTRACT

Finkel-Biskis-Reilly murine sarcoma virus (FBR-MuSV) ubiquitously expressed (FAU) gene is down-regulated in human prostate, breast and ovarian cancers. Moreover, its dysregulation is associated with poor prognosis in breast cancer. Sponges (Porifera) are animals without tissues which branched off first from the common ancestor of all metazoans. A large majority of genes implicated in human cancers have their homologues in the sponge genome. Our study suggests that FAU gene from the sponge Suberites domuncula reflects characteristics of the FAU gene from the metazoan ancestor, which have changed only slightly during the course of animal evolution. We found pro-apoptotic activity of sponge FAU protein. The same as its human homologue, sponge FAU increases apoptosis in human HEK293T cells. This indicates that the biological functions of FAU, usually associated with "higher" metazoans, particularly in cancer etiology, possess a biochemical background established early in metazoan evolution. The ancestor of all animals possibly possessed FAU protein with the structure and function similar to evolutionarily more recent versions of the protein, even before the appearance of true tissues and the origin of tumors and metastasis. It provides an opportunity to use pre-bilaterian animals as a simpler model for studying complex interactions in human cancerogenesis.


Subject(s)
Ribosomal Proteins/isolation & purification , Suberites/genetics , Animals , Apoptosis/drug effects , Biological Evolution , DNA/genetics , DNA/isolation & purification , HEK293 Cells/drug effects , HeLa Cells/drug effects , Humans , RNA, Small Untranslated/genetics , RNA, Small Untranslated/isolation & purification , Ribosomal Proteins/genetics , Ribosomal Proteins/pharmacology , Sequence Alignment , Subcellular Fractions/chemistry , Suberites/chemistry
10.
PLoS One ; 10(4): e0124670, 2015.
Article in English | MEDLINE | ID: mdl-25915617

ABSTRACT

BACKGROUND: Sirtuin 1 (SIRT1) and sirtuin 2 (SIRT2) are NAD+-dependent protein deacetylases involved in the regulation of key cancer-associated genes. In this study we evaluated the relevance of these deacetylases in lung cancer biology. MATERIAL AND METHODS: Protein levels of SIRT1 and SIRT2 were determined in non-small cell lung cancer (NSCLC) cell lines and primary tumors from 105 patients. Changes in proliferation were assessed after SIRT1 and SIRT2 downregulation in lung cancer cell lines using siRNA-mediated technology or tenovin-1, a SIRT1 and SIRT2 inhibitor. RESULTS: High SIRT1 and SIRT2 protein levels were found in NSCLC cell lines compared with non-tumor lung epithelial cells. The expression of SIRT1 and SIRT2 proteins was also significantly higher in lung primary tumors than in normal tissue (P<0.001 for both sirtuins). Stronger nuclear SIRT1 staining was observed in adenocarcinomas than in squamous cell carcinomas (P=0.033). Interestingly, in NSCLC patients, high SIRT1 and SIRT2 expression levels were associated with shorter recurrence-free survival (P=0.04 and P=0.007, respectively). Moreover, the combination of high SIRT1 and SIRT2 expression was an independent prognostic factor for shorter recurrence-free survival (P=0.002) and overall survival (P=0.022). In vitro studies showed that SIRT1 and/or SIRT2 downregulation significantly decreased proliferation of NSCLC. CONCLUSIONS: Our results support the hypothesis that SIRT1 and SIRT2 have a protumorigenic role in lung cancer, promoting cell proliferation. Moreover, the expression of these proteins is associated with poor prognosis in NSCLC patients and may help to identify those NSCLC patients with high risk of recurrence that could benefit from adjuvant therapy after resection.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Gene Expression , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Sirtuin 1/genetics , Sirtuin 2/genetics , Acetanilides/pharmacology , Aged , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , RNA Interference , RNA, Small Interfering/genetics , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/metabolism , Sirtuin 2/antagonists & inhibitors , Sirtuin 2/metabolism , Thiourea/analogs & derivatives , Thiourea/pharmacology , Tumor Suppressor Protein p53/metabolism
11.
Pathol Oncol Res ; 20(3): 687-95, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24563233

ABSTRACT

BORIS is a paralog of a highly conserved, multi-functional chromatin factor CTCF. Unlike CTCF, which has been shown to possess tumor-suppressive properties, BORIS belongs to the "cancer/testis antigen" family normally expressed only in germ cells and aberrantly activated in a variety of tumors. The consequences of BORIS expression, relative abundance of its isoforms, and its role in carcinogenesis have not been completely elucidated. It activates transcription of hTERT and MYC, genes relevant for laryngeal carcinoma progression. In this study, BORIS expression has been analyzed at the transcriptional level by RT-PCR and protein level by semi-quantitative immunohistochemistry in 32 laryngeal squamous cell carcinomas and adjacent non-tumorous tissue. BORIS was detected in 44 % (14/32) laryngeal squamous cell carcinoma samples, while it was detected only in one normal, tumor-adjacent tissue sample. Tree based survival analysis, using the recursive partitioning algorithm mvpart, extracted the ratio of relative abundance of BORIS transcript variants containing exon 7 (BORIS 7+) and those lacking exon 7 (BORIS 7-) as an independent prognostic factor associated with disease relapse during a 5-year follow-up period. Patients having BORIS 7+/BORIS 7- ratio ≥1 had a higher rate of disease relapse than patients with BORIS 7+/BORIS 7- ratio <1. Hazard ratio for that group, based on Cox Proportional Hazard Regression, was 3.53. This is the first study analyzing expression of BORIS protein and transcript variants in laryngeal squamous cell carcinoma relative to its possible prognostic value for recurrence and overall survival.


Subject(s)
Alternative Splicing/genetics , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , DNA-Binding Proteins/genetics , Laryngeal Neoplasms/genetics , Neoplasm Recurrence, Local/genetics , Algorithms , Base Sequence , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , DNA-Binding Proteins/metabolism , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Laryngeal Neoplasms/mortality , Laryngeal Neoplasms/pathology , Larynx/metabolism , Male , Middle Aged , Molecular Sequence Data , Neoplasm Grading , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Pilot Projects , Prognosis , Protein Isoforms , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate
12.
Acta Biochim Pol ; 57(2): 185-91, 2010.
Article in English | MEDLINE | ID: mdl-20512166

ABSTRACT

Liver is a unique mammalian organ with a great capacity of regeneration related to its function. After surgical resection or injury, hepatic cells, especially hepatocytes, can proliferate rapidly to repair the damage and to regenerate the structure without affecting the function of the liver. Loss of catalase activity during regeneration indicates that oxidative stress is present in the liver not only in pathological conditions but also as a 'physiological' factor during regeneration. As we have shown in our previous work, liver stem cell-like cells treated with 4-hydroxynonenal (HNE), a cytotoxic and growth regulating lipid peroxidation product, recover in the presence of spleen cells. In the current study we characterized this novel cell line as liver-derived progenitor/oval-like cells, (LDP/OCs), i.e. functional liver stem-like cells. We showed that LDP/OC were OV6 positive, with abundant glycogen content in the cytoplasm and expressed alpha-fetoprotein, albumin, biliverdin reductase and gamma-glutamyl transferase. Also, we compared their growth in vitro with the growth of cultured primary hepatocytes stressed with HNE and co-cultured with autologous spleen cells. The influence of spleen cells on HNE-treated primary hepatocytes and on LDP/OCs showed that spleen cells support in a similar manner the recovery of both types of liver cells indicating their important role in regeneration. Hence, LDP/OC cells may provide a valuable tool to study cell interactions and the role on HNE in liver regeneration.


Subject(s)
Aldehydes/pharmacology , Hepatocytes/drug effects , Liver/chemistry , Liver/cytology , Regeneration/physiology , Spleen/cytology , Stem Cells/drug effects , Cell Line , Cell Survival , Cells, Cultured , Hepatocytes/ultrastructure , Liver/drug effects , Reverse Transcriptase Polymerase Chain Reaction
13.
J Mol Med (Berl) ; 86(9): 1057-66, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18604514

ABSTRACT

Imprinting analyses of IGF2 and H19, loss of heterozygosity (LOH) analyses of IGF2R and CTCF and Helicobacter pylori detection, were performed on 35 human laryngeal squamous cell carcinomas (LSCC). Forty-six percent of the tumors were heterozygous for IGF2, and 54% were informative for the H19. Biallelic expression of IGF2 was observed in 33% (5 out of 15) of the tumors and in 27% (4 out of 15) of adjacent non-tumorous laryngeal tissues. H19 loss of imprinting (LOI) was observed in 24% (4 out of 17) of the tumors. For IGF2R and CTCF, 71% (25 out of 35) and 50% (17/34), respectively, of the samples were heterozygous, and LOH was detected in 12% (3 out of 25) and 6% (1 out of 17), respectively, of the tumors. H. pylori was found in 26% (9/35) of these tumors. Among them, four were informative for the imprinting analysis. The presence of H. pylori had no effect on IGF2/H19 imprinting. Only the H. pylori detection was further broadened with an additional 47 laryngeal tumors, resulting in a total final positivity of close to 16% (13 out of 82). This study represents the largest comprehensive IGF2/H19 imprinting study done to date on well-defined samples of human laryngeal carcinomas and corresponding non-tumorous tissue. For the first time, the analyses of IGF2/H19 imprinting have been broadened with LOH analyses of IGF2R and CTCF, with both of these genes acting as modulators of IGF2 and H19 activity. Although there were indications that H. pylori may be present in LSCC, we are the first to show its presence in LSCC by two direct techniques: Giemsa staining and nested-PCR.


Subject(s)
Carcinoma, Squamous Cell , DNA-Binding Proteins/genetics , Genomic Imprinting , Helicobacter Infections , Insulin-Like Growth Factor II/genetics , Laryngeal Neoplasms , RNA, Untranslated/genetics , Receptor, IGF Type 2/genetics , Repressor Proteins/genetics , CCCTC-Binding Factor , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/pathology , DNA-Binding Proteins/metabolism , Helicobacter pylori/metabolism , Helicobacter pylori/pathogenicity , Humans , Insulin-Like Growth Factor II/metabolism , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/microbiology , Laryngeal Neoplasms/pathology , Loss of Heterozygosity , RNA, Long Noncoding , RNA, Untranslated/metabolism , Receptor, IGF Type 2/metabolism , Repressor Proteins/metabolism
14.
J Cell Biochem ; 104(5): 1781-92, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18348204

ABSTRACT

Curcumin (diferuloymethane), a natural compound used in traditional medicine, exerts an antiproliferative effect on various tumor cell lines by an incompletely understood mechanism. It has been shown that low doses of curcumin downregulate DNA topoisomerase II alpha (TOP2A) which is upregulated in many malignances. The activity of TOP2A is required for RNA polymerase II transcription on chromatin templates. Recently, it has been reported that CTCF, a multifunctional transcription factor, recruits the largest subunit of RNA polymerase II (LS Pol II) to its target sites genome-wide. This recruitment of LS Pol II is more pronounced in proliferating cells than in fully differentiated cells. As expression of imprinted genes is often altered in tumors, we investigated the potential effect of curcumin treatment on transcription of the imprinted H19 gene, located distally from the CTCF binding site, in human tumor cell lines HCT 116, SW 620, HeLa, Cal 27, Hep-2 and Detroit 562. Transcription of TOP2A and concomitantly H19 was supressed in all tumor cell lines tested. Monoallelic IGF2 expression was maintained in curcumin-treated cancer cells, indicating the involvement of mechanism/s other than disturbance of CTCF insulator function at the IGF2/H19 locus. Curcumin did not alter H19 gene transcription in primary cell cultures derived from normal human tissues.


Subject(s)
Curcumin/pharmacology , Down-Regulation/drug effects , RNA, Untranslated/genetics , Transcription, Genetic/drug effects , Alleles , Antigens, Neoplasm/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cyclooxygenase 2/genetics , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Insulin-Like Growth Factor II/genetics , NAD/metabolism , Poly-ADP-Ribose Binding Proteins , RNA, Long Noncoding
15.
Cancer Lett ; 238(2): 224-9, 2006 Jul 18.
Article in English | MEDLINE | ID: mdl-16111804

ABSTRACT

The gene for insulin-like growth factor two, IGF2 is maternally imprinted. Fifteen heterozygous samples were analyzed for the IGF2 imprinting status and promoter usage. IGF2 LOI was detected in four non-tumorous tissues and in six laryngeal squamous cell carcinoma (LSCC) tumors. There was no clear pattern of specific promoter activity in LSCC tumors and the adjacent normal tissues. P1 promoter usage was active in eight LSCCs, among them four with LOI. As it was activated in four tumors with maintenance of imprinting (MOI) and four non-tumors, we concluded that P1 promoter is not exclusively connected with IGF2 LOI in LSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Genomic Imprinting , Insulin-Like Growth Factor II/genetics , Laryngeal Neoplasms/genetics , Promoter Regions, Genetic , Aged , Aged, 80 and over , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...