Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 71(8): 3087-97, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21363915

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is strongly associated with cancer progression, but its potential role during premalignant development has not been studied. Here, we show that a 4-week exposure of immortalized human bronchial epithelial cells (HBEC) to tobacco carcinogens can induce a persistent, irreversible, and multifaceted dedifferentiation program marked by EMT and the emergence of stem cell-like properties. EMT induction was epigenetically driven, initially by chromatin remodeling through H3K27me3 enrichment and later by ensuing DNA methylation to sustain silencing of tumor-suppressive microRNAs (miRNA), miR-200b, miR-200c, and miR-205, which were implicated in the dedifferentiation program in HBECs and also in primary lung tumors. Carcinogen-treated HBECs acquired stem cell-like features characterized by their ability to form spheroids with branching tubules and enrichment of the CD44(high)/CD24(low), CD133, and ALDH1 stem cell-like markers. miRNA overexpression studies indicated that regulation of the EMT, stem-like, and transformed phenotypes in HBECs were distinct events. Our findings extend present concepts of how EMT participates in cancer pathophysiology by showing that EMT induction can participate in cancer initiation to promote the clonal expansion of premalignant lung epithelial cells.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MicroRNAs/genetics , Carcinogens , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gene Silencing , Humans , Lung Neoplasms/chemically induced , Neoplastic Stem Cells/pathology , Nicotiana/chemistry
2.
Carcinogenesis ; 30(12): 2023-30, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19917631

ABSTRACT

Death-associated protein kinase (DAPK), a mediator of apoptotic systems, is silenced by promoter hypermethylation in lung and breast tumors. This gene has a CpG island extending 2500 bp from the translational start site; however, studies characterizing its transcriptional regulation have not been conducted. Two transcripts for DAPK were identified that code for a single protein, while being regulated by two promoters. The previously identified DAPK transcript designated as exon 1 transcript was expressed at levels 3-fold greater than the alternate exon 1b transcript. Deletion constructs of promoter 1 identified a 332 bp region containing a functional CP2-binding site important for expression of the exon 1 transcript. While moderate reporter activity was seen in promoter 2, the region comprising intron 1 and containing a HNF3B-binding site sustained expression of the alternate transcript. Sequencing the DAPK CpG island in tumor cell lines revealed dense, but heterogenous methylation of CpGs that blocked access of the CP2 and HNF3B proteins that in turn, was associated with loss of transcription that was restored by treatment with 5-aza-2'-deoxycytidine. Prevalences were similar for methylation of promoter 1 and 2 and intron 1 in lung tumors, but significantly greater in promoter 2 and intron 1 in breast tumors, indicative of tissue-specific differences in silencing these two transcripts. These studies show for the first time dual promoter regulation of DAPK, a tumor suppressor gene silenced in many cancers, and substantiate the importance of screening for silencing of both transcripts in tumors.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA Methylation , Gene Expression Regulation, Neoplastic , Gene Silencing , Neoplasms/genetics , Neoplasms/metabolism , Promoter Regions, Genetic , Apoptosis , Base Sequence , Binding Sites , Cell Line, Tumor , CpG Islands , Death-Associated Protein Kinases , Gene Deletion , Humans , Molecular Sequence Data , RNA, Messenger/metabolism
3.
Carcinogenesis ; 30(12): 2095-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19861651

ABSTRACT

There is a critical need to identify efficacious chemopreventive agents for lung cancer that can be taken chronically with no side effects and whose mechanisms of action do not involve genotoxicity that could drive, rather than impede, cancer progression. We evaluated the ability of a chemopreventive cocktail that included selenium (antioxidant), rosiglitazone (peroxisome proliferator-activated receptor gamma agonist), sodium phenylbutyrate or valproic acid (histone deacetylase inhibitors) and hydralazine (cytosine-demethylating agent) to prevent the progression of lung cancer in A/J mice treated with NNK. Agents were administered alone or in various combinations. Effects of the chemopreventive agents were quantified based on the proportion of hyperplasias and adenomas within the mouse lung. Significant effects on tumor progression were seen in all treatment groups that included rosiglitazone as reflected by a 47-57% increase in number of hyperplasias and a 10-30% decrease in adenomas. Cell proliferation was also reduced in these treatment groups by approximately 40%. Interestingly, while treatment with rosiglitazone alone did not significantly affect lesion size, striking effects were seen in the combination therapy group that included sodium phenylbutyrate, with the volume of hyperplasias and adenomas decreasing by 40 and 77%, respectively. These studies demonstrate for the first time that chronic in vivo administration of rosiglitazone, used in the management of diabetes mellitus, can significantly block the progression of premalignant lung cancer in the A/J mouse model.


Subject(s)
Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Thiazolidinediones/pharmacology , Adenoma/drug therapy , Adenoma/pathology , Animals , Antioxidants/metabolism , Cell Line, Tumor , Disease Progression , Female , Histone Deacetylases/metabolism , Humans , Hypoglycemic Agents/pharmacology , Immunohistochemistry/methods , Lung Neoplasms/metabolism , Mice , Neoplasm Invasiveness , PPAR gamma/metabolism , Rosiglitazone
4.
Dose Response ; 7(2): 104-31, 2009 Jun 11.
Article in English | MEDLINE | ID: mdl-19543479

ABSTRACT

Humans are continuously exposed to low-level ionizing radiation from natural sources. However, harsher radiation environments persisted during our planet's early years and mammals survived via an evolutionary gift--a system of radiation-induced natural protective measures (adaptive protection). This system includes antioxidants, DNA repair, apoptosis of severely damaged cells, epigenetically regulated apoptosis (epiapoptosis) pathways that selectively remove precancerous and other aberrant cells, and immunity against cancer. We propose a novel model in which the protective system is regulated at least in part via radiation-stress-stimulated epigenetic reprogramming (epireprogramming) of adaptive-response genes. High-dose radiation can promote epigenetically silencing of adaptive-response genes (episilencing), for example via promoter-associated DNA and/or histone methylation and/or histone deacetylation. Evidence is provided for low linear-energy-transfer (LET) radiation-activated natural protection (ANP) against high-LET alpha-radiation-induced lung cancer in plutonium-239 exposed rats and radon-progeny-exposed humans. Using a revised hormetic relative risk model for cancer induction that accounts for both epigenetic activation (epiactivation) and episilencing of genes, we demonstrate that, on average, >80% of alpha-radiation-induced rat lung cancers were prevented by chronic, low-rate gamma-ray ANP. Interestingly, lifetime exposure to residential radon at the Environmental Protection Agency's action level of 4 pCi L(-1) appears to be associated with on average a > 60% reduction in lung cancer cases, rather than an increase. We have used underlined italics to indicate newly introduced terminology.

5.
Cancer Res ; 68(21): 9005-14, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18974146

ABSTRACT

A better understanding of key molecular changes during transformation of lung epithelial cells could affect strategies to reduce mortality from lung cancer. This study uses an in vitro model to identify key molecular changes that drive cell transformation and the likely clonal outgrowth of preneoplastic lung epithelial cells that occurs in the chronic smoker. Here, we show differences in transformation efficiency associated with DNA repair capacity for two hTERT/cyclin-dependent kinase 4, immortalized bronchial epithelial cell lines after low-dose treatment with the carcinogens methylnitrosourea, benzo(a)pyrene-diolepoxide 1, or both for 12 weeks. Levels of cytosine-DNA methyltransferase 1 (DNMT1) protein increased significantly during carcinogen exposure and were associated with the detection of promoter hypermethylation of 5 to 10 genes in each transformed cell line. Multiple members of the cadherin gene family were commonly methylated during transformation. Stable knockdown of DNMT1 reversed transformation and gene silencing. Moreover, stable knockdown of DNMT1 protein before carcinogen treatment prevented transformation and methylation of cadherin genes. These studies provide a mechanistic link between increased DNMT1 protein, de novo methylation of tumor suppressor genes, and reduced DNA repair capacity that together seem causal for transformation of lung epithelial cells. This finding supports the development of demethylation strategies for primary prevention of lung cancer in smokers.


Subject(s)
Bronchi/metabolism , Carcinogens/toxicity , Cell Transformation, Neoplastic , DNA (Cytosine-5-)-Methyltransferases/physiology , DNA Methylation , Promoter Regions, Genetic , Bronchi/cytology , Chromatin Immunoprecipitation , DNA (Cytosine-5-)-Methyltransferase 1 , Epithelial Cells/metabolism , Humans , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...