Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Oncol Rep ; 22(4): 853-61, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19724865

ABSTRACT

Ovarian cancer ranks the most lethal among gynecologic neoplasms in women. To develop potential biomarkers for diagnosis, we have identified five novel genes (CYP39A1, GTF2A1, FOXD4L4, EBP, and HAAO) that are hypermethylated in ovarian tumors, compared with the non-malignant normal ovarian surface epithelia, using the quantitative methylation-specific polymerase chain reactions. Interestingly enough, multivariate Cox regression analysis has identified hypermethylation of CYP39A1 correlated with an increase rate of relapsing (P=0.032, hazard ratio >1). Concordant hypermethylation in at least three loci was observed in 50 out of 55 (91%) of ovarian tumors examined. The test sensitivity and specificity were assessed to be 96 and 67% for CYP39A1; 95 and 88% for GTF2A1; 93 and 67% for FOXD4L4; 81 and 67% for EBP; 89 and 82% for HAAO, respectively. Our data have identified, for the first time, GTF2A1 alone, or GTF2A1 plus HAAO are excellent candidate biomarkers for detecting this disease. Moreover, the known functions of these gene products further implicate dysregulated transcriptional control, cholesterol metabolism, or synthesis of quinolinic acids, may play important roles in attributing to ovarian neoplasm. Molecular therapies, by reversing the aberrant epigenomes using inhibitory agents or by abrogating the upstream signaling pathways that convey the epigenomic perturbations, may be developed into promising treatment regimens.


Subject(s)
Biomarkers, Tumor/genetics , DNA Methylation/genetics , Epigenesis, Genetic , Ovarian Neoplasms/genetics , 3-Hydroxyanthranilate 3,4-Dioxygenase/genetics , CpG Islands , Female , Forkhead Transcription Factors/genetics , Humans , Middle Aged , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Steroid Hydroxylases/genetics , Steroid Isomerases/genetics , Transcription Factors, TFII/genetics
2.
Methods Mol Biol ; 507: 89-106, 2009.
Article in English | MEDLINE | ID: mdl-18987809

ABSTRACT

Differential methylation hybridization (DMH) is a high-throughput DNA methylation screening tool that utilizes methylation-sensitive restriction enzymes to profile methylated fragments by hybridizing them to a CpG island microarray. This array contains probes spanning all the 27,800 islands annotated in the UCSC Genome Browser. Herein we describe a revised DMH protocol with clearly identified quality control points. In this manner, samples that are unlikely to provide good readouts for differential methylation profiles between the test and the control samples will be identified and repeated with appropriate modifications. In addition to the step-by-step laboratory DMH protocol, we also provide a detailed description regarding DMH data analysis. The suggested microarray platform contains 244,000 probes and it can be a daunting barrier for researchers with no prior experience in analyzing DNA methylation data. We have created a data analysis pipeline available in a user friendly, publicly available interface, the Broad Institute's GenePattern software, which can be accessed at http://bisr.osumc.edu :8080/gp. This permits scientists to use our existing data analysis modules on their own data. As we continue to update our analysis algorithm and approaches to integrate high-throughput methylation data with other large-scale data types, we will make these new computation protocols available through the GenePattern platform.


Subject(s)
CpG Islands , DNA Fingerprinting/methods , DNA Methylation , Oligonucleotide Array Sequence Analysis/methods , Base Sequence , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/genetics , DNA/chemistry , DNA/genetics , DNA Fingerprinting/statistics & numerical data , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Data Interpretation, Statistical , Humans , Neoplasms/chemistry , Neoplasms/genetics , Nucleic Acid Amplification Techniques , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis/statistics & numerical data
3.
Methods Mol Biol ; 556: 117-39, 2009.
Article in English | MEDLINE | ID: mdl-19488875

ABSTRACT

Differential methylation hybridization (DMH) is a high-throughput DNA methylation screening tool that utilizes methylation-sensitive restriction enzymes to profile methylated fragments by hybridizing them to a CpG island microarray. This array contains probes spanning all the 27,800 islands annotated in the UCSC Genome Browser. Herein we describe a DMH protocol with clearly identified quality control points. In this manner, samples that are unlikely to provide good read-outs for differential methylation profiles between the test and the control samples will be identified and repeated with appropriate modifications. The step-by-step laboratory DMH protocol is described. In addition, we provide descriptions regarding DMH data analysis, including image quantification, background correction, and statistical procedures for both exploratory analysis and more formal inferences. Issues regarding quality control are addressed as well.


Subject(s)
DNA Methylation , DNA/analysis , Oligonucleotide Array Sequence Analysis/methods , Humans , Oligonucleotide Array Sequence Analysis/instrumentation , Reproducibility of Results
4.
BMC Bioinformatics ; 9: 453, 2008 Oct 23.
Article in English | MEDLINE | ID: mdl-18947421

ABSTRACT

BACKGROUND: Non-biological signal (or noise) has been the bane of microarray analysis. Hybridization effects related to probe-sequence composition and DNA dye-probe interactions have been observed in differential methylation hybridization (DMH) microarray experiments as well as other effects inherent to the DMH protocol. RESULTS: We suggest two models to correct for non-biologically relevant probe signal with an overarching focus on probe-sequence composition. The estimated effects are evaluated and the strengths of the models are considered in the context of DMH analyses. CONCLUSION: The majority of estimated parameters were statistically significant in all considered models. Model selection for signal correction is based on interpretation of the estimated values and their biological significance.


Subject(s)
DNA Methylation , DNA Probes , Nucleic Acid Hybridization , Sequence Analysis, DNA , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carbocyanines/metabolism , Cell Line, Tumor , Female , Fluorescent Dyes/metabolism , Gene Expression Profiling/methods , Humans , Methylation , Models, Genetic , Models, Statistical , Oligonucleotide Array Sequence Analysis , Signal Transduction
5.
Carcinogenesis ; 29(7): 1459-65, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18499701

ABSTRACT

Several studies have reported that a high expression ratio of HOXB13 to IL17BR predicts tumor recurrence in node-negative, estrogen receptor (ER) alpha-positive breast cancer patients treated with tamoxifen. The molecular mechanisms underlying this dysregulation of gene expression remain to be explored. Our epigenetic analysis has found that increased promoter methylation of one of these genes, HOXB13, correlate with the decreased expression of its transcript in breast cancer cell lines (P < 0.005). Transcriptional silencing of this gene can be reversed by a demethylation treatment. HOXB13 is suppressed by the activation of estrogen signaling in ERalpha-positive breast cancer cells. However, treatment with 4-hydroxytamoxifen (4-OHT), an antiestrogen, abrogates the ERalpha-mediated suppression in cancer cells. The notion that this transcriptional induction of HOXB13 occurs in vitro with simultaneous exposure to both estrogen and 4-OHT may provide a biological explanation for its aberrant expression in many node-negative patients undergoing tamoxifen therapy. Interestingly, promoter hypermethylation of HOXB13 is more frequently observed in ERalpha-positive patients with increased lymph node metastasis (P = 0.031) and large tumor sizes (>5 cm) (P = 0.008). In addition, this aberrant epigenetic event is associated with shorter disease-free survival (P = 0.029) in cancer patients. These results suggest that hypermethylation of HOXB13 is a late event of breast tumorigenesis and a poor prognostic indicator of node-positive cancer patients.


Subject(s)
Breast Neoplasms/genetics , DNA Methylation , Estradiol/pharmacology , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , CpG Islands , Estrogen Receptor alpha/biosynthesis , Estrogens/metabolism , Female , Gene Silencing , Homeodomain Proteins/biosynthesis , Humans , Promoter Regions, Genetic , Signal Transduction , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
6.
BMC Bioinformatics ; 8: 38, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17270052

ABSTRACT

BACKGROUND: In order to recapitulate tumor progression pathways using epigenetic data, we developed novel clustering and pathway reconstruction algorithms, collectively referred to as heritable clustering. This approach generates a progression model of altered DNA methylation from tumor tissues diagnosed at different developmental stages. The samples act as surrogates for natural progression in breast cancer and allow the algorithm to uncover distinct epigenotypes that describe the molecular events underlying this process. Furthermore, our likelihood-based clustering algorithm has great flexibility, allowing for incomplete epigenotype or clinical phenotype data and also permitting dependencies among variables. RESULTS: Using this heritable clustering approach, we analyzed methylation data obtained from 86 primary breast cancers to recapitulate pathways of breast tumor progression. Detailed annotation and interpretation are provided to the optimal pathway recapitulated. The result confirms the previous observation that aggressive tumors tend to exhibit higher levels of promoter hypermethylation. CONCLUSION: Our results indicate that the proposed heritable clustering algorithms are a useful tool for stratifying both methylation and clinical variables of breast cancer. The application to the breast tumor data illustrates that this approach can select meaningful progression models which may aid the interpretation of pathways having biological and clinical significance. Furthermore, the framework allows for other types of biological data, such as microarray gene expression or array CGH data, to be integrated.


Subject(s)
Breast Neoplasms/genetics , Cluster Analysis , DNA Methylation , Epigenesis, Genetic , Gene Expression Profiling/methods , Neoplasm Proteins/genetics , Signal Transduction , Algorithms , Breast Neoplasms/metabolism , Chromosome Mapping , DNA, Neoplasm/genetics , Evolution, Molecular , Female , Humans , Phenotype , Systems Integration
7.
BMC Genomics ; 8: 131, 2007 May 24.
Article in English | MEDLINE | ID: mdl-17524140

ABSTRACT

BACKGROUND: Previous studies of individual genes have shown that in a self-enforcing way, dimethylation at histone 3 lysine 9 (dimethyl-H3K9) and DNA methylation cooperate to maintain a repressive mode of inactive genes. Less clear is whether this cooperation is generalized in mammalian genomes, such as mouse genome. Here we use epigenomic tools to simultaneously interrogate chromatin modifications and DNA methylation in a mouse leukemia cell line, L1210. RESULTS: Histone modifications on H3K9 and DNA methylation in L1210 were profiled by both global CpG island array and custom mouse promoter array analysis. We used chromatin immunoprecipitation microarray (ChIP-chip) to examine acetyl-H3K9 and dimethyl-H3K9. We found that the relative level of acetyl-H3K9 at different chromatin positions has a wider range of distribution than that of dimethyl-H3K9. We then used differential methylation hybridization (DMH) and the restriction landmark genome scanning (RLGS) to analyze the DNA methylation status of the same targets investigated by ChIP-chip. The results of epigenomic profiling, which have been independently confirmed for individual loci, show an inverse relationship between DNA methylation and histone acetylation in regulating gene silencing. In contrast to the previous notion, dimethyl-H3K9 seems to be less distinct in specifying silencing for the genes tested. CONCLUSION: This study demonstrates in L1210 leukemia cells a diverse relationship between histone modifications and DNA methylation in the maintenance of gene silencing. Acetyl-H3K9 shows an inverse relationship between DNA methylation and histone acetylation in regulating gene silencing as expected. However, dimethyl-H3K9 seems to be less distinct in relation to promoter methylation. Meanwhile, a combination of epigenomic tools is of help in understanding the heterogeneity of epigenetic regulation, which may further our vision accumulated from single-gene studies.


Subject(s)
DNA Methylation , Gene Silencing , Histones/metabolism , Protein Processing, Post-Translational , Acetylation , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , Chromatin Immunoprecipitation , CpG Islands/genetics , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Gene Expression Regulation/drug effects , Hydroxamic Acids/pharmacology , Leukemia L1210/genetics , Leukemia L1210/metabolism , Leukemia L1210/pathology , Lysine/metabolism , Methylation , Mice , Mice, Inbred DBA , Oligonucleotide Array Sequence Analysis
8.
BMC Syst Biol ; 3: 73, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19615063

ABSTRACT

BACKGROUND: The TGF-beta/SMAD pathway is part of a broader signaling network in which crosstalk between pathways occurs. While the molecular mechanisms of TGF-beta/SMAD signaling pathway have been studied in detail, the global networks downstream of SMAD remain largely unknown. The regulatory effect of SMAD complex likely depends on transcriptional modules, in which the SMAD binding elements and partner transcription factor binding sites (SMAD modules) are present in specific context. RESULTS: To address this question and develop a computational model for SMAD modules, we simultaneously performed chromatin immunoprecipitation followed by microarray analysis (ChIP-chip) and mRNA expression profiling to identify TGF-beta/SMAD regulated and synchronously coexpressed gene sets in ovarian surface epithelium. Intersecting the ChIP-chip and gene expression data yielded 150 direct targets, of which 141 were grouped into 3 co-expressed gene sets (sustained up-regulated, transient up-regulated and down-regulated), based on their temporal changes in expression after TGF-beta activation. We developed a data-mining method driven by the Random Forest algorithm to model SMAD transcriptional modules in the target sequences. The predicted SMAD modules contain SMAD binding element and up to 2 of 7 other transcription factor binding sites (E2F, P53, LEF1, ELK1, COUPTF, PAX4 and DR1). CONCLUSION: Together, the computational results further the understanding of the interactions between SMAD and other transcription factors at specific target promoters, and provide the basis for more targeted experimental verification of the co-regulatory modules.


Subject(s)
Chromatin/metabolism , Smad Proteins/metabolism , Algorithms , Animals , Base Sequence , Cell Line , Gene Expression Profiling , Genome , Humans , Immunoprecipitation , Mice , Models, Biological , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Rats , Reproducibility of Results , Sequence Analysis, DNA , Smad Proteins/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
9.
Cancer Inform ; 6: 111-25, 2008.
Article in English | MEDLINE | ID: mdl-19259406

ABSTRACT

We present the implementation of an application using caGrid, which is the service-oriented Grid software infrastructure of the NCI cancer Biomedical Informatics Grid (caBIG), to support design and analysis of custom microarray experiments in the study of epigenetic alterations in cancer. The design and execution of these experiments requires synthesis of information from multiple data types and datasets. In our implementation, each data source is implemented as a caGrid Data Service, and analytical resources are wrapped as caGrid Analytical Services. This service-based implementation has several advantages. A backend resource can be modified or upgraded, without needing to change other components in the application. A remote resource can be added easily, since resources are not required to be collected in a centralized infrastructure.

10.
Cancer Res ; 68(24): 10257-66, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19074894

ABSTRACT

The interplay between histone modifications and promoter hypermethylation provides a causative explanation for epigenetic gene silencing in cancer. Less is known about the upstream initiators that direct this process. Here, we report that the Cystatin M (CST6) tumor suppressor gene is concurrently down-regulated with other loci in breast epithelial cells cocultured with cancer-associated fibroblasts (CAF). Promoter hypermethylation of CST6 is associated with aberrant AKT1 activation in epithelial cells, as well as the disabled INNP4B regulator resulting from the suppression by CAFs. Repressive chromatin, marked by trimethyl-H3K27 and dimethyl-H3K9, and de novo DNA methylation is established at the promoter. The findings suggest that microenvironmental stimuli are triggers in this epigenetic cascade, leading to the long-term silencing of CST6 in breast tumors. Our present findings implicate a causal mechanism defining how tumor stromal fibroblasts support neoplastic progression by manipulating the epigenome of mammary epithelial cells. The result also highlights the importance of direct cell-cell contact between epithelial cells and the surrounding fibroblasts that confer this epigenetic perturbation. Because this two-way interaction is anticipated, the described coculture system can be used to determine the effect of epithelial factors on fibroblasts in future studies.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cystatin M/genetics , Fibroblasts/pathology , Gene Silencing , Proto-Oncogene Proteins c-akt/metabolism , Breast Neoplasms/enzymology , Cell Communication/genetics , Cell Line, Tumor , Coculture Techniques , DNA Methylation , Down-Regulation , Enzyme Activation , Epithelial Cells/pathology , Female , Fibroblasts/enzymology , Gene Expression Regulation, Neoplastic , Humans , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Transfection
11.
Cancer Inform ; 3: 43-54, 2007 Feb 07.
Article in English | MEDLINE | ID: mdl-19455234

ABSTRACT

With state-of-the-art microarray technologies now available for whole genome CpG island (CGI) methylation profiling, there is a need to develop statistical models that are specifically geared toward the analysis of such data. In this article, we propose a Gamma-Normal-Gamma (GNG) mixture model for describing three groups of CGI loci: hypomethylated, undifferentiated, and hypermethylated, from a single methylation microarray. This model was applied to study the methylation signatures of three breast cancer cell lines: MCF7, T47D, and MDAMB361. Biologically interesting and interpretable results are obtained, which highlights the heterogeneity nature of the three cell lines. This underlies the premise for the need of analyzing each of the microarray slides individually as opposed to pooling them together for a single analysis. Our comparisons with the fitted densities from the Normal-Uniform (NU) mixture model in the literature proposed for gene expression analysis show an improved goodness of fit of the GNG model over the NU model. Although the GNG model was proposed in the context of single-slide methylation analysis, it can be readily adapted to analyze multi-slide methylation data as well as other types of microarray data.

SELECTION OF CITATIONS
SEARCH DETAIL