Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Genomics ; 111(1): 17-23, 2019 01.
Article in English | MEDLINE | ID: mdl-27453286

ABSTRACT

To develop accurate prognostic models is one of the biggest challenges in "omics"-based cancer research. Here, we propose a novel computational method for identifying dysregulated gene subnetworks as biomarkers to predict cancer recurrence. Applying our method to the DNA methylome of endometrial cancer patients, we identified a subnetwork consisting of differentially methylated (DM) genes, and non-differentially methylated genes, termed Epigenetic Connectors (EC), that are topologically important for connecting the DM genes in a protein-protein interaction network. The ECs are statistically significantly enriched in well-known tumorgenesis and metastasis pathways, and include known epigenetic regulators. Importantly, combining the DMs and ECs as features using a novel random walk procedure, we constructed a support vector machine classifier that significantly improved the prediction accuracy of cancer recurrence and outperformed several alternative methods, demonstrating the effectiveness of our network-based approach.


Subject(s)
Algorithms , Biomarkers, Tumor , DNA Methylation , Endometrial Neoplasms , Neoplasm Recurrence, Local , CpG Islands , DNA, Neoplasm , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Epigenomics , Female , Gene Expression Profiling , Gene Regulatory Networks , High-Throughput Nucleotide Sequencing , Humans , Models, Genetic , Prognosis , Protein Interaction Domains and Motifs , Sequence Analysis, DNA
2.
Nat Commun ; 7: 10913, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26941120

ABSTRACT

The breast cancer susceptibility gene BRCA1 is well known for its function in double-strand break (DSB) DNA repair. While BRCA1 is also implicated in transcriptional regulation, the physiological significance remains unclear. COBRA1 (also known as NELF-B) is a BRCA1-binding protein that regulates RNA polymerase II (RNAPII) pausing and transcription elongation. Here we interrogate functional interaction between BRCA1 and COBRA1 during mouse mammary gland development. Tissue-specific deletion of Cobra1 reduces mammary epithelial compartments and blocks ductal morphogenesis, alveologenesis and lactogenesis, demonstrating a pivotal role of COBRA1 in adult tissue development. Remarkably, these developmental deficiencies due to Cobra1 knockout are largely rescued by additional loss of full-length Brca1. Furthermore, Brca1/Cobra1 double knockout restores developmental transcription at puberty, alters luminal epithelial homoeostasis, yet remains deficient in homologous recombination-based DSB repair. Thus our genetic suppression analysis uncovers a previously unappreciated, DNA repair-independent function of BRCA1 in antagonizing COBRA1-dependent transcription programme during mammary gland development.


Subject(s)
DNA Repair/physiology , Mammary Glands, Animal/growth & development , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Aging , Animals , BRCA1 Protein , DNA Breaks, Double-Stranded , Epithelial Cells , Estrogens/metabolism , Female , Gene Expression Regulation, Developmental/physiology , Homeostasis , Mice , Mice, Knockout , Nuclear Proteins/genetics , Progestins/metabolism , RNA-Binding Proteins , Sexual Maturation , Transcriptome , Tumor Suppressor Proteins/genetics
3.
Bioinformatics ; 30(13): 1858-66, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24618465

ABSTRACT

MOTIVATION: Metastasis prediction is a well-known problem in breast cancer research. As breast cancer is a complex and heterogeneous disease with many molecular subtypes, predictive models trained for one cohort often perform poorly on other cohorts, and a combined model may be suboptimal for individual patients. Furthermore, attempting to develop subtype-specific models is hindered by the ambiguity and stereotypical definitions of subtypes. RESULTS: Here, we propose a personalized approach by relaxing the definition of breast cancer subtypes. We assume that each patient belongs to a distinct subtype, defined implicitly by a set of patients with similar molecular characteristics, and construct a different predictive model for each patient, using as training data, only the patients defining the subtype. To increase robustness, we also develop a committee-based prediction method by pooling together multiple personalized models. Using both intra- and inter-dataset validations, we show that our approach can significantly improve the prediction accuracy of breast cancer metastasis compared with several popular approaches, especially on those hard-to-learn cases. Furthermore, we find that breast cancer patients belonging to different canonical subtypes tend to have different predictive models and gene signatures, suggesting that metastasis in different canonical subtypes are likely governed by different molecular mechanisms. AVAILABILITY AND IMPLEMENTATION: Source code implemented in MATLAB and Java available at www.cs.utsa.edu/∼jruan/PCC/.


Subject(s)
Breast Neoplasms/pathology , Algorithms , Cluster Analysis , Datasets as Topic , Humans , Neoplasm Metastasis
4.
Nat Commun ; 4: 1821, 2013.
Article in English | MEDLINE | ID: mdl-23652009

ABSTRACT

Adipose stromal cells are the primary source of local oestrogens in adipose tissue, aberrant production of which promotes oestrogen receptor-positive breast cancer. Here we show that extracellular matrix compliance and cell contractility are two opposing determinants for oestrogen output of adipose stromal cells. Using synthetic extracellular matrix and elastomeric micropost arrays with tunable rigidity, we find that increasing matrix compliance induces transcription of aromatase, a rate-limiting enzyme in oestrogen biosynthesis. This mechanical cue is transduced sequentially by discoidin domain receptor 1, c-Jun N-terminal kinase 1, and phosphorylated JunB, which binds to and activates two breast cancer-associated aromatase promoters. In contrast, elevated cell contractility due to actin stress fibre formation dampens aromatase transcription. Mechanically stimulated stromal oestrogen production enhances oestrogen-dependent transcription in oestrogen receptor-positive tumour cells and promotes their growth. This novel mechanotransduction pathway underlies communications between extracellular matrix, stromal hormone output, and cancer cell growth within the same microenvironment.


Subject(s)
Adipose Tissue/metabolism , Estrogens/metabolism , Stress, Mechanical , Adipose Tissue/cytology , Adipose Tissue/drug effects , Animals , Aromatase/genetics , Aromatase/metabolism , Biomechanical Phenomena/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cattle , Cell Culture Techniques , Collagen/metabolism , Culture Media, Conditioned/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Discoidin Domain Receptor 1 , Estrogens/biosynthesis , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Female , Gene Knockdown Techniques , Humans , Integrin beta1/metabolism , MAP Kinase Signaling System , Mechanotransduction, Cellular/drug effects , Models, Biological , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Estrogen/metabolism , Stromal Cells/drug effects , Stromal Cells/enzymology , Transcription, Genetic/drug effects
5.
Article in English | MEDLINE | ID: mdl-25327524

ABSTRACT

In silico prediction of drug side-effects in early stage of drug development is becoming more popular now days, which not only reduces the time for drug design but also reduces the drug development costs. In this article we propose an ensemble approach to predict drug side-effects of drug molecules based on their chemical structure. Our idea originates from the observation that similar drugs have similar side-effects. Based on this observation we design an ensemble approach that combine the results from different classification models where each model is generated by a different set of similar drugs. We applied our approach to 1385 side-effects in the SIDER database for 888 drugs. Results show that our approach outperformed previously published approaches and standard classifiers. Furthermore, we applied our method to a number of uncharacterized drug molecules in DrugBank database and predict their side-effect profiles for future usage. Results from various sources confirm that our method is able to predict the side-effects for uncharacterized drugs and more importantly able to predict rare side-effects which are often ignored by other approaches. The method described in this article can be useful to predict side-effects in drug design in an early stage to reduce experimental cost and time.

6.
BMC Genomics ; 13 Suppl 6: S8, 2012.
Article in English | MEDLINE | ID: mdl-23134806

ABSTRACT

BACKGROUND: Metastatic breast cancer is a leading cause of cancer-related deaths in women worldwide. DNA microarray has become an important tool to help identify biomarker genes for improving the prognosis of breast cancer. Recently, it was shown that pathway-level relationships between genes can be incorporated to build more robust classification models and to obtain more useful biological insight from such models. Due to the unavailability of complete pathways, protein-protein interaction (PPI) network is becoming more popular to researcher and opens a new way to investigate the developmental process of breast cancer. METHODS: In this study, a network-based method is proposed to combine microarray gene expression profiles and PPI network for biomarker discovery for breast cancer metastasis. The key idea in our approach is to identify a small number of genes to connect differentially expressed genes into a single component in a PPI network; these intermediate genes contain important information about the pathways involved in metastasis and have a high probability of being biomarkers. RESULTS: We applied this approach on two breast cancer microarray datasets, and for both cases we identified significant numbers of well-known biomarker genes for breast cancer metastasis. Those selected genes are significantly enriched with biological processes and pathways related to cancer carcinogenic process, and, importantly, have much higher stability across different datasets than in previous studies. Furthermore, our selected genes significantly increased cross-data classification accuracy of breast cancer metastasis. CONCLUSIONS: The randomized Steiner tree based approach described in this study is a new way to discover biomarker genes for breast cancer, and improves the prediction accuracy of metastasis. Though the analysis is limited here only to breast cancer, it can be easily applied to other diseases.


Subject(s)
Algorithms , Biomarkers/metabolism , Breast Neoplasms/metabolism , Neoplasm Metastasis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Databases, Factual , Female , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Prognosis , Protein Interaction Mapping
SELECTION OF CITATIONS
SEARCH DETAIL
...