1,364 research outputs found

    Using gene and microRNA expression in the human airway for lung cancer diagnosis

    Full text link
    Lung cancer surpasses all other causes of cancer-related deaths worldwide. Gene-expression microarrays have shown that differences in the cytologically normal bronchial airway can distinguish between patients with and without lung cancer. In research reported here, we have used microRNA expression in bronchial epithelium and gene expression in nasal epithelium to advance biological understanding of the lung-cancer "field of injury" and develop new biomarkers for lung cancer diagnosis. MicroRNAs are known to mediate the airway response to tobacco smoke exposure but their role in the lung-cancer-associated field of injury was previously unknown. Microarrays can measure microRNA expression; however, they are probe-based and limited to detecting annotated microRNAs. MicroRNA sequencing, on the other hand, allows the identification of novel microRNAs that may play important biological roles. We have used microRNA sequencing to discover novel microRNAs in the bronchial epithelium. One of the predicted microRNAs, now known as miR-4423, is associated with lung cancer and airway development. This finding demonstrates for the first time a microRNA expression change associated with the lung-cancer field of injury and microRNA mediation of gene expression changes within that field. The National Lung Screening Trial showed that screening high-risk smokers using CT scans decreases lung-cancer-associated mortality. Nodules were detected in over 20% of participants; however, the overwhelming majority of screening-detected nodules were non-malignant. We therefore need biomarkers to determine which screening-detected nodules are benign and do not require further invasive testing. Given that the lung-cancer-associated field of injury extends to the bronchial epithelium, our group hypothesized that the field of injury may extend farther up in the airway. Using gene expression microarrays, we have identified a nasal epithelium gene-expression signature associated with lung cancer. Using samples from the bronchial epithelium and the nasal epithelium, we have established that there is a common lung-cancer-associated gene-expression signature throughout the airway. In addition, we have developed a nasal epithelium gene-expression biomarker for lung cancer together with a clinico-genomic classifier that includes both clinical factors and gene expression. Our data suggests that gene expression profiling in nasal epithelium might serve as a non-invasive approach for lung cancer diagnosis and screenin

    Network Inference Algorithms Elucidate Nrf2 Regulation of Mouse Lung Oxidative Stress

    Get PDF
    A variety of cardiovascular, neurological, and neoplastic conditions have been associated with oxidative stress, i.e., conditions under which levels of reactive oxygen species (ROS) are elevated over significant periods. Nuclear factor erythroid 2-related factor (Nrf2) regulates the transcription of several gene products involved in the protective response to oxidative stress. The transcriptional regulatory and signaling relationships linking gene products involved in the response to oxidative stress are, currently, only partially resolved. Microarray data constitute RNA abundance measures representing gene expression patterns. In some cases, these patterns can identify the molecular interactions of gene products. They can be, in effect, proxies for protein–protein and protein–DNA interactions. Traditional techniques used for clustering coregulated genes on high-throughput gene arrays are rarely capable of distinguishing between direct transcriptional regulatory interactions and indirect ones. In this study, newly developed information-theoretic algorithms that employ the concept of mutual information were used: the Algorithm for the Reconstruction of Accurate Cellular Networks (ARACNE), and Context Likelihood of Relatedness (CLR). These algorithms captured dependencies in the gene expression profiles of the mouse lung, allowing the regulatory effect of Nrf2 in response to oxidative stress to be determined more precisely. In addition, a characterization of promoter sequences of Nrf2 regulatory targets was conducted using a Support Vector Machine classification algorithm to corroborate ARACNE and CLR predictions. Inferred networks were analyzed, compared, and integrated using the Collective Analysis of Biological Interaction Networks (CABIN) plug-in of Cytoscape. Using the two network inference algorithms and one machine learning algorithm, a number of both previously known and novel targets of Nrf2 transcriptional activation were identified. Genes predicted as novel Nrf2 targets include Atf1, Srxn1, Prnp, Sod2, Als2, Nfkbib, and Ppp1r15b. Furthermore, microarray and quantitative RT-PCR experiments following cigarette-smoke-induced oxidative stress in Nrf2+/+ and Nrf2−/− mouse lung affirmed many of the predictions made. Several new potential feed-forward regulatory loops involving Nrf2, Nqo1, Srxn1, Prdx1, Als2, Atf1, Sod1, and Park7 were predicted. This work shows the promise of network inference algorithms operating on high-throughput gene expression data in identifying transcriptional regulatory and other signaling relationships implicated in mammalian disease

    Robust selection of cancer survival signatures from high-throughput genomic data using two-fold subsampling

    Get PDF
    Identifying relevant signatures for clinical patient outcome is a fundamental task in high-throughput studies. Signatures, composed of features such as mRNAs, miRNAs, SNPs or other molecular variables, are often non-overlapping, even though they have been identified from similar experiments considering samples with the same type of disease. The lack of a consensus is mostly due to the fact that sample sizes are far smaller than the numbers of candidate features to be considered, and therefore signature selection suffers from large variation. We propose a robust signature selection method that enhances the selection stability of penalized regression algorithms for predicting survival risk. Our method is based on an aggregation of multiple, possibly unstable, signatures obtained with the preconditioned lasso algorithm applied to random (internal) subsamples of a given cohort data, where the aggregated signature is shrunken by a simple thresholding strategy. The resulting method, RS-PL, is conceptually simple and easy to apply, relying on parameters automatically tuned by cross validation. Robust signature selection using RS-PL operates within an (external) subsampling framework to estimate the selection probabilities of features in multiple trials of RS-PL. These probabilities are used for identifying reliable features to be included in a signature. Our method was evaluated on microarray data sets from neuroblastoma, lung adenocarcinoma, and breast cancer patients, extracting robust and relevant signatures for predicting survival risk. Signatures obtained by our method achieved high prediction performance and robustness, consistently over the three data sets. Genes with high selection probability in our robust signatures have been reported as cancer-relevant. The ordering of predictor coefficients associated with signatures was well-preserved across multiple trials of RS-PL, demonstrating the capability of our method for identifying a transferable consensus signature. The software is available as an R package rsig at CRAN (http://cran.r-project.org)

    VARIATIONS IN MICROARRAY BASED GENE EXPRESSION PROFILING: IDENTIFYING SOURCES AND IMPROVING RESULTS

    Get PDF
    Two major issues hinder the application of microarray based gene expression profiling in clinical laboratories as a diagnostic or prognostic tool. The first issue is the sheer volume and high-dimensionality of gene expression data from microarray experiments, which require advanced algorithms to extract meaningful gene expression patterns that correlate with biological impact. The second issue is the substantial amount of variation in microarray gene expression data, which impairs the performance of analysis method and makes sharing or integrating microarray data very difficult. Variations can be introduced by all possible sources including the DNA microarray technology itself and the experimental procedures. Many of these variations have not been characterized, measured, or linked to the sources. In the first part of this dissertation, a decision tree learning method was demonstrated to perform as well as more popularly accepted classification methods in partitioning cancer samples with microarray data. More importantly, results demonstrate that variation introduced into microarray data by tissue sampling and tissue handling compromised the performance of classification methods. In the second part of this dissertation, variations introduced by the T7 based in vitro transcription labeling methods were investigated in detail. Results demonstrated that individual amplification methods significantly biased gene expression data even though the methods compared in this study were all derivatives of the T7 RNA polymerase based in vitro transcription labeling approach. Variations observed can be partially explained by the number of biotinylated nucleotides used for labeling and the incubation time of the in vitro transcription experiments. These variations can generate discordant gene expression results even using the same RNA samples and cannot be corrected by post experiment analysis including advanced normalization techniques. Studies in this dissertation stress the concept that experimental and analytical methods must work together. This dissertation also emphasizes the importance of standardizing the DNA microarray technology and experimental procedures in order to optimize gene expression analysis and create quality standards compatible with the clinical application of this technology. These findings should be taken into account especially when comparing data from different platforms, and in standardizing protocols for clinical applications in pathology

    Multiple platform assessment of the EGF dependent transcriptome by microarray and deep tag sequencing analysis

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Epidermal Growth Factor (EGF) is a key regulatory growth factor activating many processes relevant to normal development and disease, affecting cell proliferation and survival. Here we use a combined approach to study the EGF dependent transcriptome of HeLa cells by using multiple long oligonucleotide based microarray platforms (from Agilent, Operon, and Illumina) in combination with digital gene expression profiling (DGE) with the Illumina Genome Analyzer.</p> <p>Results</p> <p>By applying a procedure for cross-platform data meta-analysis based on RankProd and GlobalAncova tests, we establish a well validated gene set with transcript levels altered after EGF treatment. We use this robust gene list to build higher order networks of gene interaction by interconnecting associated networks, supporting and extending the important role of the EGF signaling pathway in cancer. In addition, we find an entirely new set of genes previously unrelated to the currently accepted EGF associated cellular functions.</p> <p>Conclusions</p> <p>We propose that the use of global genomic cross-validation derived from high content technologies (microarrays or deep sequencing) can be used to generate more reliable datasets. This approach should help to improve the confidence of downstream <it>in silico </it>functional inference analyses based on high content data.</p

    Interpretation of Mutations, Expression, Copy Number in Somatic Breast Cancer: Implications for Metastasis and Chemotherapy

    Get PDF
    Breast cancer (BC) patient management has been transformed over the last two decades due to the development and application of genome-wide technologies. The vast amounts of data generated by these assays, however, create new challenges for accurate and comprehensive analysis and interpretation. This thesis describes novel methods for fluorescence in-situ hybridization (FISH), array comparative genomic hybridization (aCGH), and next generation DNA- and RNA-sequencing, to improve upon current approaches used for these technologies. An ab initio algorithm was implemented to identify genomic intervals of single copy and highly divergent repetitive sequences that were applied to FISH and aCGH probe design. FISH probes with higher resolution than commercially available reagents were developed and validated on metaphase chromosomes. An aCGH microarray was developed that had improved reproducibility compared to the standard Agilent 44K array, which was achieved by placing oligonucleotide probes distant from conserved repetitive sequences. Splicing mutations are currently underrepresented in genome-wide sequencing analyses, and there are limited methods to validate genome-wide mutation predictions. This thesis describes Veridical, a program developed to statistically validate aberrant splicing caused by a predicted mutation. Splicing mutation analysis was performed on a large subset of BC patients previously analyzed by the Cancer Genome Atlas. This analysis revealed an elevated number of splicing mutations in genes involved in NCAM pathways in basal-like and HER2-enriched lymph node positive tumours. Genome-wide technologies were leveraged further to develop chemosensitivity models that predict BC response to paclitaxel and gemcitabine. A type of machine learning, called support vector machines (SVM), was used to create predictive models from small sets of biologically-relevant genes to drug disposition or resistance. SVM models generated were able to predict sensitivity in two groups of independent patient data. High variability between individuals requires more accurate and higher resolution genomic data. However the data themselves are insufficient; also needed are more insightful analytical methods to fully exploit these data. This dissertation presents both improvements in data quality and accuracy as well as analytical procedures, with the aim of detecting and interpreting critical genomic abnormalities that are hallmarks of BC subtypes, metastasis and therapy response

    Developing RNA diagnostics for studying healthy human ageing

    Get PDF
    Developing strategies to cope with increase in the ageing population and age-related chronic diseases is one of the societies biggest challenges. The characteristics of the ageing process shows significant inter-individual variation. Building genomic signatures that could account for variation in health outcomes with age may facilitate early prognosis of individual age-correlated diseases (e.g. cancer, coronary artery diseases and dementia) and help in developing better targeted treatments provided years in advance of acquiring disabling symptoms for these diseases. The aim of this thesis was to explore methods for diagnosing molecular features of human ageing. In particular, we utilise multi-platform transcriptomics, independent clinical data and classification methods to evaluate which human tissues demonstrate a reproducible molecular signature for age and which clinical phenotypes correlated with these new RNA biomarkers. [Continues.

    Isoform-level gene signature improves prognostic stratification and accurately classifies glioblastoma subtypes.

    Get PDF
    Molecular stratification of tumors is essential for developing personalized therapies. Although patient stratification strategies have been successful; computational methods to accurately translate the gene-signature from high-throughput platform to a clinically adaptable low-dimensional platform are currently lacking. Here, we describe PIGExClass (platform-independent isoform-level gene-expression based classification-system), a novel computational approach to derive and then transfer gene-signatures from one analytical platform to another. We applied PIGExClass to design a reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) based molecular-subtyping assay for glioblastoma multiforme (GBM), the most aggressive primary brain tumors. Unsupervised clustering of TCGA (the Cancer Genome Altas Consortium) GBM samples, based on isoform-level gene-expression profiles, recaptured the four known molecular subgroups but switched the subtype for 19% of the samples, resulting in significant (P = 0.0103) survival differences among the refined subgroups. PIGExClass derived four-class classifier, which requires only 121 transcript-variants, assigns GBM patients' molecular subtype with 92% accuracy. This classifier was translated to an RT-qPCR assay and validated in an independent cohort of 206 GBM samples. Our results demonstrate the efficacy of PIGExClass in the design of clinically adaptable molecular subtyping assay and have implications for developing robust diagnostic assays for cancer patient stratification

    Establishment of predictive blood-based signatures in medical large scale genomic data sets : Development of novel diagnostic tests

    Get PDF
    Increasing data has led to tremendous success in discovering molecular biomarkers based on high throughput data. However, the translation of these so-called genomic signatures into clinical practice has been limited. The complexity and volume of genomic profiling requires heightened attention to robust design, methodological details, and avoidance of bias. During this thesis, novel strategies aimed at closing the gap from initially promising pilot studies to the clinical application of novel biomarkers are evaluated. First, a conventional process for genomic biomarker development comprising feature selection, algorithm and parameter optimization, and performance assessment was established. Using this approach, a RNA-stabilized whole blood diagnostic classifier for non-small cell lung cancer was built in a training set that can be used as a biomarker to discriminate between patients and control samples. Subsequently, this optimized classifier was successfully applied to two independent and blinded validation sets. Extensive permutation analysis using random feature lists supports the specificity of the established transcriptional classifier. Next, it was demonstrated that a combined approach of clinical trial simulation and adaptive learning strategies can be used to speed up biomarker development. As a model, genome-wide expression data derived from over 4,700 individuals in 37 studies addressing four clinical endpoints were used to assess over 1,800,000 classifiers. In addition to current approaches determining optimal classifiers within a defined study setting, randomized clinical trial simulation unequivocally uncovered the overall variance in the prediction performance of potential disease classifiers to predict the outcome of a large biomarker validation study from a pilot trial. Furthermore, most informative features were identified by feature ranking according to an individual classification performance score. Applying an adaptive learning strategy based on data extrapolation led to a datadriven prediction of the study size required for larger validation studies based on small pilot trials and an estimate of the expected statistical performance during validation. With these significant improvements, exceedingly robust and clinically applicable gene signatures for the diagnosis and detection of acute myeloid leukemia, active tuberculosis, HIV infection, and non-small cell lung cancer are established which could demonstrate disease-related enrichment of the obtained signatures and phenotype-related feature ranking. In further research, platform requirements for blood-based biomarker development were exemplarily examined for micro RNA expression profiling. The performance as well as the technical sample handling to provide reliable strategies for platform implementation in clinical applications were investigated. Overall, all introduced methods improve and accelerate the development of biomarker signatures for molecular diagnostics and can easily be extended to other high throughput data and other disease settings

    Cell Cycle Gene Networks Are Associated with Melanoma Prognosis

    Get PDF
    BACKGROUND: Our understanding of the molecular pathways that underlie melanoma remains incomplete. Although several published microarray studies of clinical melanomas have provided valuable information, we found only limited concordance between these studies. Therefore, we took an in vitro functional genomics approach to understand melanoma molecular pathways. METHODOLOGY/PRINCIPAL FINDINGS: Affymetrix microarray data were generated from A375 melanoma cells treated in vitro with siRNAs against 45 transcription factors and signaling molecules. Analysis of this data using unsupervised hierarchical clustering and Bayesian gene networks identified proliferation-association RNA clusters, which were co-ordinately expressed across the A375 cells and also across melanomas from patients. The abundance in metastatic melanomas of these cellular proliferation clusters and their putative upstream regulators was significantly associated with patient prognosis. An 8-gene classifier derived from gene network hub genes correctly classified the prognosis of 23/26 metastatic melanoma patients in a cross-validation study. Unlike the RNA clusters associated with cellular proliferation described above, co-ordinately expressed RNA clusters associated with immune response were clearly identified across melanoma tumours from patients but not across the siRNA-treated A375 cells, in which immune responses are not active. Three uncharacterised genes, which the gene networks predicted to be upstream of apoptosis- or cellular proliferation-associated RNAs, were found to significantly alter apoptosis and cell number when over-expressed in vitro. CONCLUSIONS/SIGNIFICANCE: This analysis identified co-expression of RNAs that encode functionally-related proteins, in particular, proliferation-associated RNA clusters that are linked to melanoma patient prognosis. Our analysis suggests that A375 cells in vitro may be valid models in which to study the gene expression modules that underlie some melanoma biological processes (e.g., proliferation) but not others (e.g., immune response). The gene expression modules identified here, and the RNAs predicted by Bayesian network inference to be upstream of these modules, are potential prognostic biomarkers and drug targets
    corecore