91 research outputs found

    Biomarker discovery across annotated and unannotated microarray datasets using semi-supervised learning

    Get PDF
    The growing body of DNA microarray data has the potential to advance our understanding of the molecular basis of disease. However annotating microarray datasets with clinically useful information is not always possible, as this often requires access to detailed patient records. In this study we introduce GLAD, a new Semi-Supervised Learning (SSL) method for combining independent annotated datasets and unannotated datasets with the aim of identifying more robust sample classifiers

    Genomics, Molecular Imaging, Bioinformatics, and Bio-Nano-Info Integration are Synergistic Components of Translational Medicine and Personalized Healthcare Research

    Get PDF
    Supported by National Science Foundation (NSF), International Society of Intelligent Biological Medicine (ISIBM), International Journal of Computational Biology and Drug Design and International Journal of Functional Informatics and Personalized Medicine, IEEE 7th Bioinformatics and Bioengineering attracted more than 600 papers and 500 researchers and medical doctors. It was the only synergistic inter/multidisciplinary IEEE conference with 24 Keynote Lectures, 7 Tutorials, 5 Cutting-Edge Research Workshops and 32 Scientific Sessions including 11 Special Research Interest Sessions that were designed dynamically at Harvard in response to the current research trends and advances. The committee was very grateful for the IEEE Plenary Keynote Lectures given by: Dr. A. Keith Dunker (Indiana), Dr. Jun Liu (Harvard), Dr. Brian Athey (Michigan), Dr. Mark Borodovsky (Georgia Tech and President of ISIBM), Dr. Hamid Arabnia (Georgia and Vice-President of ISIBM), Dr. Ruzena Bajcsy (Berkeley and Member of United States National Academy of Engineering and Member of United States Institute of Medicine of the National Academies), Dr. Mary Yang (United States National Institutes of Health and Oak Ridge, DOE), Dr. Chih-Ming Ho (UCLA and Member of United States National Academy of Engineering and Academician of Academia Sinica), Dr. Andy Baxevanis (United States National Institutes of Health), Dr. Arif Ghafoor (Purdue), Dr. John Quackenbush (Harvard), Dr. Eric Jakobsson (UIUC), Dr. Vladimir Uversky (Indiana), Dr. Laura Elnitski (United States National Institutes of Health) and other world-class scientific leaders. The Harvard meeting was a large academic event 100% full-sponsored by IEEE financially and academically. After a rigorous peer-review process, the committee selected 27 high-quality research papers from 600 submissions. The committee is grateful for contributions from keynote speakers Dr. Russ Altman (IEEE BIBM conference keynote lecturer on combining simulation and machine learning to recognize function in 4D), Dr. Mary Qu Yang (IEEE BIBM workshop keynote lecturer on new initiatives of detecting microscopic disease using machine learning and molecular biology, http://ieeexplore.ieee.org/servlet/opac? punumber=4425386) and Dr. Jack Y.Yang (IEEE BIBM workshop keynote lecturer on data mining and knowledge discovery in translational medicine) from the first IEEE Computer Society BioInformatics and BioMedicine (IEEE BIBM) international conference and workshops, November 2- 4, 2007, Silicon Valley, California, USA

    Biomarker lists stability in genomic studies: analysis and improvement by prior biological knowledge integration into the learning process

    Get PDF
    The analysis of high-throughput sequencing, microarray and mass spectrometry data has been demonstrated extremely helpful for the identification of those genes and proteins, called biomarkers, helpful for answering to both diagnostic/prognostic and functional questions. In this context, robustness of the results is critical both to understand the biological mechanisms underlying diseases and to gain sufficient reliability for clinical/pharmaceutical applications. Recently, different studies have proved that the lists of identified biomarkers are poorly reproducible, making the validation of biomarkers as robust predictors of a disease a still open issue. The reasons of these differences are referable to both data dimensions (few subjects with respect to the number of features) and heterogeneity of complex diseases, characterized by alterations of multiple regulatory pathways and of the interplay between different genes and the environment. Typically in an experimental design, data to analyze come from different subjects and different phenotypes (e.g. normal and pathological). The most widely used methodologies for the identification of significant genes related to a disease from microarray data are based on computing differential gene expression between different phenotypes by univariate statistical tests. Such approach provides information on the effect of specific genes as independent features, whereas it is now recognized that the interplay among weakly up/down regulated genes, although not significantly differentially expressed, might be extremely important to characterize a disease status. Machine learning algorithms are, in principle, able to identify multivariate nonlinear combinations of features and have thus the possibility to select a more complete set of experimentally relevant features. In this context, supervised classification methods are often used to select biomarkers, and different methods, like discriminant analysis, random forests and support vector machines among others, have been used, especially in cancer studies. Although high accuracy is often achieved in classification approaches, the reproducibility of biomarker lists still remains an open issue, since many possible sets of biological features (i.e. genes or proteins) can be considered equally relevant in terms of prediction, thus it is in principle possible to have a lack of stability even by achieving the best accuracy. This thesis represents a study of several computational aspects related to biomarker discovery in genomic studies: from the classification and feature selection strategies to the type and the reliability of the biological information used, proposing new approaches able to cope with the problem of the reproducibility of biomarker lists. The study has highlighted that, although reasonable and comparable classification accuracy can be achieved by different methods, further developments are necessary to achieve robust biomarker lists stability, because of the high number of features and the high correlation among them. In particular, this thesis proposes two different approaches to improve biomarker lists stability by using prior information related to biological interplay and functional correlation among the analyzed features. Both approaches were able to improve biomarker selection. The first approach, using prior information to divide the application of the method into different subproblems, improves results interpretability and offers an alternative way to assess lists reproducibility. The second, integrating prior information in the kernel function of the learning algorithm, improves lists stability. Finally, the interpretability of results is strongly affected by the quality of the biological information available and the analysis of the heterogeneities performed in the Gene Ontology database has revealed the importance of providing new methods able to verify the reliability of the biological properties which are assigned to a specific feature, discriminating missing or less specific information from possible inconsistencies among the annotations. These aspects will be more and more deepened in the future, as the new sequencing technologies will monitor an increasing number of features and the number of functional annotations from genomic databases will considerably grow in the next years.L’analisi di dati high-throughput basata sull’utilizzo di tecnologie di sequencing, microarray e spettrometria di massa si è dimostrata estremamente utile per l’identificazione di quei geni e proteine, chiamati biomarcatori, utili per rispondere a quesiti sia di tipo diagnostico/prognostico che funzionale. In tale contesto, la stabilità dei risultati è cruciale sia per capire i meccanismi biologici che caratterizzano le malattie sia per ottenere una sufficiente affidabilità per applicazioni in campo clinico/farmaceutico. Recentemente, diversi studi hanno dimostrato che le liste di biomarcatori identificati sono scarsamente riproducibili, rendendo la validazione di tali biomarcatori come indicatori stabili di una malattia un problema ancora aperto. Le ragioni di queste differenze sono imputabili sia alla dimensione dei dataset (pochi soggetti rispetto al numero di variabili) sia all’eterogeneità di malattie complesse, caratterizzate da alterazioni di più pathway di regolazione e delle interazioni tra diversi geni e l’ambiente. Tipicamente in un disegno sperimentale, i dati da analizzare provengono da diversi soggetti e diversi fenotipi (e.g. normali e patologici). Le metodologie maggiormente utilizzate per l’identificazione di geni legati ad una malattia si basano sull’analisi differenziale dell’espressione genica tra i diversi fenotipi usando test statistici univariati. Tale approccio fornisce le informazioni sull’effetto di specifici geni considerati come variabili indipendenti tra loro, mentre è ormai noto che l’interazione tra geni debolmente up/down regolati, sebbene non differenzialmente espressi, potrebbe rivelarsi estremamente importante per caratterizzare lo stato di una malattia. Gli algoritmi di machine learning sono, in linea di principio, capaci di identificare combinazioni non lineari delle variabili e hanno quindi la possibilità di selezionare un insieme più dettagliato di geni che sono sperimentalmente rilevanti. In tale contesto, i metodi di classificazione supervisionata vengono spesso utilizzati per selezionare i biomarcatori, e diversi approcci, quali discriminant analysis, random forests e support vector machines tra altri, sono stati utilizzati, soprattutto in studi oncologici. Sebbene con tali approcci di classificazione si ottenga un alto livello di accuratezza di predizione, la riproducibilità delle liste di biomarcatori rimane ancora una questione aperta, dato che esistono molteplici set di variabili biologiche (i.e. geni o proteine) che possono essere considerati ugualmente rilevanti in termini di predizione. Quindi in teoria è possibile avere un’insufficiente stabilità anche raggiungendo il massimo livello di accuratezza. Questa tesi rappresenta uno studio su diversi aspetti computazionali legati all’identificazione di biomarcatori in genomica: dalle strategie di classificazione e di feature selection adottate alla tipologia e affidabilità dell’informazione biologica utilizzata, proponendo nuovi approcci in grado di affrontare il problema della riproducibilità delle liste di biomarcatori. Tale studio ha evidenziato che sebbene un’accettabile e comparabile accuratezza nella predizione può essere ottenuta attraverso diversi metodi, ulteriori sviluppi sono necessari per raggiungere una robusta stabilità nelle liste di biomarcatori, a causa dell’alto numero di variabili e dell’alto livello di correlazione tra loro. In particolare, questa tesi propone due diversi approcci per migliorare la stabilità delle liste di biomarcatori usando l’informazione a priori legata alle interazioni biologiche e alla correlazione funzionale tra le features analizzate. Entrambi gli approcci sono stati in grado di migliorare la selezione di biomarcatori. Il primo approccio, usando l’informazione a priori per dividere l’applicazione del metodo in diversi sottoproblemi, migliora l’interpretabilità dei risultati e offre un modo alternativo per verificare la riproducibilità delle liste. Il secondo, integrando l’informazione a priori in una funzione kernel dell’algoritmo di learning, migliora la stabilità delle liste. Infine, l’interpretabilità dei risultati è fortemente influenzata dalla qualità dell’informazione biologica disponibile e l’analisi delle eterogeneità delle annotazioni effettuata sul database Gene Ontology rivela l’importanza di fornire nuovi metodi in grado di verificare l’attendibilità delle proprietà biologiche che vengono assegnate ad una specifica variabile, distinguendo la mancanza o la minore specificità di informazione da possibili inconsistenze tra le annotazioni. Questi aspetti verranno sempre più approfonditi in futuro, dato che le nuove tecnologie di sequencing monitoreranno un maggior numero di variabili e il numero di annotazioni funzionali derivanti dai database genomici crescer`a considerevolmente nei prossimi anni

    Multi-class gene expression biomarker panel identification for the diagnosis of paediatric febrile illness

    Get PDF
    Febrile illness in children can result from infections by diverse viral or bacterial pathogens as well as inflammatory conditions or cancer. The limitations of the existing diagnostic pipeline, which relies on clinical symptoms and signs, pathogen detection, empirical treatment and diagnoses of exclusion, contribute to missed or de- layed diagnosis and unnecessary antibiotic use. The potential of host gene expression biomarkers measured in blood has been demonstrated for simplified binary diagnostic questions however, the clinical reality is that multiple potential aetiologies must be considered and prioritised on the basis of likelihood and risks of severe disease. In order to identify a biomarker panel which better reflects this clinical reality, we applied a multi-class supervised learning approach to whole blood transcriptomic datasets from children with infectious and inflammatory disease. Three datasets were used for the analyses presented here, a single microarray dataset, a meta-analysis of 12 publicly available microarray datasets and a newly generated RNA-sequencing dataset. These were used for preliminary investigations of the approach, discovery of a multi-class biomarker panel of febrile illness and valida- tion of the biomarker panel respectively. In the merged microarray discovery dataset a two-stage approach to feature selection and classification, based on LASSO and Ridge penalised regression was applied to distinguish 18 disease classes. Cost-sensitivity was incorporated in the approach as aetiologies of febrile illness vary considerably in the risk of severe disease. The resulting 161 transcript biomarker panel could reliably distinguish bacterial, viral, inflammatory, tuberculosis and malarial disease as well as pathogen specific aetiologies. The panel was then validated in a newly generated RNA-Seq dataset and compared to previously published binary biomarker panels. The analyses presented here demonstrate that a single test for the diagnosis of acute febrile illness in children is possible using host RNA biomarkers. A test which could distinguish multiple aetiologies soon after presentation could be used to reduce unnecessary antibiotic use, improve targetting of antibiotics to bacterial species and reduce delays in the diagnosis of inflammatory diseases.Open Acces

    Discovering Biomarkers of Alzheimer's Disease by Statistical Learning Approaches

    Get PDF
    In this work, statistical learning approaches are exploited to discover biomarkers for Alzheimer's disease (AD). The contributions has been made in the fields of both biomarker and software driven studies. Surprising discoveries were made in the field of blood-based biomarker search. With the inclusion of existing biological knowledge and a proposed novel feature selection method, several blood-based protein models were discovered to have promising ability to separate AD patients from healthy individuals. A new statistical pattern was discovered which can be potential new guideline for diagnosis methodology. In the field of brain-based biomarker, the positive contribution of covariates such as age, gender and APOE genotype to a AD classifier was verified, as well as the discovery of panel of highly informative biomarkers comprising 26 RNA transcripts. The classifier trained by the panetl of genes shows excellent capacity in discriminating patients from control. Apart from biomarker driven studies, the development of statistical packages or application were also involved. R package metaUnion was designed and developed to provide advanced meta-analytic approach applicable for microarray data. This package overcomes the defects appearing in previous meta-analytic packages { 1) the neglection of missing data, 2) the in exibility of feature dimension 3) the lack of functions to support post-analysis summary. R package metaUnion has been applied in a published study as part of the integrated genomic approaches and resulted in significant findings. To provide benchmark references about significance of features for dementia researchers, a web-based platform AlzExpress was built to provide researchers with granular level of differential expression test and meta-analysis results. A combination of fashionable big data technologies and robust data mining algorithms make AlzExpress flexible, scalable and comprehensive platform of valuable bioinformatics in dementia research.Plymouth Universit

    Integrative Transcriptomic Analysis of Long Intergenic Non-Coding RNAs in Cancer.

    Get PDF
    Ph.D. Thesis. University of Hawaiʻi at Mānoa 2017

    Bioinformatics applied to human genomics and proteomics: development of algorithms and methods for the discovery of molecular signatures derived from omic data and for the construction of co-expression and interaction networks

    Get PDF
    [EN] The present PhD dissertation develops and applies Bioinformatic methods and tools to address key current problems in the analysis of human omic data. This PhD has been organised by main objectives into four different chapters focused on: (i) development of an algorithm for the analysis of changes and heterogeneity in large-scale omic data; (ii) development of a method for non-parametric feature selection; (iii) integration and analysis of human protein-protein interaction networks and (iv) integration and analysis of human co-expression networks derived from tissue expression data and evolutionary profiles of proteins. In the first chapter, we developed and tested a new robust algorithm in R, called DECO, for the discovery of subgroups of features and samples within large-scale omic datasets, exploring all feature differences possible heterogeneity, through the integration of both data dispersion and predictor-response information in a new statistic parameter called h (heterogeneity score). In the second chapter, we present a simple non-parametric statistic to measure the cohesiveness of categorical variables along any quantitative variable, applicable to feature selection in all types of big data sets. In the third chapter, we describe an analysis of the human interactome integrating two global datasets from high-quality proteomics technologies: HuRI (a human protein-protein interaction network generated by a systematic experimental screening based on Yeast-Two-Hybrid technology) and Cell-Atlas (a comprehensive map of subcellular localization of human proteins generated by antibody imaging). This analysis aims to create a framework for the subcellular localization characterization supported by the human protein-protein interactome. In the fourth chapter, we developed a full integration of three high-quality proteome-wide resources (Human Protein Atlas, OMA and TimeTree) to generate a robust human co-expression network across tissues assigning each human protein along the evolutionary timeline. In this way, we investigate how old in evolution and how correlated are the different human proteins, and we place all them in a common interaction network. As main general comment, all the work presented in this PhD uses and develops a wide variety of bioinformatic and statistical tools for the analysis, integration and enlighten of molecular signatures and biological networks using human omic data. Most of this data corresponds to sample cohorts generated in recent biomedical studies on specific human diseases

    Machine learning and computational methods to identify molecular and clinical markers for complex diseases – case studies in cancer and obesity

    Get PDF
    In biomedical research, applied machine learning and bioinformatics are the essential disciplines heavily involved in translating data-driven findings into medical practice. This task is especially accomplished by developing computational tools and algorithms assisting in detection and clarification of underlying causes of the diseases. The continuous advancements in high-throughput technologies coupled with the recently promoted data sharing policies have contributed to presence of a massive wealth of data with remarkable potential to improve human health care. In concordance with this massive boost in data production, innovative data analysis tools and methods are required to meet the growing demand. The data analyzed by bioinformaticians and computational biology experts can be broadly divided into molecular and conventional clinical data categories. The aim of this thesis was to develop novel statistical and machine learning tools and to incorporate the existing state-of-the-art methods to analyze bio-clinical data with medical applications. The findings of the studies demonstrate the impact of computational approaches in clinical decision making by improving patients risk stratification and prediction of disease outcomes. This thesis is comprised of five studies explaining method development for 1) genomic data, 2) conventional clinical data and 3) integration of genomic and clinical data. With genomic data, the main focus is detection of differentially expressed genes as the most common task in transcriptome profiling projects. In addition to reviewing available differential expression tools, a data-adaptive statistical method called Reproducibility Optimized Test Statistic (ROTS) is proposed for detecting differential expression in RNA-sequencing studies. In order to prove the efficacy of ROTS in real biomedical applications, the method is used to identify prognostic markers in clear cell renal cell carcinoma (ccRCC). In addition to previously known markers, novel genes with potential prognostic and therapeutic role in ccRCC are detected. For conventional clinical data, ensemble based predictive models are developed to provide clinical decision support in treatment of patients with metastatic castration resistant prostate cancer (mCRPC). The proposed predictive models cover treatment and survival stratification tasks for both trial-based and realworld patient cohorts. Finally, genomic and conventional clinical data are integrated to demonstrate the importance of inclusion of genomic data in predictive ability of clinical models. Again, utilizing ensemble-based learners, a novel model is proposed to predict adulthood obesity using both genetic and social-environmental factors. Overall, the ultimate objective of this work is to demonstrate the importance of clinical bioinformatics and machine learning for bio-clinical marker discovery in complex disease with high heterogeneity. In case of cancer, the interpretability of clinical models strongly depends on predictive markers with high reproducibility supported by validation data. The discovery of these markers would increase chance of early detection and improve prognosis assessment and treatment choice

    Different approaches to measuring gene expression and DNA methylation and their application in cancer research

    Get PDF
    corecore