576 research outputs found

    Current Challenges in Modeling Cellular Metabolism

    Get PDF
    Mathematical and computational models play an essential role in understanding the cellular metabolism. They are used as platforms to integrate current knowledge on a biological system and to systematically test and predict the effect of manipulations to such systems. The recent advances in genome sequencing techniques have facilitated the reconstruction of genome-scale metabolic networks for a wide variety of organisms from microbes to human cells. These models have been successfully used in multiple biotechnological applications. Despite these advancements, modeling cellular metabolism still presents many challenges. The aim of this Research Topic is not only to expose and consolidate the state-of-the-art in metabolic modeling approaches, but also to push this frontier beyond the current edge through the introduction of innovative solutions. The articles presented in this e-book address some of the main challenges in the field, including the integration of different modeling formalisms, the integration of heterogeneous data sources into metabolic models, explicit representation of other biological processes during phenotype simulation, and standardization efforts in the representation of metabolic models and simulation results

    Designing Data-Driven Learning Algorithms: A Necessity to Ensure Effective Post-Genomic Medicine and Biomedical Research

    Get PDF
    Advances in sequencing technology have significantly contributed to shaping the area of genetics and enabled the identification of genetic variants associated with complex traits through genome-wide association studies. This has provided insights into genetic medicine, in which case, genetic factors influence variability in disease and treatment outcomes. On the other side, the missing or hidden heritability has suggested that the host quality of life and other environmental factors may also influence differences in disease risk and drug/treatment responses in genomic medicine, and orient biomedical research, even though this may be highly constrained by genetic capabilities. It is expected that combining these different factors can yield a paradigm-shift of personalized medicine and lead to a more effective medical treatment. With existing “big data” initiatives and high-performance computing infrastructures, there is a need for data-driven learning algorithms and models that enable the selection and prioritization of relevant genetic variants (post-genomic medicine) and trigger effective translation into clinical practice. In this chapter, we survey and discuss existing machine learning algorithms and post-genomic analysis models supporting the process of identifying valuable markers

    Developments in the tools and methodologies of synthetic biology.

    Get PDF
    Synthetic biology is principally concerned with the rational design and engineering of biologically based parts, devices, or systems. However, biological systems are generally complex and unpredictable, and are therefore, intrinsically difficult to engineer. In order to address these fundamental challenges, synthetic biology is aiming to unify a body of knowledge from several foundational scientific fields, within the context of a set of engineering principles. This shift in perspective is enabling synthetic biologists to address complexity, such that robust biological systems can be designed, assembled, and tested as part of a biological design cycle. The design cycle takes a forward-design approach in which a biological system is specified, modeled, analyzed, assembled, and its functionality tested. At each stage of the design cycle, an expanding repertoire of tools is being developed. In this review, we highlight several of these tools in terms of their applications and benefits to the synthetic biology community

    Elucidating dynamic metabolic physiology through network integration of quantitative time-course metabolomics.

    Get PDF
    Efst á síðunni er hægt að nálgast greinina í heild sinni með því að smella á hlekkinn To access publisher's full text version of this article. Please click on the hyperlink in Additional Links field.The increasing availability of metabolomics data necessitates novel methods for deeper data analysis and interpretation. We present a flux balance analysis method that allows for the computation of dynamic intracellular metabolic changes at the cellular scale through integration of time-course absolute quantitative metabolomics. This approach, termed "unsteady-state flux balance analysis" (uFBA), is applied to four cellular systems: three dynamic and one steady-state as a negative control. uFBA and FBA predictions are contrasted, and uFBA is found to be more accurate in predicting dynamic metabolic flux states for red blood cells, platelets, and Saccharomyces cerevisiae. Notably, only uFBA predicts that stored red blood cells metabolize TCA intermediates to regenerate important cofactors, such as ATP, NADH, and NADPH. These pathway usage predictions were subsequently validated through (13)C isotopic labeling and metabolic flux analysis in stored red blood cells. Utilizing time-course metabolomics data, uFBA provides an accurate method to predict metabolic physiology at the cellular scale for dynamic systems.National Heart Lung and Blood Institute European Research Council U.S. Department of Energ

    Doctor of Philosophy

    Get PDF
    dissertationCancer is extremely challenging to treat as every patient responds differently to treatments, depending on the specific molecular aberrations and deregulated signaling pathways driving their tumors. To address this heterogeneity and improve patient outcomes, therapies targeting specific pathways have been developed. The use of computational pathway analysis tools and genomic data can help guide the use of targeted therapies by assessing which pathways are deregulated in patient subpopulations and individual tumors. However, most pathway analysis tools do not account for complex interactions inherent to signaling pathways, and are not capable of integrating different types of genomic data (multiomic data). To address these limitations, this dissertation focuses on developing user-friendly multiomic gene set analysis tools, and utilizing bioinformatics tools to measure pathway activation for multiple pathways simultaneously in cancer. Chapter 2 first describes the need for genomics and pathway-based analyses in cancer using the commonly aberrant RAS pathway as an example. Chapter 3 utilizes pathway-based gene expression signatures and the pathway analysis toolkit ASSIGN to interrogate pathways from the growth factor receptor network (GFRN) in breast cancer. Two discrete phenotypes, which correlated with mechanisms of apoptosis and drug response, were characterized from GFRN activity. These phenotypes have the potential to pinpoint more effective breast cancer treatments. Chapter 4 describes the development of Gene Set Omic Analysis (GSOA), a novel gene set analysis tool which uses machine learning to identify pathway differences between two given biologicalconditions from multiomic data. GSOA demonstrated its capacity to identify pathways known to play a role in various cancers, and improves upon other methods because of its ability to decipher complex multigene and multiomic patterns. Chapter 5 describes GSOA-shiny, a novel web application for GSOA, which provides biologists with lack of bioinformatics experience access to multiomic gene set analysis from an easy-to-use interface. Overall, this dissertation presents novel breast cancer phenotypes with clinical implications, provides the research community with gene expression signatures for GFRN components, and presents an innovative method and web application for gene set analysisâ€"all contributing to furthering the field of personalized oncology

    Toward Multiscale Models of Cyanobacterial Growth

    Get PDF
    Oxygenic photosynthesis dominates global primary productivity ever since its evolution more than three billion years ago. While many aspects of phototrophic growth are well understood, it remains a considerable challenge to elucidate the manifold dependencies and interconnections between the diverse cellular processes that together facilitate the synthesis of new cells. Phototrophic growth involves the coordinated action of several layers of cellular functioning, ranging from the photosynthetic light reactions and the electron transport chain, to carbon-concentrating mechanisms and the assimilation of inorganic carbon. It requires the synthesis of new building blocks by cellular metabolism, protection against excessive light, as well as diurnal regulation by a circadian clock and the orchestration of gene expression and cell division. Computational modeling allows us to quantitatively describe these cellular functions and processes relevant for phototrophic growth. As yet, however, computational models are mostly confined to the inner workings of individual cellular processes, rather than describing the manifold interactions between them in the context of a living cell. Using cyanobacteria as model organisms, this contribution seeks to summarize existing computational models that are relevant to describe phototrophic growth and seeks to outline their interactions and dependencies. Our ultimate aim is to understand cellular functioning and growth as the outcome of a coordinated operation of diverse yet interconnected cellular processes.Peer Reviewe

    Doctor of Philosophy

    Get PDF
    dissertationGene expression data repositories provide large and ever increasing data for secondary use by translational informatics methods. For example, Gene Expression Omnibus (GEO) houses over 37,000 experiments with the goal of supporting further research. To use these published results in a larger meta-analysis, consolidation of the data are needed; however, the data are largely unstructured, thus hindering data integration efforts. Here, I propose the use of a novel pipeline, Ontology Based Data Integration (OBDI), which uses an ontological approach to combine the samples across multiple GEO experiments. The ODBI pipeline uses machine learning algorithms that permit researchers to consolidate and analyze data across GEO experiments. Here, I demonstrate how using an ontological approach to integrate samples across experiments can be used to explore the immune response at a molecular level. As part of this process, a Web Ontology Language (OWL) was developed for each data platform used. OWL serves as a core component in successfully processing different sample types. Immunological experiments from GEO were consolidated to evaluate this methodology. The experiments included samples analyzed on expression arrays, BeadChips, and sequencing technologies. The integration of a complex biological system and the incorporation of different biological data types will validate the potential of OBDI. iv The nature of biological data is highly dimensional. OBDI incorporates tools and techniques that can handle the analysis of various biological data. The machine learning analysis performed within the OBDI pipeline successfully evaluated the newly annotated experiments and provides insights that can be further explored. The OBDI pipeline can help researchers annotate experiments using ontologies and analyze the annotated experiments. To successfully build the pipeline, ontologies served as the backbone of integrating samples from GEO Series records into machine learning experiments using ML-Flex. By using the OBDI pipeline, researchers can access the uncurated experiments from GEO (GEO Data Series) and annotate the data using the terms in the ontologies. This mechanism allows for the organization of data sets in relationship to new experiments independent of GEO's GDS curation process. The OBDI system allows ontologies to grow organically around a cluster of experiments. These experiments are then further analyzed in ML-Flex using machine learning algorithms. The curated experiments are analyzed in silico and the computational analyses are supported by the OBDI ontological system

    Computational Logic for Biomedicine and Neurosciences

    Get PDF
    We advocate here the use of computational logic for systems biology, as a \emph{unified and safe} framework well suited for both modeling the dynamic behaviour of biological systems, expressing properties of them, and verifying these properties. The potential candidate logics should have a traditional proof theoretic pedigree (including either induction, or a sequent calculus presentation enjoying cut-elimination and focusing), and should come with certified proof tools. Beyond providing a reliable framework, this allows the correct encodings of our biological systems. % For systems biology in general and biomedicine in particular, we have so far, for the modeling part, three candidate logics: all based on linear logic. The studied properties and their proofs are formalized in a very expressive (non linear) inductive logic: the Calculus of Inductive Constructions (CIC). The examples we have considered so far are relatively simple ones; however, all coming with formal semi-automatic proofs in the Coq system, which implements CIC. In neuroscience, we are directly using CIC and Coq, to model neurons and some simple neuronal circuits and prove some of their dynamic properties. % In biomedicine, the study of multi omic pathway interactions, together with clinical and electronic health record data should help in drug discovery and disease diagnosis. Future work includes using more automatic provers. This should enable us to specify and study more realistic examples, and in the long term to provide a system for disease diagnosis and therapy prognosis
    corecore