Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Sci Adv ; 10(19): eadj1424, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38718126

ABSTRACT

The ongoing expansion of human genomic datasets propels therapeutic target identification; however, extracting gene-disease associations from gene annotations remains challenging. Here, we introduce Mantis-ML 2.0, a framework integrating AstraZeneca's Biological Insights Knowledge Graph and numerous tabular datasets, to assess gene-disease probabilities throughout the phenome. We use graph neural networks, capturing the graph's holistic structure, and train them on hundreds of balanced datasets via a robust semi-supervised learning framework to provide gene-disease probabilities across the human exome. Mantis-ML 2.0 incorporates natural language processing to automate disease-relevant feature selection for thousands of diseases. The enhanced models demonstrate a 6.9% average classification power boost, achieving a median receiver operating characteristic (ROC) area under curve (AUC) score of 0.90 across 5220 diseases from Human Phenotype Ontology, OpenTargets, and Genomics England. Notably, Mantis-ML 2.0 prioritizes associations from an independent UK Biobank phenome-wide association study (PheWAS), providing a stronger form of triaging and mitigating against underpowered PheWAS associations. Results are exposed through an interactive web resource.


Subject(s)
Biological Specimen Banks , Neural Networks, Computer , Humans , Genome-Wide Association Study/methods , Phenotype , United Kingdom , Phenomics/methods , Genetic Predisposition to Disease , Genomics/methods , Databases, Genetic , Algorithms , Computational Biology/methods , UK Biobank
2.
Bioresour Technol ; 387: 129683, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37597572

ABSTRACT

Anaerobic digestion is an established method for the biological conversion of waste feedstocks to biogas and biomethane. While anaerobic digestion is an excellent waste management technique, it can be susceptible to toxins and pollutants from contaminated feedstocks, which may have a detrimental impact on a digester's efficiency and productivity. Ethylene glycol (EG) is readily used in the heat-transfer loops of anaerobic digestion facilities to maintain reactor temperature. Failure of the structural integrity of these heat transfer loops can cause EG to leak into the digester, potentially causing a decrease in the resultant gas yields. Batch fermentations were incubated with 0, 10, 100 and 500 ppm (parts per million) of EG, and analysis showed that the EG was completely metabolised by the digester microbiome. The concentrations of EG tested showed significant increases in gas yields, however there were no significant changes to the digester microbiome.


Subject(s)
Metagenome , Microbiota , Anaerobiosis , Biofuels , Ethylene Glycols
3.
Sci Rep ; 9(1): 19995, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31882654

ABSTRACT

B cells are postulated to be central in seropositive rheumatoid arthritis (RA). Here, we use exploratory mass cytometry (n = 23) and next-generation sequencing (n = 19) to study B-cell repertoire shifts in RA patients. Expression of several B-cell markers were significantly different in ACPA+ RA compared to healthy controls, including an increase in HLA-DR across subsets, CD22 in clusters of IgM+ B cells and CD11c in IgA+ memory. Moreover, both IgA+ and IgG+ double negative (IgD- CD27-) CD11c+ B cells were increased in ACPA+ RA, and there was a trend for elevation in a CXCR5/CCR6high transitional B-cell cluster. In the RA BCR repertoire, there were significant differences in subclass distribution and, notably, the frequency of VH with low somatic hypermutation (SHM) was strikingly higher, especially in IgG1 (p < 0.0001). Furthermore, both ACPA+ and ACPA- RA patients had significantly higher total serum IgA and IgM compared to controls, based on serology of larger cohorts (n = 3494 IgA; n = 397 IgM). The observed elevated Ig-levels, distortion in IgM+ B cells, increase in double negative B cells, change in B-cell markers, and elevation of unmutated IgG+ B cells suggests defects in B-cell tolerance in RA. This may represent an underlying cause of increased polyreactivity and autoimmunity in RA.


Subject(s)
Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Disease Susceptibility , Immune Tolerance , Adaptive Immunity , Arthritis, Rheumatoid/pathology , Autoantibodies/immunology , Autoimmunity , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Biomarkers , CD11c Antigen/metabolism , HLA-DR Antigens/immunology , Humans , Immunoglobulin A/immunology , Immunoglobulin M/immunology , Immunologic Memory , Receptors, Antigen, B-Cell/metabolism
4.
Methods Mol Biol ; 1939: 181-198, 2019.
Article in English | MEDLINE | ID: mdl-30848462

ABSTRACT

In the era of big data and informatics, computational integration of data across the hierarchical structures of human biology enables discovery of new druggable targets of disease and new mode of action of a drug. We present herein a computational framework and guide of integrating drug targets, gene expression data, transcription factors, and prior knowledge of protein interactions to computationally construct the signaling network (mode of action) of a drug. In a similar manner, a disease network is constructed using its disease targets. And then, drug candidates are computationally prioritized by computationally ranking the closeness between a disease network and a drug's signaling network. Furthermore, we describe the use of the most perturbed HLA genes to assess the safety risk for immune-mediated adverse reactions such as Stevens-Johnson syndrome/toxic epidermal necrolysis.


Subject(s)
Computational Biology/methods , Drug Discovery/methods , Big Data , Databases, Factual , Humans , Precision Medicine/methods , Programming, Linear , Protein Interaction Maps/drug effects , Stevens-Johnson Syndrome/etiology , Transcriptome/drug effects
5.
CPT Pharmacometrics Syst Pharmacol ; 7(3): 166-174, 2018 03.
Article in English | MEDLINE | ID: mdl-29341478

ABSTRACT

Drug-induced cardiomyopathy contributes to drug attrition. We compared two pipelines of predictive modeling: (1) applying elastic net (EN) to differentially expressed genes (DEGs) of drugs; (2) applying integer linear programming (ILP) to construct each drug's signaling pathway starting from its targets to downstream proteins, to transcription factors, and to its DEGs in human cardiomyocytes, and then subjecting the genes/proteins in the drugs' signaling networks to EN regression. We classified 31 drugs with availability of DEGs into 13 toxic and 18 nontoxic drugs based on a clinical cardiomyopathy incidence cutoff of 0.1%. The ILP-augmented modeling increased prediction accuracy from 79% to 88% (sensitivity: 88%; specificity: 89%) under leave-one-out cross validation. The ILP-constructed signaling networks of drugs were better predictors than DEGs. Per literature, the microRNAs that reportedly regulate expression of our six top predictors are of diagnostic value for natural heart failure or doxorubicin-induced cardiomyopathy. This translational predictive modeling might uncover potential biomarkers.


Subject(s)
Cardiomyopathies/chemically induced , Doxorubicin/pharmacology , Gene Regulatory Networks/drug effects , Signal Transduction/drug effects , Cardiomyopathies/metabolism , Cardiotoxicity , Computational Biology , Doxorubicin/adverse effects , Humans , Models, Biological , Molecular Targeted Therapy , Regression Analysis , Translational Research, Biomedical
6.
Acta Orthop Belg ; 84(4): 443-451, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30879449

ABSTRACT

The present study aims to evaluate the efficacy of matrix-induced adipose-derived mesenchymal stem cells (Ad-MSCs) for cartilage repair of focal chondral knee lesions. Twenty patients were initially treated for symptomatic full-thickness chondral defects and then prospectively followed for two years. All patients underwent a single- stage procedure consisting in filling each defect with autologous culture-expanded mesenchymal stem cells embedded in a trimmed-to-fit commercially available biodegradable matrix. Knee-related function was evaluated based on subjective scores given by two self-reported questionnaires (KOOS and IKDC). Data analysis shows significant improvements (p<0.001) in all values. The mean preoperative scores in the subscales of KOOS, as well as in the IKDC subjective score were constantly increased during the follow-up period with statistically significant differences at 6, 12 and 24 months follow-up. The findings of this study indicate that matrix- induced adipose-derived mesenchymal stem cells implantation is an effective and safe single-staged cell-based procedure to manage full-thickness focal chondral lesions of the knee.


Subject(s)
Cartilage Diseases/surgery , Cartilage, Articular/surgery , Knee Injuries/surgery , Knee Joint/surgery , Mesenchymal Stem Cell Transplantation/methods , Adolescent , Adult , Female , Follow-Up Studies , Humans , Male , Tissue Scaffolds , Treatment Outcome , Young Adult
8.
Adv Protein Chem Struct Biol ; 102: 147-79, 2016.
Article in English | MEDLINE | ID: mdl-26827605

ABSTRACT

Some successes have been achieved in the war on cancer over the past 30 years with recent efforts on protein kinase inhibitors. Nonetheless, we are still facing challenges due to cancer evolution. Cancers are complex and heterogeneous due to primary and secondary mutations, with phenotypic and molecular heterogeneity manifested among patients of a cancer, and within an individual patient throughout the disease course. Our understanding of cancer genomes has been facilitated by advances in omics and in bioinformatics technologies; major areas in cancer research are advancing in parallel on many fronts. Computational methods have been developed to decipher the molecular complexity of cancer and to identify driver mutations in cancers. Utilizing the identified driver mutations to develop effective therapy would require biological linkages from cellular context to clinical implication; for this purpose, computational mining of biomedical literature facilitates utilization of a huge volume of biomedical research data and knowledge. In addition, frontier technologies, such as genome editing technologies, are facilitating investigation of cancer mutations, and opening the door for developing novel treatments to treat diseases. We will review and highlight the challenges of treating cancers, which behave like moving targets due to mutation and evolution, and the current state-of-the-art research in the areas mentioned above.


Subject(s)
Computational Biology/methods , Neoplasms/genetics , Patient Care , Precision Medicine , Biomedical Research/trends , Data Mining/methods , Humans , Mutation
9.
Integr Biol (Camb) ; 7(8): 904-20, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25932872

ABSTRACT

Identification of signaling pathways that are functional in a specific biological context is a major challenge in systems biology, and could be instrumental to the study of complex diseases and various aspects of drug discovery. Recent approaches have attempted to combine gene expression data with prior knowledge of protein connectivity in the form of a PPI network, and employ computational methods to identify subsets of the protein-protein-interaction (PPI) network that are functional, based on the data at hand. However, the use of undirected networks limits the mechanistic insight that can be drawn, since it does not allow for following mechanistically signal transduction from one node to the next. To address this important issue, we used a directed, signaling network as a scaffold to represent protein connectivity, and implemented an Integer Linear Programming (ILP) formulation to model the rules of signal transduction from one node to the next in the network. We then optimized the structure of the network to best fit the gene expression data at hand. We illustrated the utility of ILP modeling with a case study of drug induced lung injury. We identified the modes of action of 200 lung toxic drugs based on their gene expression profiles and, subsequently, merged the drug specific pathways to construct a signaling network that captured the mechanisms underlying Drug Induced Lung Disease (DILD). We further demonstrated the predictive power and biological relevance of the DILD network by applying it to identify drugs with relevant pharmacological mechanisms for treating lung injury.


Subject(s)
Gene Expression Profiling/methods , Lung Injury/chemically induced , Lung Injury/metabolism , Lung/metabolism , Models, Biological , Proteome/metabolism , Drug-Related Side Effects and Adverse Reactions/etiology , Drug-Related Side Effects and Adverse Reactions/metabolism , Humans , Lung/drug effects , Metabolic Networks and Pathways/drug effects , Protein Interaction Mapping/methods , Signal Transduction/drug effects
10.
PLoS One ; 10(5): e0128411, 2015.
Article in English | MEDLINE | ID: mdl-26020784

ABSTRACT

Modeling of signal transduction pathways is instrumental for understanding cells' function. People have been tackling modeling of signaling pathways in order to accurately represent the signaling events inside cells' biochemical microenvironment in a way meaningful for scientists in a biological field. In this article, we propose a method to interrogate such pathways in order to produce cell-specific signaling models. We integrate available prior knowledge of protein connectivity, in a form of a Prior Knowledge Network (PKN) with phosphoproteomic data to construct predictive models of the protein connectivity of the interrogated cell type. Several computational methodologies focusing on pathways' logic modeling using optimization formulations or machine learning algorithms have been published on this front over the past few years. Here, we introduce a light and fast approach that uses a breadth-first traversal of the graph to identify the shortest pathways and score proteins in the PKN, fitting the dependencies extracted from the experimental design. The pathways are then combined through a heuristic formulation to produce a final topology handling inconsistencies between the PKN and the experimental scenarios. Our results show that the algorithm we developed is efficient and accurate for the construction of medium and large scale signaling networks. We demonstrate the applicability of the proposed approach by interrogating a manually curated interaction graph model of EGF/TNFA stimulation against made up experimental data. To avoid the possibility of erroneous predictions, we performed a cross-validation analysis. Finally, we validate that the introduced approach generates predictive topologies, comparable to the ILP formulation. Overall, an efficient approach based on graph theory is presented herein to interrogate protein-protein interaction networks and to provide meaningful biological insights.


Subject(s)
Algorithms , Epidermal Growth Factor/metabolism , Models, Statistical , Phosphoproteins/metabolism , Proteomics/statistics & numerical data , Tumor Necrosis Factor-alpha/metabolism , Computer Graphics , Epidermal Growth Factor/genetics , Gene Expression Regulation , Gene Regulatory Networks , Humans , Phosphoproteins/genetics , Protein Interaction Mapping , Protein Interaction Maps , Proteomics/methods , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , User-Computer Interface
11.
Mult Scler ; 21(2): 138-46, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25112814

ABSTRACT

The pathogenesis of multiple sclerosis (MS) involves alterations to multiple pathways and processes, which represent a significant challenge for developing more-effective therapies. Systems biology approaches that study pathway dysregulation should offer benefits by integrating molecular networks and dynamic models with current biological knowledge for understanding disease heterogeneity and response to therapy. In MS, abnormalities have been identified in several cytokine-signaling pathways, as well as those of other immune receptors. Among the downstream molecules implicated are Jak/Stat, NF-Kb, ERK1/3, p38 or Jun/Fos. Together, these data suggest that MS is likely to be associated with abnormalities in apoptosis/cell death, microglia activation, blood-brain barrier functioning, immune responses, cytokine production, and/or oxidative stress, although which pathways contribute to the cascade of damage and can be modulated remains an open question. While current MS drugs target some of these pathways, others remain untouched. Here, we propose a pragmatic systems analysis approach that involves the large-scale extraction of processes and pathways relevant to MS. These data serve as a scaffold on which computational modeling can be performed to identify disease subgroups based on the contribution of different processes. Such an analysis, targeting these relevant MS-signaling pathways, offers the opportunity to accelerate the development of novel individual or combination therapies.


Subject(s)
Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Drug Discovery , Humans
12.
Bioinformatics ; 31(4): 484-91, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25294919

ABSTRACT

MOTIVATION: Animal models are important tools in drug discovery and for understanding human biology in general. However, many drugs that initially show promising results in rodents fail in later stages of clinical trials. Understanding the commonalities and differences between human and rat cell signaling networks can lead to better experimental designs, improved allocation of resources and ultimately better drugs. RESULTS: The sbv IMPROVER Species-Specific Network Inference challenge was designed to use the power of the crowds to build two species-specific cell signaling networks given phosphoproteomics, transcriptomics and cytokine data generated from NHBE and NRBE cells exposed to various stimuli. A common literature-inspired reference network with 220 nodes and 501 edges was also provided as prior knowledge from which challenge participants could add or remove edges but not nodes. Such a large network inference challenge not based on synthetic simulations but on real data presented unique difficulties in scoring and interpreting the results. Because any prior knowledge about the networks was already provided to the participants for reference, novel ways for scoring and aggregating the results were developed. Two human and rat consensus networks were obtained by combining all the inferred networks. Further analysis showed that major signaling pathways were conserved between the two species with only isolated components diverging, as in the case of ribosomal S6 kinase RPS6KA1. Overall, the consensus between inferred edges was relatively high with the exception of the downstream targets of transcription factors, which seemed more difficult to predict. CONTACT: ebilal@us.ibm.com or gustavo@us.ibm.com. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Algorithms , Crowdsourcing , Cytokines/metabolism , Gene Expression Profiling , Gene Regulatory Networks , Phosphoproteins/metabolism , Software , Systems Biology/methods , Animals , Bronchi/cytology , Bronchi/metabolism , Cell Communication , Cells, Cultured , Databases, Factual , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation , Humans , Models, Animal , Oligonucleotide Array Sequence Analysis , Phosphorylation , Rats , Signal Transduction , Species Specificity
13.
Drug Discov Today ; 19(4): 425-32, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24141136

ABSTRACT

Several important aspects of the drug discovery process, including target identification, mechanism of action determination and biomarker identification as well as drug repositioning, require complete understanding of the effects of drugs on protein phosphorylation in relevant biological systems. Novel high-throughput phosphoproteomic technologies can be employed to measure these phosphorylation events. In this review, we describe the advantages and limitations of state-of-the-art phosphoproteomic approaches such as mass spectrometry and antibody-based technologies in terms of sample and data throughput as well as data quality. We then discuss how datasets from each technology can be analyzed and how the results can be and have been applied to advance different aspects of the drug discovery process.


Subject(s)
Drug Discovery , Phosphoproteins/metabolism , Proteomics , Drug Repositioning , Humans , Mass Spectrometry , Precision Medicine , Protein Array Analysis
14.
Sci Data ; 1: 140009, 2014.
Article in English | MEDLINE | ID: mdl-25977767

ABSTRACT

The biological responses to external cues such as drugs, chemicals, viruses and hormones, is an essential question in biomedicine and in the field of toxicology, and cannot be easily studied in humans. Thus, biomedical research has continuously relied on animal models for studying the impact of these compounds and attempted to 'translate' the results to humans. In this context, the SBV IMPROVER (Systems Biology Verification for Industrial Methodology for PROcess VErification in Research) collaborative initiative, which uses crowd-sourcing techniques to address fundamental questions in systems biology, invited scientists to deploy their own computational methodologies to make predictions on species translatability. A multi-layer systems biology dataset was generated that was comprised of phosphoproteomics, transcriptomics and cytokine data derived from normal human (NHBE) and rat (NRBE) bronchial epithelial cells exposed in parallel to more than 50 different stimuli under identical conditions. The present manuscript describes in detail the experimental settings, generation, processing and quality control analysis of the multi-layer omics dataset accessible in public repositories for further intra- and inter-species translation studies.


Subject(s)
Bronchi/metabolism , Cytokines , Epithelial Cells/metabolism , Proteomics , Transcriptome , Animals , Bronchi/cytology , Cytokines/metabolism , Humans , Models, Animal , Rats , Systems Biology/methods , Translational Research, Biomedical
15.
PLoS Comput Biol ; 9(9): e1003204, 2013.
Article in English | MEDLINE | ID: mdl-24039561

ABSTRACT

Cross-referencing experimental data with our current knowledge of signaling network topologies is one central goal of mathematical modeling of cellular signal transduction networks. We present a new methodology for data-driven interrogation and training of signaling networks. While most published methods for signaling network inference operate on Bayesian, Boolean, or ODE models, our approach uses integer linear programming (ILP) on interaction graphs to encode constraints on the qualitative behavior of the nodes. These constraints are posed by the network topology and their formulation as ILP allows us to predict the possible qualitative changes (up, down, no effect) of the activation levels of the nodes for a given stimulus. We provide four basic operations to detect and remove inconsistencies between measurements and predicted behavior: (i) find a topology-consistent explanation for responses of signaling nodes measured in a stimulus-response experiment (if none exists, find the closest explanation); (ii) determine a minimal set of nodes that need to be corrected to make an inconsistent scenario consistent; (iii) determine the optimal subgraph of the given network topology which can best reflect measurements from a set of experimental scenarios; (iv) find possibly missing edges that would improve the consistency of the graph with respect to a set of experimental scenarios the most. We demonstrate the applicability of the proposed approach by interrogating a manually curated interaction graph model of EGFR/ErbB signaling against a library of high-throughput phosphoproteomic data measured in primary hepatocytes. Our methods detect interactions that are likely to be inactive in hepatocytes and provide suggestions for new interactions that, if included, would significantly improve the goodness of fit. Our framework is highly flexible and the underlying model requires only easily accessible biological knowledge. All related algorithms were implemented in a freely available toolbox SigNetTrainer making it an appealing approach for various applications.


Subject(s)
Computer Graphics , Signal Transduction , Models, Biological
16.
Biopharm Drug Dispos ; 34(9): 477-88, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23983165

ABSTRACT

Computational modeling has been adopted in all aspects of drug research and development, from the early phases of target identification and drug discovery to the late-stage clinical trials. The different questions addressed during each stage of drug R&D has led to the emergence of different modeling methodologies. In the research phase, systems biology couples experimental data with elaborate computational modeling techniques to capture lifecycle and effector cellular functions (e.g. metabolism, signaling, transcription regulation, protein synthesis and interaction) and integrates them in quantitative models. These models are subsequently used in various ways, i.e. to identify new targets, generate testable hypotheses, gain insights on the drug's mode of action (MOA), translate preclinical findings, and assess the potential of clinical drug efficacy and toxicity. In the development phase, pharmacokinetic/pharmacodynamic (PK/PD) modeling is the established way to determine safe and efficacious doses for testing at increasingly larger, and more pertinent to the target indication, cohorts of subjects. First, the relationship between drug input and its concentration in plasma is established. Second, the relationship between this concentration and desired or undesired PD responses is ascertained. Recognizing that the interface of systems biology with PK/PD will facilitate drug development, systems pharmacology came into existence, combining methods from PK/PD modeling and systems engineering explicitly to account for the implicated mechanisms of the target system in the study of drug-target interactions. Herein, a number of popular system biology methodologies are discussed, which could be leveraged within a systems pharmacology framework to address major issues in drug development.


Subject(s)
Drug Discovery , Models, Biological , Pharmacology, Clinical , Systems Biology , Humans , Pharmacokinetics
17.
Methods Mol Biol ; 930: 179-214, 2013.
Article in English | MEDLINE | ID: mdl-23086842

ABSTRACT

Mathematical models are useful tools for understanding protein signaling networks because they provide an integrated view of pharmacological and toxicological processes at the molecular level. Here we describe an approach previously introduced based on logic modeling to generate cell-specific, mechanistic and predictive models of signal transduction. Models are derived from a network encoding prior knowledge that is trained to signaling data, and can be either binary (based on Boolean logic) or quantitative (using a recently developed formalism, constrained fuzzy logic). The approach is implemented in the freely available tool CellNetOptimizer (CellNOpt). We explain the process CellNOpt uses to train a prior knowledge network to data and illustrate its application with a toy example as well as a realistic case describing signaling networks in the HepG2 liver cancer cell line.


Subject(s)
Algorithms , Fuzzy Logic , Models, Biological , Organ Specificity , Signal Transduction , Computer Simulation , Hep G2 Cells , Humans
18.
Article in English | MEDLINE | ID: mdl-25729777

ABSTRACT

Hepatocellular Carcinoma (HCC) is one of the leading causes of death worldwide, with only a handful of treatments effective in unresectable HCC. Most of the clinical trials for HCC using new generation interventions (drug-targeted therapies) have poor efficacy whereas just a few of them show some promising clinical outcomes [1]. This is amongst the first studies where the mode of action of some of the compounds extensively used in clinical trials is interrogated on the phosphoproteomic level, in an attempt to build predictive models for clinical efficacy. Signaling data are combined with previously published gene expression and clinical data within a consistent framework that identifies drug effects on the phosphoproteomic level and translates them to the gene expression level. The interrogated drugs are then correlated with genes differentially expressed in normal versus tumor tissue, and genes predictive of patient survival. Although the number of clinical trial results considered is small, our approach shows potential for discerning signaling activities that may help predict drug efficacy for HCC.

19.
PLoS One ; 7(11): e50085, 2012.
Article in English | MEDLINE | ID: mdl-23226239

ABSTRACT

Modeling of signal transduction pathways plays a major role in understanding cells' function and predicting cellular response. Mathematical formalisms based on a logic formalism are relatively simple but can describe how signals propagate from one protein to the next and have led to the construction of models that simulate the cells response to environmental or other perturbations. Constrained fuzzy logic was recently introduced to train models to cell specific data to result in quantitative pathway models of the specific cellular behavior. There are two major issues in this pathway optimization: i) excessive CPU time requirements and ii) loosely constrained optimization problem due to lack of data with respect to large signaling pathways. Herein, we address both issues: the former by reformulating the pathway optimization as a regular nonlinear optimization problem; and the latter by enhanced algorithms to pre/post-process the signaling network to remove parts that cannot be identified given the experimental conditions. As a case study, we tackle the construction of cell type specific pathways in normal and transformed hepatocytes using medium and large-scale functional phosphoproteomic datasets. The proposed Non Linear Programming (NLP) formulation allows for fast optimization of signaling topologies by combining the versatile nature of logic modeling with state of the art optimization algorithms.


Subject(s)
Algorithms , Hepatocytes/metabolism , Models, Biological , Nonlinear Dynamics , Phosphoproteins/metabolism , Signal Transduction , Computer Simulation , Fuzzy Logic , Humans , Information Theory , Primary Cell Culture , Protein Interaction Mapping , Proteome
20.
Mol Biosyst ; 8(5): 1571-84, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22446821

ABSTRACT

Construction of large and cell-specific signaling pathways is essential to understand information processing under normal and pathological conditions. On this front, gene-based approaches offer the advantage of large pathway exploration whereas phosphoproteomic approaches offer a more reliable view of pathway activities but are applicable to small pathway sizes. In this paper, we demonstrate an experimentally adaptive approach to construct large signaling pathways from phosphoproteomic data within a 3-day time frame. Our approach--taking advantage of the fast turnaround time of the xMAP technology--is carried out in four steps: (i) screen optimal pathway inducers, (ii) select the responsive ones, (iii) combine them in a combinatorial fashion to construct a phosphoproteomic dataset, and (iv) optimize a reduced generic pathway via an Integer Linear Programming formulation. As a case study, we uncover novel players and their corresponding pathways in primary human hepatocytes by interrogating the signal transduction downstream of 81 receptors of interest and constructing a detailed model for the responsive part of the network comprising 177 species (of which 14 are measured) and 365 interactions.


Subject(s)
Computational Biology/methods , Phosphoproteins/metabolism , Proteomics/methods , Signal Transduction , Cluster Analysis , Cytokines/pharmacology , Feedback, Physiological/drug effects , Humans , Ligands , Programming, Linear , Signal Transduction/drug effects , Statistics as Topic
SELECTION OF CITATIONS
SEARCH DETAIL
...