Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Methods Mol Biol ; 1939: 181-198, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30848462

RESUMO

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.


Assuntos
Biologia Computacional/métodos , Descoberta de Drogas/métodos , Big Data , Bases de Dados Factuais , Humanos , Medicina de Precisão/métodos , Programação Linear , Mapas de Interação de Proteínas/efeitos dos fármacos , Síndrome de Stevens-Johnson/etiologia , Transcriptoma/efeitos dos fármacos
2.
CPT Pharmacometrics Syst Pharmacol ; 7(3): 166-174, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29341478

RESUMO

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.


Assuntos
Cardiomiopatias/induzido quimicamente , Doxorrubicina/farmacologia , Redes Reguladoras de Genes/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Cardiomiopatias/metabolismo , Cardiotoxicidade , Biologia Computacional , Doxorrubicina/efeitos adversos , Humanos , Modelos Biológicos , Terapia de Alvo Molecular , Análise de Regressão , Pesquisa Translacional Biomédica
4.
Adv Protein Chem Struct Biol ; 102: 147-79, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26827605

RESUMO

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.


Assuntos
Biologia Computacional/métodos , Neoplasias/genética , Assistência ao Paciente , Medicina de Precisão , Pesquisa Biomédica/tendências , Mineração de Dados/métodos , Humanos , Mutação
5.
Integr Biol (Camb) ; 7(8): 904-20, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25932872

RESUMO

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.


Assuntos
Perfilação da Expressão Gênica/métodos , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/metabolismo , Pulmão/metabolismo , Modelos Biológicos , Proteoma/metabolismo , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/etiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/metabolismo , Humanos , Pulmão/efeitos dos fármacos , Redes e Vias Metabólicas/efeitos dos fármacos , Mapeamento de Interação de Proteínas/métodos , Transdução de Sinais/efeitos dos fármacos
6.
PLoS One ; 10(5): e0128411, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26020784

RESUMO

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.


Assuntos
Algoritmos , Fator de Crescimento Epidérmico/metabolismo , Modelos Estatísticos , Fosfoproteínas/metabolismo , Proteômica/estatística & dados numéricos , Fator de Necrose Tumoral alfa/metabolismo , Gráficos por Computador , Fator de Crescimento Epidérmico/genética , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Fosfoproteínas/genética , Mapeamento de Interação de Proteínas , Mapas de Interação de Proteínas , Proteômica/métodos , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética , Interface Usuário-Computador
7.
Mult Scler ; 21(2): 138-46, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25112814

RESUMO

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.


Assuntos
Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Descoberta de Drogas , Humanos
8.
Bioinformatics ; 31(4): 484-91, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25294919

RESUMO

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.


Assuntos
Algoritmos , Crowdsourcing , Citocinas/metabolismo , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Fosfoproteínas/metabolismo , Software , Biologia de Sistemas/métodos , Animais , Brônquios/citologia , Brônquios/metabolismo , Comunicação Celular , Células Cultivadas , Bases de Dados Factuais , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Humanos , Modelos Animais , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Ratos , Transdução de Sinais , Especificidade da Espécie
9.
Drug Discov Today ; 19(4): 425-32, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24141136

RESUMO

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.


Assuntos
Descoberta de Drogas , Fosfoproteínas/metabolismo , Proteômica , Reposicionamento de Medicamentos , Humanos , Espectrometria de Massas , Medicina de Precisão , Análise Serial de Proteínas
10.
Sci Data ; 1: 140009, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25977767

RESUMO

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.


Assuntos
Brônquios/metabolismo , Citocinas , Células Epiteliais/metabolismo , Proteômica , Transcriptoma , Animais , Brônquios/citologia , Citocinas/metabolismo , Humanos , Modelos Animais , Ratos , Biologia de Sistemas/métodos , Pesquisa Translacional Biomédica
11.
PLoS Comput Biol ; 9(9): e1003204, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24039561

RESUMO

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.


Assuntos
Gráficos por Computador , Transdução de Sinais , Modelos Biológicos
12.
Biopharm Drug Dispos ; 34(9): 477-88, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23983165

RESUMO

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.


Assuntos
Descoberta de Drogas , Modelos Biológicos , Farmacologia Clínica , Biologia de Sistemas , Humanos , Farmacocinética
13.
Artigo em Inglês | MEDLINE | ID: mdl-25729777

RESUMO

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.

14.
PLoS One ; 7(11): e50085, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23226239

RESUMO

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.


Assuntos
Algoritmos , Hepatócitos/metabolismo , Modelos Biológicos , Dinâmica não Linear , Fosfoproteínas/metabolismo , Transdução de Sinais , Simulação por Computador , Lógica Fuzzy , Humanos , Teoria da Informação , Cultura Primária de Células , Mapeamento de Interação de Proteínas , Proteoma
15.
Mol Biosyst ; 8(5): 1571-84, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22446821

RESUMO

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.


Assuntos
Biologia Computacional/métodos , Fosfoproteínas/metabolismo , Proteômica/métodos , Transdução de Sinais , Análise por Conglomerados , Citocinas/farmacologia , Retroalimentação Fisiológica/efeitos dos fármacos , Humanos , Ligantes , Programação Linear , Transdução de Sinais/efeitos dos fármacos , Estatística como Assunto
16.
BMC Syst Biol ; 5: 107, 2011 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-21729292

RESUMO

BACKGROUND: Signalling pathways are the cornerstone on understanding cell function and predicting cell behavior. Recently, logical models of canonical pathways have been optimised with high-throughput phosphoproteomic data to construct cell-type specific pathways. However, less is known on how signalling pathways can be linked to a cellular response such as cell growth, death, cytokine secretion, or transcriptional activity. RESULTS: In this work, we measure the signalling activity (phosphorylation levels) and phenotypic behavior (cytokine secretion) of normal and cancer hepatocytes treated with a combination of cytokines and inhibitors. Using the two datasets, we construct "extended" pathways that integrate intracellular activity with cellular responses using a hybrid logical/data-driven computational approach. Boolean logic is used whenever a priori knowledge is accessible (i.e., construction of canonical pathways), whereas a data-driven approach is used for linking cellular behavior to signalling activity via non-canonical edges. The extended pathway is subsequently optimised to fit signalling and behavioural data using an Integer Linear Programming formulation. As a result, we are able to construct maps of primary and transformed hepatocytes downstream of 7 receptors that are capable of explaining the secretion of 22 cytokines. CONCLUSIONS: We developed a method for constructing extended pathways that start at the receptor level and via a complex intracellular signalling pathway identify those mechanisms that drive cellular behaviour. Our results constitute a proof-of-principle for construction of "extended pathways" that are capable of linking pathway activity to diverse responses such as growth, death, differentiation, gene expression, or cytokine secretion.


Assuntos
Lógica , Modelos Biológicos , Transdução de Sinais , Calibragem , Linhagem Celular Tumoral , Bases de Dados Factuais , Humanos , Modelos Lineares , Reprodutibilidade dos Testes
17.
Artigo em Inglês | MEDLINE | ID: mdl-22255146

RESUMO

Chondrocytes, the only cell type in articular cartilage, are responsible for maintaining the composition of cartilage extracellular matrix (ECM) through a complex interplay of anabolic and catabolic stimuli. Although understanding the way chondrocytes respond to stimuli is of utmost importance for shedding light into the etiology of joint diseases, an integrative approach to studying their signaling transduction mechanisms is yet to be introduced. Herein, we propose an approach that combines high throughput proteomic measurements and state of the art optimization algorithms to construct a predictive model of chondrocyte signaling network, downstream of 78 receptors of interest.


Assuntos
Cartilagem Articular/metabolismo , Modelos Biológicos , Transdução de Sinais , Humanos , Fosfoproteínas/metabolismo , Proteômica
18.
Artigo em Inglês | MEDLINE | ID: mdl-21096084

RESUMO

Construction of signaling pathway maps and identification of drug effects are major challenge for pharmaceutical industries. Signaling maps are usually obtained from manual literature search, automated text mining algorithms, or canonical pathway databases (i.e. Reactome, KEGG, STKE, Pathway Studio, Ingenuity etc.) and in some cases they are used in combination with gene expression or mass spec data in an effort to create pathways specific to cell types or diseases. Our approach combines computational models with novel multicombinatorial high-throughput phosphoproteomic data for the functional analysis of signalling networks in mammalian cells. On the experimental front, we subject the cells with hundreds of co-treatment with a diverse set of ligands and inhibitors and we measure phosphorylation events on key signaling proteins using the xMAP technology. On the computational front, we create pathway maps that are cell type specific by fitting our phosphoprotein dataset into generic signaling maps via an Integer Linear programming formulation. To identify drug effects, we monitor the differences of topologies created with and without the presence of drug. In the present work, we use this approach to identify the effects of Nilotinib, a well known anti-cancer drug.


Assuntos
Antineoplásicos/farmacologia , Biologia Computacional/métodos , Pirimidinas/farmacologia , Algoritmos , Mineração de Dados , Células Hep G2 , Humanos , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
19.
PLoS Comput Biol ; 5(12): e1000591, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19997482

RESUMO

Understanding the mechanisms of cell function and drug action is a major endeavor in the pharmaceutical industry. Drug effects are governed by the intrinsic properties of the drug (i.e., selectivity and potency) and the specific signaling transduction network of the host (i.e., normal vs. diseased cells). Here, we describe an unbiased, phosphoproteomic-based approach to identify drug effects by monitoring drug-induced topology alterations. With our proposed method, drug effects are investigated under diverse stimulations of the signaling network. Starting with a generic pathway made of logical gates, we build a cell-type specific map by constraining it to fit 13 key phopshoprotein signals under 55 experimental conditions. Fitting is performed via an Integer Linear Program (ILP) formulation and solution by standard ILP solvers; a procedure that drastically outperforms previous fitting schemes. Then, knowing the cell's topology, we monitor the same key phosphoprotein signals under the presence of drug and we re-optimize the specific map to reveal drug-induced topology alterations. To prove our case, we make a topology for the hepatocytic cell-line HepG2 and we evaluate the effects of 4 drugs: 3 selective inhibitors for the Epidermal Growth Factor Receptor (EGFR) and a non-selective drug. We confirm effects easily predictable from the drugs' main target (i.e., EGFR inhibitors blocks the EGFR pathway) but we also uncover unanticipated effects due to either drug promiscuity or the cell's specific topology. An interesting finding is that the selective EGFR inhibitor Gefitinib inhibits signaling downstream the Interleukin-1alpha (IL1alpha) pathway; an effect that cannot be extracted from binding affinity-based approaches. Our method represents an unbiased approach to identify drug effects on small to medium size pathways which is scalable to larger topologies with any type of signaling interventions (small molecules, RNAi, etc). The method can reveal drug effects on pathways, the cornerstone for identifying mechanisms of drug's efficacy.


Assuntos
Modelos Biológicos , Farmacologia/métodos , Fosfoproteínas/metabolismo , Proteômica/métodos , Transdução de Sinais/efeitos dos fármacos , Algoritmos , Antineoplásicos/farmacologia , Bases de Dados de Proteínas , Células Hep G2 , Humanos , Reprodutibilidade dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA