Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Toxicol Sci ; 198(1): 14-30, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38015832

ABSTRACT

Drug-induced liver injury (DILI) remains the main reason for drug development attritions largely due to poor mechanistic understanding. Toxicogenomic to interrogate the mechanism of DILI has been broadly performed. Gene coregulation network-based transcriptome analysis is a bioinformatics approach that potentially contributes to improve mechanistic interpretation of toxicogenomic data. Here we performed an extensive concentration time course response-toxicogenomic study in the HepG2 cell line exposed to 20 DILI compounds, 7 reference compounds for stress response pathways, and 10 agonists for cytokines and growth factor receptors. We performed whole transcriptome targeted RNA sequencing to more than 500 conditions and applied weighted gene coregulated network analysis to the transcriptomics data followed by the identification of gene coregulated networks (modules) that were strongly modulated upon the exposure of DILI compounds. Preservation analysis on the module responses of HepG2 and PHH demonstrated highly preserved adaptive stress response gene coregulated networks. We correlated gene coregulated networks with cell death onset and causal relationships of 67 critical target genes of these modules with the onset of cell death was evaluated using RNA interference screening. We identified GTPBP2, HSPA1B, IRF1, SIRT1, and TSC22D3 as essential modulators of DILI compound-induced cell death. These genes were also induced by DILI compounds in PHH. Altogether, we demonstrate the application of large transcriptome datasets combined with network-based analysis and biological validation to uncover the candidate determinants of DILI.


Subject(s)
Chemical and Drug Induced Liver Injury , Transcriptome , Humans , Hep G2 Cells , Gene Expression Profiling , Gene Regulatory Networks , Chemical and Drug Induced Liver Injury/genetics
2.
Genome Biol ; 23(1): 97, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35422018

ABSTRACT

The advancement of highly multiplexed spatial technologies requires scalable methods that can leverage spatial information. We present MISTy, a flexible, scalable, and explainable machine learning framework for extracting relationships from any spatial omics data, from dozens to thousands of measured markers. MISTy builds multiple views focusing on different spatial or functional contexts to dissect different effects. We evaluated MISTy on in silico and breast cancer datasets measured by imaging mass cytometry and spatial transcriptomics. We estimated structural and functional interactions coming from different spatial contexts in breast cancer and demonstrated how to relate MISTy's results to clinical features.


Subject(s)
Breast Neoplasms , Machine Learning , Breast Neoplasms/genetics , Female , Humans
3.
Mol Syst Biol ; 17(10): e10402, 2021 10.
Article in English | MEDLINE | ID: mdl-34661974

ABSTRACT

Recent technological developments allow us to measure the status of dozens of proteins in individual cells. This opens the way to understand the heterogeneity of complex multi-signaling networks across cells and cell types, with important implications to understand and treat diseases such as cancer. These technologies are, however, limited to proteins for which antibodies are available and are fairly costly, making predictions of new markers and of existing markers under new conditions a valuable alternative. To assess our capacity to make such predictions and boost further methodological development, we organized the Single Cell Signaling in Breast Cancer DREAM challenge. We used a mass cytometry dataset, covering 36 markers in over 4,000 conditions totaling 80 million single cells across 67 breast cancer cell lines. Through four increasingly difficult subchallenges, the participants predicted missing markers, new conditions, and the time-course response of single cells to stimuli in the presence and absence of kinase inhibitors. The challenge results show that despite the stochastic nature of signal transduction in single cells, the signaling events are tightly controlled and machine learning methods can accurately predict new experimental data.


Subject(s)
Breast Neoplasms , Signal Transduction , Breast Neoplasms/genetics , Female , Humans , Machine Learning , Proteins
4.
Arch Toxicol ; 95(8): 2691-2718, 2021 08.
Article in English | MEDLINE | ID: mdl-34151400

ABSTRACT

5-Fluorouracil (5-FU) is a widely used chemotherapeutical that induces acute toxicity in the small and large intestine of patients. Symptoms can be severe and lead to the interruption of cancer treatments. However, there is limited understanding of the molecular mechanisms underlying 5-FU-induced intestinal toxicity. In this study, well-established 3D organoid models of human colon and small intestine (SI) were used to characterize 5-FU transcriptomic and metabolomic responses. Clinically relevant 5-FU concentrations for in vitro testing in organoids were established using physiologically based pharmacokinetic simulation of dosing regimens recommended for cancer patients, resulting in exposures to 10, 100 and 1000 µM. After treatment, different measurements were performed: cell viability and apoptosis; image analysis of cell morphological changes; RNA sequencing; and metabolome analysis of supernatant from organoids cultures. Based on analysis of the differentially expressed genes, the most prominent molecular pathways affected by 5-FU included cell cycle, p53 signalling, mitochondrial ATP synthesis and apoptosis. Short time-series expression miner demonstrated tissue-specific mechanisms affected by 5-FU, namely biosynthesis and transport of small molecules, and mRNA translation for colon; cell signalling mediated by Rho GTPases and fork-head box transcription factors for SI. Metabolomic analysis showed that in addition to the effects on TCA cycle and oxidative stress in both organoids, tissue-specific metabolic alterations were also induced by 5-FU. Multi-omics integration identified transcription factor E2F1, a regulator of cell cycle and apoptosis, as the best key node across all samples. These results provide new insights into 5-FU toxicity mechanisms and underline the relevance of human organoid models in the safety assessment in drug development.


Subject(s)
Colon/drug effects , Fluorouracil/toxicity , Intestine, Small/drug effects , Models, Biological , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/toxicity , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Survival/drug effects , Colon/pathology , Dose-Response Relationship, Drug , Female , Fluorouracil/administration & dosage , Fluorouracil/pharmacokinetics , Humans , Intestine, Small/pathology , Male , Metabolomics , Organoids/drug effects , Oxidative Stress/drug effects , Transcriptome
5.
Cell Syst ; 12(5): 401-418.e12, 2021 05 19.
Article in English | MEDLINE | ID: mdl-33932331

ABSTRACT

One goal of precision medicine is to tailor effective treatments to patients' specific molecular markers of disease. Here, we used mass cytometry to characterize the single-cell signaling landscapes of 62 breast cancer cell lines and five lines from healthy tissue. We quantified 34 markers in each cell line upon stimulation by the growth factor EGF in the presence or absence of five kinase inhibitors. These data-on more than 80 million single cells from 4,000 conditions-were used to fit mechanistic signaling network models that provide insight into how cancer cells process information. Our dynamic single-cell-based models accurately predicted drug sensitivity and identified genomic features associated with drug sensitivity, including a missense mutation in DDIT3 predictive of PI3K-inhibition sensitivity. We observed similar trends in genotype-drug sensitivity associations in patient-derived xenograft mouse models. This work provides proof of principle that patient-specific single-cell measurements and modeling could inform effective precision medicine strategies.


Subject(s)
Breast Neoplasms , Pharmaceutical Preparations , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Female , Genomics , Humans , Mice , Signal Transduction
6.
Biochem Pharmacol ; 190: 114591, 2021 08.
Article in English | MEDLINE | ID: mdl-33957093

ABSTRACT

Drug-induced liver injury (DILI) is the most prevalent adversity encountered in drug development and clinical settings leading to urgent needs to understand the underlying mechanisms. In this study, we have systematically investigated the dynamics of the activation of cellular stress response pathways and cell death outcomes upon exposure of a panel of liver toxicants using live cell imaging of fluorescent reporter cell lines. We established a comprehensive temporal dynamic response profile of a large set of BAC-GFP HepG2 cell lines representing the following components of stress signaling: i) unfolded protein response (UPR) [ATF4, XBP1, BIP and CHOP]; ii) oxidative stress [NRF2, SRXN1, HMOX1]; iii) DNA damage [P53, P21, BTG2, MDM2]; and iv) NF-κB pathway [A20, ICAM1]. We quantified the single cell GFP expression as a surrogate for endogenous protein expression using live cell imaging over > 60 h upon exposure to 14 DILI compounds at multiple concentrations. Using logic-based ordinary differential equation (Logic-ODE), we modelled the dynamic profiles of the different stress responses and extracted specific descriptors potentially predicting the progressive outcomes. We identified the activation of ATF4-CHOP axis of the UPR as the key pathway showing the highest correlation with cell death upon DILI compound perturbation. Knocking down main components of the UPR provided partial protection from compound-induced cytotoxicity, indicating a complex interplay among UPR components as well as other stress pathways. Our results suggest that a systematic analysis of the temporal dynamics of ATF4-CHOP axis activation can support the identification of DILI risk for new candidate drugs.


Subject(s)
Activating Transcription Factor 4/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Models, Biological , Oxidative Stress/physiology , Single-Cell Analysis/methods , Transcription Factor CHOP/metabolism , Chemical and Drug Induced Liver Injury/pathology , Forecasting , Hep G2 Cells , Humans , Unfolded Protein Response/drug effects , Unfolded Protein Response/physiology
7.
Mol Syst Biol ; 17(3): e9923, 2021 03.
Article in English | MEDLINE | ID: mdl-33749993

ABSTRACT

Molecular knowledge of biological processes is a cornerstone in omics data analysis. Applied to single-cell data, such analyses provide mechanistic insights into individual cells and their interactions. However, knowledge of intercellular communication is scarce, scattered across resources, and not linked to intracellular processes. To address this gap, we combined over 100 resources covering interactions and roles of proteins in inter- and intracellular signaling, as well as transcriptional and post-transcriptional regulation. We added protein complex information and annotations on function, localization, and role in diseases for each protein. The resource is available for human, and via homology translation for mouse and rat. The data are accessible via OmniPath's web service (https://omnipathdb.org/), a Cytoscape plug-in, and packages in R/Bioconductor and Python, providing access options for computational and experimental scientists. We created workflows with tutorials to facilitate the analysis of cell-cell interactions and affected downstream intracellular signaling processes. OmniPath provides a single access point to knowledge spanning intra- and intercellular processes for data analysis, as we demonstrate in applications studying SARS-CoV-2 infection and ulcerative colitis.


Subject(s)
COVID-19/metabolism , Colitis, Ulcerative/metabolism , Computational Biology/methods , Proteins/metabolism , Signal Transduction , Animals , Cell Communication , Colitis, Ulcerative/pathology , Databases, Factual , Enzymes/metabolism , Humans , Mice , Protein Processing, Post-Translational , Proteins/genetics , Rats , Single-Cell Analysis , Software , Workflow
8.
Mol Syst Biol ; 17(1): e9730, 2021 01.
Article in English | MEDLINE | ID: mdl-33502086

ABSTRACT

Multi-omics datasets can provide molecular insights beyond the sum of individual omics. Various tools have been recently developed to integrate such datasets, but there are limited strategies to systematically extract mechanistic hypotheses from them. Here, we present COSMOS (Causal Oriented Search of Multi-Omics Space), a method that integrates phosphoproteomics, transcriptomics, and metabolomics datasets. COSMOS combines extensive prior knowledge of signaling, metabolic, and gene regulatory networks with computational methods to estimate activities of transcription factors and kinases as well as network-level causal reasoning. COSMOS provides mechanistic hypotheses for experimental observations across multi-omics datasets. We applied COSMOS to a dataset comprising transcriptomics, phosphoproteomics, and metabolomics data from healthy and cancerous tissue from eleven clear cell renal cell carcinoma (ccRCC) patients. COSMOS was able to capture relevant crosstalks within and between multiple omics layers, such as known ccRCC drug targets. We expect that our freely available method will be broadly useful to extract mechanistic insights from multi-omics studies.


Subject(s)
Carcinoma, Renal Cell/genetics , Computational Biology/methods , Gene Regulatory Networks , Kidney Neoplasms/genetics , Carcinoma, Renal Cell/metabolism , Case-Control Studies , Gene Expression Profiling , Humans , Kidney Neoplasms/metabolism , Metabolomics , Phosphoproteins
9.
Life Sci Alliance ; 3(11)2020 11.
Article in English | MEDLINE | ID: mdl-32972997

ABSTRACT

Single-cell RNA-sequencing (scRNAseq) technologies are rapidly evolving. Although very informative, in standard scRNAseq experiments, the spatial organization of the cells in the tissue of origin is lost. Conversely, spatial RNA-seq technologies designed to maintain cell localization have limited throughput and gene coverage. Mapping scRNAseq to genes with spatial information increases coverage while providing spatial location. However, methods to perform such mapping have not yet been benchmarked. To fill this gap, we organized the DREAM Single-Cell Transcriptomics challenge focused on the spatial reconstruction of cells from the Drosophila embryo from scRNAseq data, leveraging as silver standard, genes with in situ hybridization data from the Berkeley Drosophila Transcription Network Project reference atlas. The 34 participating teams used diverse algorithms for gene selection and location prediction, while being able to correctly localize clusters of cells. Selection of predictor genes was essential for this task. Predictor genes showed a relatively high expression entropy, high spatial clustering and included prominent developmental genes such as gap and pair-rule genes and tissue markers. Application of the top 10 methods to a zebra fish embryo dataset yielded similar performance and statistical properties of the selected genes than in the Drosophila data. This suggests that methods developed in this challenge are able to extract generalizable properties of genes that are useful to accurately reconstruct the spatial arrangement of cells in tissues.


Subject(s)
Computational Biology/methods , Gene Expression Profiling/methods , Single-Cell Analysis/methods , Spatial Analysis , Algorithms , Animals , Databases, Genetic , Drosophila/genetics , Forecasting/methods , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Sequence Analysis, RNA/methods , Transcriptome/genetics , Zebrafish/genetics
10.
Bioinformatics ; 36(16): 4523-4524, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32516357

ABSTRACT

SUMMARY: The molecular changes induced by perturbations such as drugs and ligands are highly informative of the intracellular wiring. Our capacity to generate large datasets is increasing steadily. A useful way to extract mechanistic insight from the data is by integrating them with a prior knowledge network of signalling to obtain dynamic models. CellNOpt is a collection of Bioconductor R packages for building logic models from perturbation data and prior knowledge of signalling networks. We have recently developed new components and refined the existing ones to keep up with the computational demand of increasingly large datasets, including (i) an efficient integer linear programming, (ii) a probabilistic logic implementation for semi-quantitative datasets, (iii) the integration of a stochastic Boolean simulator, (iv) a tool to identify missing links, (v) systematic post-hoc analyses and (vi) an R-Shiny tool to run CellNOpt interactively. AVAILABILITY AND IMPLEMENTATION: R-package(s): https://github.com/saezlab/cellnopt. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Signal Transduction , Software , Logic
11.
Bioinformatics ; 36(8): 2632-2633, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31886476

ABSTRACT

SUMMARY: Multiple databases provide valuable information about curated pathways and other resources that can be used to build and analyze networks. OmniPath combines 61 (and continuously growing) network resources into a comprehensive collection, with over 120 000 interactions. We present here the OmniPath App, a Cytoscape plugin to flexibly import data from OmniPath via a simple and intuitive interface. Thus, it makes possible to directly access the large body of high-quality knowledge provided by OmniPath within Cytoscape for inspection and further use with other tools. AVAILABILITY AND IMPLEMENTATION: The OmniPath App has been developed for Cytoscape 3 in the Java programing language. The latest source code and the plugin can be found at: https://github.com/saezlab/Omnipath_Cytoscape and http://apps.cytoscape.org/apps/omnipath, respectively. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Software , Databases, Factual
12.
Mol Cell ; 74(5): 1086-1102.e5, 2019 06 06.
Article in English | MEDLINE | ID: mdl-31101498

ABSTRACT

Kinase and phosphatase overexpression drives tumorigenesis and drug resistance. We previously developed a mass-cytometry-based single-cell proteomics approach that enables quantitative assessment of overexpression effects on cell signaling. Here, we applied this approach in a human kinome- and phosphatome-wide study to assess how 649 individually overexpressed proteins modulated cancer-related signaling in HEK293T cells in an abundance-dependent manner. Based on these data, we expanded the functional classification of human kinases and phosphatases and showed that the overexpression effects include non-catalytic roles. We detected 208 previously unreported signaling relationships. The signaling dynamics analysis indicated that the overexpression of ERK-specific phosphatases sustains proliferative signaling. This suggests a phosphatase-driven mechanism of cancer progression. Moreover, our analysis revealed a drug-resistant mechanism through which overexpression of tyrosine kinases, including SRC, FES, YES1, and BLK, induced MEK-independent ERK activation in melanoma A375 cells. These proteins could predict drug sensitivity to BRAF-MEK concurrent inhibition in cells carrying BRAF mutations.


Subject(s)
Carcinogenesis/genetics , Melanoma/genetics , Phosphoric Monoester Hydrolases/genetics , Phosphotransferases/genetics , Proto-Oncogene Proteins B-raf/genetics , Cell Proliferation/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Melanoma/enzymology , Melanoma/pathology , Mutation , Phosphorylation/genetics , Protein Kinase Inhibitors/pharmacology , Proteomics , Signal Transduction/drug effects
13.
BMC Syst Biol ; 11(1): 54, 2017 May 05.
Article in English | MEDLINE | ID: mdl-28476119

ABSTRACT

BACKGROUND: Kinetic models of biochemical systems usually consist of ordinary differential equations that have many unknown parameters. Some of these parameters are often practically unidentifiable, that is, their values cannot be uniquely determined from the available data. Possible causes are lack of influence on the measured outputs, interdependence among parameters, and poor data quality. Uncorrelated parameters can be seen as the key tuning knobs of a predictive model. Therefore, before attempting to perform parameter estimation (model calibration) it is important to characterize the subset(s) of identifiable parameters and their interplay. Once this is achieved, it is still necessary to perform parameter estimation, which poses additional challenges. METHODS: We present a methodology that (i) detects high-order relationships among parameters, and (ii) visualizes the results to facilitate further analysis. We use a collinearity index to quantify the correlation between parameters in a group in a computationally efficient way. Then we apply integer optimization to find the largest groups of uncorrelated parameters. We also use the collinearity index to identify small groups of highly correlated parameters. The results files can be visualized using Cytoscape, showing the identifiable and non-identifiable groups of parameters together with the model structure in the same graph. RESULTS: Our contributions alleviate the difficulties that appear at different stages of the identifiability analysis and parameter estimation process. We show how to combine global optimization and regularization techniques for calibrating medium and large scale biological models with moderate computation times. Then we evaluate the practical identifiability of the estimated parameters using the proposed methodology. The identifiability analysis techniques are implemented as a MATLAB toolbox called VisId, which is freely available as open source from GitHub ( https://github.com/gabora/visid ). CONCLUSIONS: Our approach is geared towards scalability. It enables the practical identifiability analysis of dynamic models of large size, and accelerates their calibration. The visualization tool allows modellers to detect parts that are problematic and need refinement or reformulation, and provides experimentalists with information that can be helpful in the design of new experiments.


Subject(s)
Models, Biological , Systems Biology , Arabidopsis/physiology , Circadian Clocks , Kinetics , Linear Models , Signal Transduction , Transforming Growth Factor beta/metabolism
14.
Bioinformatics ; 32(21): 3357-3359, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27378288

ABSTRACT

MOTIVATION: Many problems of interest in dynamic modeling and control of biological systems can be posed as non-linear optimization problems subject to algebraic and dynamic constraints. In the context of modeling, this is the case of, e.g. parameter estimation, optimal experimental design and dynamic flux balance analysis. In the context of control, model-based metabolic engineering or drug dose optimization problems can be formulated as (multi-objective) optimal control problems. Finding a solution to those problems is a very challenging task which requires advanced numerical methods. RESULTS: This work presents the AMIGO2 toolbox: the first multiplatform software tool that automatizes the solution of all those problems, offering a suite of state-of-the-art (multi-objective) global optimizers and advanced simulation approaches. AVAILABILITY AND IMPLEMENTATION: The toolbox and its documentation are available at: sites.google.com/site/amigo2toolbox CONTACT: ebalsa@iim.csic.esSupplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Software , Systems Biology , Algorithms , Animals , Humans , Metabolic Engineering , Models, Biological
15.
BMC Syst Biol ; 9: 74, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26515482

ABSTRACT

BACKGROUND: Dynamic modelling provides a systematic framework to understand function in biological systems. Parameter estimation in nonlinear dynamic models remains a very challenging inverse problem due to its nonconvexity and ill-conditioning. Associated issues like overfitting and local solutions are usually not properly addressed in the systems biology literature despite their importance. Here we present a method for robust and efficient parameter estimation which uses two main strategies to surmount the aforementioned difficulties: (i) efficient global optimization to deal with nonconvexity, and (ii) proper regularization methods to handle ill-conditioning. In the case of regularization, we present a detailed critical comparison of methods and guidelines for properly tuning them. Further, we show how regularized estimations ensure the best trade-offs between bias and variance, reducing overfitting, and allowing the incorporation of prior knowledge in a systematic way. RESULTS: We illustrate the performance of the presented method with seven case studies of different nature and increasing complexity, considering several scenarios of data availability, measurement noise and prior knowledge. We show how our method ensures improved estimations with faster and more stable convergence. We also show how the calibrated models are more generalizable. Finally, we give a set of simple guidelines to apply this strategy to a wide variety of calibration problems. CONCLUSIONS: Here we provide a parameter estimation strategy which combines efficient global optimization with a regularization scheme. This method is able to calibrate dynamic models in an efficient and robust way, effectively fighting overfitting and allowing the incorporation of prior information.


Subject(s)
Models, Biological , Systems Biology/methods , Algorithms , Metabolic Networks and Pathways , Nonlinear Dynamics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...