1
|
Tien NTN, Thu NQ, Kim DH, Park S, Long NP. EasyPubPlot: A Shiny Web Application for Rapid Omics Data Exploration and Visualization. J Proteome Res 2025; 24:2188-2195. [PMID: 40053871 DOI: 10.1021/acs.jproteome.4c01068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2025]
Abstract
Computational toolkits for data exploration and visualization from widely used omics platforms often lack flexibility and customization. While many tools generate standardized output, advanced programming skills are necessary to create high-quality visualizations. Therefore, user-friendly tools that simplify this crucial, yet time-consuming, step are essential. We developed EasyPubPlot (Easy Publishable Plotting), a straightforward, easy-to-use, no-coding, user experience-oriented, open-source, and shiny web application along with its associated R package to streamline data exploration and visualization for functional omics-empowered research. EasyPubPlot generates publishable scores plots, volcano plots, heatmaps, box plots, dot plots, and bubble plots with minimal necessary steps. The tool was designed to guide new users to accurate and efficient navigation. Step-by-step tutorials for each type of plot are also provided. Herein, we demonstrated EasyPubPlot's competent functionality and versatility by showcasing metabolomics, proteomics, and transcriptomics data. Collectively, EasyPubPlot reduces the gap between data analysis and stunning visualization, thereby diminishing friction and focusing on science. The app can be downloaded and installed locally (https://github.com/Pharmaco-OmicsLab/EasyPubPlot) or used through a web application (https://pharmaco-omicslab.shinyapps.io/EasyPubPlot).
Collapse
Affiliation(s)
- Nguyen Tran Nam Tien
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Nguyen Quang Thu
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Dong Hyun Kim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Seongoh Park
- School of Mathematics, Statistics and Data Science, Sungshin Women's University, Seoul 02844, Republic of Korea
- Data Science Center, Sungshin Women's University, Seoul 02844, Republic of Korea
| | - Nguyen Phuoc Long
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| |
Collapse
|
2
|
Monfort-Lanzas P, Gostner JM, Hackl H. Modeling omics dose-response at the pathway level with DoseRider. Comput Struct Biotechnol J 2025; 27:1440-1448. [PMID: 40242291 PMCID: PMC12001094 DOI: 10.1016/j.csbj.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
The generation of omics data sets has become an important approach in modern pharmacological and toxicological research as it can provide mechanistic and quantitative information on a large scale. Analyses of these data frequently revealed a non-linear dose-response relationship underscoring the importance of the modeling process to infer biological exposure limits. A number of tools have been developed for dose-response modeling and various thresholds have been defined as a quantitative representation of the effect of a substance, such as effective concentrations or benchmark doses (BMD). Here we present DoseRider an easy-to-use web application and a companion R package for linear and non-linear dose-response modeling and assessment of BMD at the level of biological pathways or signatures using generalized mixed effect models. This approach allows to analyze custom or provided multi-omics data such as RNA sequencing or metabolomics data and its annotation of a collection of pathways and gene sets from various species. Moreover, we introduce the concept of the trend change doses (TCDs) as a numerical descriptor of effects derived from complex dose-response curves. The usability of DoseRider was demonstrated by analyses of RNA sequencing data of bisphenol AF (BPAF) treatment of a human breast cancer cell line (MCF-7) at 8 different concentrations using gene sets for chemical and genetic perturbations (MSigDB). The BMD for BPAF and a set of genes upregulated by estrogen in breast cancer was 0.2 µM (95 %-CI 0.1-0.5 µM) and the lowest TCD (TCD1) was 0.003 µM (95 %-CI 0.0006-0.01 µM). The comprehensive presentation of the results underlines the suitability of the system for pharmacogenomics, toxicogenomics, and applications beyond.
Collapse
Affiliation(s)
- Pablo Monfort-Lanzas
- Institute of Medical Biochemistry, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
- Institute of Bioinformatics, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Johanna M. Gostner
- Institute of Medical Biochemistry, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Hubert Hackl
- Institute of Bioinformatics, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
3
|
Drake C, Zobl W, Escher SE. Assessment of pulmonary fibrosis using weighted gene co-expression network analysis. FRONTIERS IN TOXICOLOGY 2024; 6:1465704. [PMID: 39512679 PMCID: PMC11540828 DOI: 10.3389/ftox.2024.1465704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024] Open
Abstract
For many industrial chemicals toxicological data is sparse regarding several regulatory endpoints, so there is a high and often unmet demand for NAMs that allow for screening and prioritization of these chemicals. In this proof of concept case study we propose multi-gene biomarkers of compounds' ability to induce lung fibrosis and demonstrate their application in vitro. For deriving these biomarkers we used weighted gene co-expression network analysis to reanalyze a study where the time-dependent pulmonary gene-expression in mice treated with bleomycin had been documented. We identified eight modules of 58 to 273 genes each which were particularly activated during the different phases (inflammatory; acute and late fibrotic) of the developing fibrosis. The modules' relation to lung fibrosis was substantiated by comparison to known markers of lung fibrosis from DisGenet. Finally, we show the modules' application as biomarkers of chemical inducers of lung fibrosis based on an in vitro study of four diketones. Clear differences could be found between the lung fibrosis inducing diketones and other compounds with regard to their tendency to induce dose-dependent increases of module activation as determined using a previously proposed differential activation score and the fraction of differentially expressed genes in the modules. Accordingly, this study highlights the potential use of composite biomarkers mechanistic screening for compound-induced lung fibrosis.
Collapse
|
4
|
McClure RS, Rericha Y, Waters KM, Tanguay RL. 3' RNA-seq is superior to standard RNA-seq in cases of sparse data but inferior at identifying toxicity pathways in a model organism. FRONTIERS IN BIOINFORMATICS 2023; 3:1234218. [PMID: 37576716 PMCID: PMC10414111 DOI: 10.3389/fbinf.2023.1234218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction: The application of RNA-sequencing has led to numerous breakthroughs related to investigating gene expression levels in complex biological systems. Among these are knowledge of how organisms, such as the vertebrate model organism zebrafish (Danio rerio), respond to toxicant exposure. Recently, the development of 3' RNA-seq has allowed for the determination of gene expression levels with a fraction of the required reads compared to standard RNA-seq. While 3' RNA-seq has many advantages, a comparison to standard RNA-seq has not been performed in the context of whole organism toxicity and sparse data. Methods and results: Here, we examined samples from zebrafish exposed to perfluorobutane sulfonamide (FBSA) with either 3' or standard RNA-seq to determine the advantages of each with regards to the identification of functionally enriched pathways. We found that 3' and standard RNA-seq showed specific advantages when focusing on annotated or unannotated regions of the genome. We also found that standard RNA-seq identified more differentially expressed genes (DEGs), but that this advantage disappeared under conditions of sparse data. We also found that standard RNA-seq had a significant advantage in identifying functionally enriched pathways via analysis of DEG lists but that this advantage was minimal when identifying pathways via gene set enrichment analysis of all genes. Conclusions: These results show that each approach has experimental conditions where they may be advantageous. Our observations can help guide others in the choice of 3' RNA-seq vs standard RNA sequencing to query gene expression levels in a range of biological systems.
Collapse
Affiliation(s)
- Ryan S. McClure
- Biological Sciences Division, Pacific Northwest Laboratory, Richland, WA, United States
| | - Yvonne Rericha
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States
| | - Katrina M. Waters
- Biological Sciences Division, Pacific Northwest Laboratory, Richland, WA, United States
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States
| | - Robyn L. Tanguay
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States
| |
Collapse
|
5
|
Cecchetto M, Peruzza L, Giubilato E, Bernardini I, Rovere GD, Marcomini A, Regoli F, Bargelloni L, Patarnello T, Semenzin E, Milan M. An innovative index to incorporate transcriptomic data into weight of evidence approaches for environmental risk assessment. ENVIRONMENTAL RESEARCH 2023; 227:115745. [PMID: 36972774 DOI: 10.1016/j.envres.2023.115745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/15/2023] [Accepted: 03/22/2023] [Indexed: 05/08/2023]
Abstract
The sharp decrease in the cost of RNA-sequencing and the rapid improvement in computational analysis of eco-toxicogenomic data have brought new insights into the adverse effects of chemicals on aquatic organisms. Yet, transcriptomics is generally applied qualitatively in environmental risk assessments, hampering more effective exploitation of this evidence through multidisciplinary studies. In view of this limitation, a methodology is here presented to quantitatively elaborate transcriptional data in support to environmental risk assessment. The proposed methodology makes use of results from the application of Gene Set Enrichment Analysis to recent studies investigating the response of Mytilus galloprovincialis and Ruditapes philippinarum exposed to contaminants of emerging concern. The degree of changes in gene sets and the relevance of physiological reactions are integrated in the calculation of a hazard index. The outcome is then classified according to five hazard classes (from absent to severe), providing an evaluation of whole-transcriptome effects of chemical exposure. The application to experimental and simulated datasets proved that the method can effectively discriminate different levels of altered transcriptomic responses when compared to expert judgement (Spearman correlation coefficient of 0.96). A further application to data collected in two independent studies of Salmo trutta and Xenopus tropicalis exposed to contaminants confirmed the potential extension of the methodology to other aquatic species. This methodology can serve as a proof of concept for the integration of "genomic tools" in environmental risk assessment based on multidisciplinary investigations. To this end, the proposed transcriptomic hazard index can now be incorporated into quantitative Weight of Evidence approaches and weighed, with results from other types of analysis, to elucidate the role of chemicals in adverse ecological effects.
Collapse
Affiliation(s)
- Martina Cecchetto
- Department of Environmental Sciences, Informatics and Statistics, Ca' Foscari University of Venice, via Torino 155, 30172, Mestre-Venezia, Italy
| | - Luca Peruzza
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020, Legnaro, Padova, Italy
| | - Elisa Giubilato
- Department of Environmental Sciences, Informatics and Statistics, Ca' Foscari University of Venice, via Torino 155, 30172, Mestre-Venezia, Italy
| | - Ilaria Bernardini
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020, Legnaro, Padova, Italy
| | - Giulia Dalla Rovere
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020, Legnaro, Padova, Italy
| | - Antonio Marcomini
- Department of Environmental Sciences, Informatics and Statistics, Ca' Foscari University of Venice, via Torino 155, 30172, Mestre-Venezia, Italy
| | - Francesco Regoli
- Department of Life and Environmental Sciences, Marche Polytechnic University, Via Brecce Bianche, 60131, Ancona, Italy; NFBC, National Future Biodiversity Center, Palermo, Italy
| | - Luca Bargelloni
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020, Legnaro, Padova, Italy
| | - Tomaso Patarnello
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020, Legnaro, Padova, Italy
| | - Elena Semenzin
- Department of Environmental Sciences, Informatics and Statistics, Ca' Foscari University of Venice, via Torino 155, 30172, Mestre-Venezia, Italy.
| | - Massimo Milan
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020, Legnaro, Padova, Italy; NFBC, National Future Biodiversity Center, Palermo, Italy
| |
Collapse
|
6
|
Lizano-Fallas V, Carrasco del Amor A, Cristobal S. Prediction of Molecular Initiating Events for Adverse Outcome Pathways Using High-Throughput Identification of Chemical Targets. TOXICS 2023; 11:189. [PMID: 36851063 PMCID: PMC9965981 DOI: 10.3390/toxics11020189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/13/2023] [Indexed: 06/18/2023]
Abstract
The impact of exposure to multiple chemicals raises concerns for human and environmental health. The adverse outcome pathway method offers a framework to support mechanism-based assessment in environmental health starting by describing which mechanisms are triggered upon interaction with different stressors. The identification of the molecular initiating event and the molecular interaction between a chemical and a protein target is still a challenge for the development of adverse outcome pathways. The cellular response to chemical exposure studied with omics could not directly identify the protein targets. However, recent mass spectrometry-based methods are offering a proteome-wide identification of protein targets interacting with s but unrevealing a molecular initiating event from a set of targets is still dependent on available knowledge. Here, we directly coupled the target identification findings from the proteome integral solubility alteration assay with an analytical hierarchy process for the prediction of a prioritized molecular initiating event. We demonstrate the applicability of this combination of methodologies with a test compound (TCDD), and it could be further studied and integrated into AOPs. From the eight protein targets identified by the proteome integral solubility alteration assay after analyzing 2824 human hepatic proteins, the analytical hierarchy process can select the most suitable protein for an AOP. Our combined method solves the missing links between high-throughput target identification and prediction of the molecular initiating event. We anticipate its utility to decipher new molecular initiating events and support more sustainable methodologies to gain time and resources in chemical assessment.
Collapse
Affiliation(s)
- Veronica Lizano-Fallas
- Department of Biomedical and Clinical Sciences, Cell Biology, Faculty of Medicine, Linköping University, 581 85 Linköping, Sweden
| | - Ana Carrasco del Amor
- Department of Biomedical and Clinical Sciences, Cell Biology, Faculty of Medicine, Linköping University, 581 85 Linköping, Sweden
| | - Susana Cristobal
- Department of Biomedical and Clinical Sciences, Cell Biology, Faculty of Medicine, Linköping University, 581 85 Linköping, Sweden
- Ikerbasque, Basque Foundation for Sciences, Department of Physiology, Faculty of Medicine, and Nursing, University of the Basque Country (UPV/EHU), 489 40 Leioa, Spain
| |
Collapse
|
7
|
Oku Y, Madia F, Lau P, Paparella M, McGovern T, Luijten M, Jacobs MN. Analyses of Transcriptomics Cell Signalling for Pre-Screening Applications in the Integrated Approach for Testing and Assessment of Non-Genotoxic Carcinogens. Int J Mol Sci 2022; 23:ijms232112718. [PMID: 36361516 PMCID: PMC9659232 DOI: 10.3390/ijms232112718] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/18/2022] [Accepted: 10/18/2022] [Indexed: 12/03/2022] Open
Abstract
With recent rapid advancement of methodological tools, mechanistic understanding of biological processes leading to carcinogenesis is expanding. New approach methodologies such as transcriptomics can inform on non-genotoxic mechanisms of chemical carcinogens and can be developed for regulatory applications. The Organisation for the Economic Cooperation and Development (OECD) expert group developing an Integrated Approach to the Testing and Assessment (IATA) of Non-Genotoxic Carcinogens (NGTxC) is reviewing the possible assays to be integrated therein. In this context, we review the application of transcriptomics approaches suitable for pre-screening gene expression changes associated with phenotypic alterations that underlie the carcinogenic processes for subsequent prioritisation of downstream test methods appropriate to specific key events of non-genotoxic carcinogenesis. Using case studies, we evaluate the potential of gene expression analyses especially in relation to breast cancer, to identify the most relevant approaches that could be utilised as (pre-) screening tools, for example Gene Set Enrichment Analysis (GSEA). We also consider how to address the challenges to integrate gene panels and transcriptomic assays into the IATA, highlighting the pivotal omics markers identified for assay measurement in the IATA key events of inflammation, immune response, mitogenic signalling and cell injury.
Collapse
Affiliation(s)
- Yusuke Oku
- The Organisation for Economic Cooperation and Development (OECD), 2 Rue Andre Pascal, 75016 Paris, France
- Correspondence: (Y.O.); (M.N.J.)
| | - Federica Madia
- European Commission, Joint Research Centre (JRC), Via Enrico Fermi, 2749, 21027 Ispra, Italy
| | - Pierre Lau
- Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Martin Paparella
- Institute of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Innrain 80, 6020 Innbruck, Austria
| | - Timothy McGovern
- US Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20901, USA
| | - Mirjam Luijten
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, Bilthoven, 3721 MA Utrecht, The Netherlands
| | - Miriam N. Jacobs
- Centre for Radiation, Chemical and Environmental Hazard (CRCE), Public Health England (PHE), Chilton OX11 0RQ, Oxfordshire, UK
- Correspondence: (Y.O.); (M.N.J.)
| |
Collapse
|
8
|
Bundy JL, Judson R, Williams AJ, Grulke C, Shah I, Everett LJ. Predicting molecular initiating events using chemical target annotations and gene expression. BioData Min 2022; 15:7. [PMID: 35246223 PMCID: PMC8895536 DOI: 10.1186/s13040-022-00292-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 02/10/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The advent of high-throughput transcriptomic screening technologies has resulted in a wealth of publicly available gene expression data associated with chemical treatments. From a regulatory perspective, data sets that cover a large chemical space and contain reference chemicals offer utility for the prediction of molecular initiating events associated with chemical exposure. Here, we integrate data from a large compendium of transcriptomic responses to chemical exposure with a comprehensive database of chemical-protein associations to train binary classifiers that predict mechanism(s) of action from transcriptomic responses. First, we linked reference chemicals present in the LINCS L1000 gene expression data collection to chemical identifiers in RefChemDB, a database of chemical-protein interactions. Next, we trained binary classifiers on MCF7 human breast cancer cell line derived gene expression profiles and chemical-protein labels using six classification algorithms to identify optimal analysis parameters. To validate classifier accuracy, we used holdout data sets, training-excluded reference chemicals, and empirical significance testing of null models derived from permuted chemical-protein associations. To identify classifiers that have variable predicting performance across training data derived from different cellular contexts, we trained a separate set of binary classifiers on the PC3 human prostate cancer cell line. RESULTS We trained classifiers using expression data associated with chemical treatments linked to 51 molecular initiating events. This analysis identified and validated 9 high-performing classifiers with empirical p-values lower than 0.05 and internal accuracies ranging from 0.73 to 0.94 and holdout accuracies of 0.68 to 0.92. High-ranking predictions for training-excluded reference chemicals demonstrating that predictive accuracy extends beyond the set of chemicals used in classifier training. To explore differences in classifier performance as a function of training data cellular context, MCF7-trained classifier accuracies were compared to classifiers trained on the PC3 gene expression data for the same molecular initiating events. CONCLUSIONS This methodology can offer insight in prioritizing candidate perturbagens of interest for targeted screens. This approach can also help guide the selection of relevant cellular contexts for screening classes of candidate perturbagens using cell line specific model performance.
Collapse
Affiliation(s)
- Joseph L Bundy
- Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, 109 T.W. Alexander Drive, Durham, NC, 27709, USA
| | - Richard Judson
- Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, 109 T.W. Alexander Drive, Durham, NC, 27709, USA
| | - Antony J Williams
- Chemical Characterization and Exposure Division, Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, 109 T.W. Alexander Drive, Durham, NC, 27709, USA
| | - Chris Grulke
- Chemical Characterization and Exposure Division, Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, 109 T.W. Alexander Drive, Durham, NC, 27709, USA
| | - Imran Shah
- Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, 109 T.W. Alexander Drive, Durham, NC, 27709, USA
| | - Logan J Everett
- Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, 109 T.W. Alexander Drive, Durham, NC, 27709, USA.
| |
Collapse
|
9
|
ALOHA: Aggregated local extrema splines for high-throughput dose-response analysis. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 21:100196. [PMID: 35083394 PMCID: PMC8785973 DOI: 10.1016/j.comtox.2021.100196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Computational methods for genomic dose-response integrate dose-response modeling with bioinformatics tools to evaluate changes in molecular and cellular functions related to pathogenic processes. These methods use parametric models to describe each gene's dose-response, but such models may not adequately capture expression changes. Additionally, current approaches do not consider gene co-expression networks. When assessing co-expression networks, one typically does not consider the dose-response relationship, resulting in 'co-regulated' gene sets containing genes having different dose-response patterns. To avoid these limitations, we develop an analysis pipeline called Aggregated Local Extrema Splines for High-throughput Analysis (ALOHA), which computes individual genomic dose-response functions using a flexible class Bayesian shape constrained splines and clusters gene co-regulation based upon these fits. Using splines, we reduce information loss due to parametric lack-of-fit issues, and because we cluster on dose-response relationships, we better identify co-regulation clusters for genes that have co-expressed dose-response patterns from chemical exposure. The clustered pathways can then be used to estimate a dose associated with a pre-specified biological response, i.e., the benchmark dose (BMD), and approximate a point of departure dose corresponding to minimal adverse response in the whole tissue/organism. We compare our approach to current parametric methods and our biologically enriched gene sets to cluster on normalized expression data. Using this methodology, we can more effectively extract the underlying structure leading to more cohesive estimates of gene set potency.
Collapse
|
10
|
Zhou G, Chen J, Wu C, Jiang P, Wang Y, Zhang Y, Jiang Y, Li X. Deciphering the Protein, Modular Connections and Precision Medicine for Heart Failure With Preserved Ejection Fraction and Hypertension Based on TMT Quantitative Proteomics and Molecular Docking. Front Physiol 2021; 12:607089. [PMID: 34721049 PMCID: PMC8552070 DOI: 10.3389/fphys.2021.607089] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 09/23/2021] [Indexed: 12/16/2022] Open
Abstract
Background: Exploring the potential biological relationships between heart failure with preserved ejection fraction (HFpEF) and concomitant diseases has been the focus of many studies for the establishment of personalized therapies. Hypertension (HTN) is the most common concomitant disease in HFpEF patients, but the functional connections between HFpEF and HTN are still not fully understood and effective treatment strategies are still lacking. Methods: In this study, tandem mass tag (TMT) quantitative proteomics was used to identify disease-related proteins and construct disease-related networks. Furthermore, functional enrichment analysis of overlapping network modules was used to determine the functional similarities between HFpEF and HTN. Molecular docking and module analyses were combined to identify therapeutic targets for HFpEF and HTN. Results: Seven common differentially expressed proteins (co-DEPs) and eight overlapping modules were identified in HFpEF and HTN. The common biological processes between HFpEF and HTN were mainly related to energy metabolism. Myocardial contraction, energy metabolism, apoptosis, oxidative stress, immune response, and cardiac hypertrophy were all closely associated with HFpEF and HTN. Epinephrine, sulfadimethoxine, chloroform, and prednisolone acetate were best matched with the co-DEPs by molecular docking analyses. Conclusion: Myocardial contraction, energy metabolism, apoptosis, oxidative stress, immune response, and cardiac hypertrophy were the main functional connections between HFpEF and HTN. Epinephrine, sulfadimethoxine, chloroform, and prednisolone acetate could potentially be effective for the treatment of HTN and HFpEF.
Collapse
Affiliation(s)
- Guofeng Zhou
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiye Chen
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chuanhong Wu
- The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
| | - Ping Jiang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yongcheng Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yongjian Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuehua Jiang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
11
|
Kim M, Jee SC, Kim S, Hwang KH, Sung JS. Identification and Characterization of mRNA Biomarkers for Sodium Cyanide Exposure. TOXICS 2021; 9:toxics9110288. [PMID: 34822678 PMCID: PMC8624962 DOI: 10.3390/toxics9110288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 10/27/2021] [Accepted: 11/01/2021] [Indexed: 11/16/2022]
Abstract
Biomarkers in exposure assessment are defined as the quantifiable targets that indicate the exposure to hazardous chemicals and their resulting health effect. In this study, we aimed to identify, validate, and characterize the mRNA biomarker that can detect the exposure of sodium cyanide. To identify reliable biomarkers for sodium cyanide exposure, critical criteria were defined for candidate selection: (1) the expression level of mRNA significantly changes in response to sodium thiocyanate treatment in transcriptomics results (fold change > 2.0 or <0.50, adjusted p-value < 0.05); and (2) the mRNA level is significantly modulated by sodium cyanide exposure in both normal human lung cells and rat lung tissue. We identified the following mRNA biomarker candidates: ADCY5, ANGPTL4, CCNG2, CD9, COL1A2, DACT3, GGCX, GRB14, H1F0, HSPA1A, MAF, MAT2A, PPP1R10, and PPP4C. The expression levels of these candidates were commonly downregulated by sodium cyanide exposure both in vitro and in vivo. We functionally characterized the biomarkers and established the impact of sodium cyanide on transcriptomic profiles using in silico approaches. Our results suggest that the biomarkers may contribute to the regulation and degradation of the extracellular matrix, leading to a negative effect on surrounding lung cells.
Collapse
Affiliation(s)
- Min Kim
- Department of Life Science, Biomedi Campus, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang 10326, Gyeonggi-do, Korea; (M.K.); (S.-C.J.); (S.K.)
| | - Seung-Cheol Jee
- Department of Life Science, Biomedi Campus, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang 10326, Gyeonggi-do, Korea; (M.K.); (S.-C.J.); (S.K.)
| | - Soee Kim
- Department of Life Science, Biomedi Campus, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang 10326, Gyeonggi-do, Korea; (M.K.); (S.-C.J.); (S.K.)
| | - Kyung-Hwa Hwang
- Jeonbuk Branch, Korea Institute of Toxicology, KIT, KRICT, 30 Baehak 1-gil, Jeongeup-si 56212, Jeollabuk-do, Korea;
| | - Jung-Suk Sung
- Department of Life Science, Biomedi Campus, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang 10326, Gyeonggi-do, Korea; (M.K.); (S.-C.J.); (S.K.)
- Correspondence: ; Tel.: +82-31-961-5132; Fax: +82-31-961-5108
| |
Collapse
|
12
|
Lee F, Shah I, Soong YT, Xing J, Ng IC, Tasnim F, Yu H. Reproducibility and robustness of high-throughput S1500+ transcriptomics on primary rat hepatocytes for chemical-induced hepatotoxicity assessment. Curr Res Toxicol 2021; 2:282-295. [PMID: 34467220 PMCID: PMC8384775 DOI: 10.1016/j.crtox.2021.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/15/2021] [Accepted: 07/31/2021] [Indexed: 11/06/2022] Open
Abstract
TempO-Seq assays of rat hepatocytes in collagen sandwich are highly reproducible. Gene expression analysis shows S1500+ is representative of the whole transcriptome. Connectivity mapping shows consistency between TempO-Seq and Affymetrix data. Gene set enrichment shows consistency between S1500+ and the whole transcriptome. Gene set enrichment using hallmark gene sets informs hepatotoxicity.
Cell-based in vitro models coupled with high-throughput transcriptomics (HTTr) are increasingly utilized as alternative methods to animal-based toxicity testing. Here, using a panel of 14 chemicals with different risks of human drug-induced liver injury (DILI) and two dosing concentrations, we evaluated an HTTr platform comprised of collagen sandwich primary rat hepatocyte culture and the TempO-Seq surrogate S1500+ (ST) assay. First, the HTTr platform was found to exhibit high reproducibility between technical and biological replicates (r greater than 0.85). Connectivity mapping analysis further demonstrated a high level of inter-platform reproducibility between TempO-Seq data and Affymetrix GeneChip data from the Open TG-GATES project. Second, the TempO-Seq ST assay was shown to be a robust surrogate to the whole transcriptome (WT) assay in capturing chemical-induced changes in gene expression, as evident from correlation analysis, PCA and unsupervised hierarchical clustering. Gene set enrichment analysis (GSEA) using the Hallmark gene set collection also demonstrated consistency in enrichment scores between ST and WT assays. Lastly, unsupervised hierarchical clustering of hallmark enrichment scores broadly divided the samples into hepatotoxic, intermediate, and non-hepatotoxic groups. Xenobiotic metabolism, bile acid metabolism, apoptosis, p53 pathway, and coagulation were found to be the key hallmarks driving the clustering. Taken together, our results established the reproducibility and performance of collagen sandwich culture in combination with TempO-Seq S1500+ assay, and demonstrated the utility of GSEA using the hallmark gene set collection to identify potential hepatotoxicants for further validation.
Collapse
Affiliation(s)
- Fan Lee
- Innovations in Food & Chemical Safety Program (IFCS), Institute of Bioengineering and Bioimaging (IBB), Agency for Science Technology and Research, Singapore
| | - Imran Shah
- Center for Computational Toxicology & Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
| | - Yun Ting Soong
- Innovations in Food & Chemical Safety Program (IFCS), Institute of Bioengineering and Bioimaging (IBB), Agency for Science Technology and Research, Singapore
| | - Jiangwa Xing
- Innovations in Food & Chemical Safety Program (IFCS), Institute of Bioengineering and Bioimaging (IBB), Agency for Science Technology and Research, Singapore
| | - Inn Chuan Ng
- Department of Physiology and Mechanobiology Institute, National University of Singapore, Singapore
| | - Farah Tasnim
- Innovations in Food & Chemical Safety Program (IFCS), Institute of Bioengineering and Bioimaging (IBB), Agency for Science Technology and Research, Singapore
| | - Hanry Yu
- Innovations in Food & Chemical Safety Program (IFCS), Institute of Bioengineering and Bioimaging (IBB), Agency for Science Technology and Research, Singapore.,Department of Physiology and Mechanobiology Institute, National University of Singapore, Singapore.,Critical Analytics for Manufacturing Personalized-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore
| |
Collapse
|
13
|
Manem VS, Sazonova O, Gagné A, Orain M, Khoshkrood-Mansoori B, Gaudreault N, Bossé Y, Joubert P. Unravelling actionable biology using transcriptomic data to integrate mitotic index and Ki-67 in the management of lung neuroendocrine tumors. Oncotarget 2021; 12:209-220. [PMID: 33613848 PMCID: PMC7869577 DOI: 10.18632/oncotarget.27874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 01/19/2021] [Indexed: 11/25/2022] Open
Abstract
Pulmonary neuroendocrine tumors (NETs) are a heterogeneous family of malignancies whose classification relies on morphology and mitotic rate, unlike extrapulmonary neuroendocrine tumors that require both mitotic rate and Ki-67. As mitotic count is proportional to Ki-67, it is crucial to understand if Ki-67 can complement the existing diagnostic guidelines, as well as discover the benefit of these two markers to unravel the biological heterogeneity. In this study, we investigated the association of mitotic rate and Ki-67 at gene- and pathway-level using transcriptomic data in lung NET malignancies. Lung resection tumor specimens obtained from 28 patients diagnosed with NETs were selected. Mitotic rate, Ki-67 and transcriptomic data were obtained for all samples. The concordance between mitotic rate and Ki-67 was evaluated at gene-level and pathway-level using gene expression data. Our analysis revealed a strong association between mitotic rate and Ki-67 across all samples and cell cycle genes were found to be differentially ranked between them. Pathway analysis indicated that a greater number of pathways overlapped between these markers. Analyses based on lung NET subtypes revealed that mitotic rate in carcinoids and Ki-67 in large cell neuroendocrine carcinomas provided comprehensive characterization of pathways among these malignancies. Among the two subtypes, we found distinct leading-edge gene sets that drive the enrichment signal of commonly enriched pathways between mitotic index and Ki-67. Overall, our findings delineated the degree of benefit of the two proliferation markers, and offers new layer to predict the biological behavior and identify high-risk patients using a more comprehensive diagnostic workup.
Collapse
Affiliation(s)
- Venkata S.K. Manem
- Quebec Heart and Lung Institute Research Center, Quebec City, QC G1V4G5, Canada
| | - Olga Sazonova
- Quebec Heart and Lung Institute Research Center, Quebec City, QC G1V4G5, Canada
| | - Andréanne Gagné
- Quebec Heart and Lung Institute Research Center, Quebec City, QC G1V4G5, Canada
| | - Michèle Orain
- Quebec Heart and Lung Institute Research Center, Quebec City, QC G1V4G5, Canada
| | | | - Nathalie Gaudreault
- Quebec Heart and Lung Institute Research Center, Quebec City, QC G1V4G5, Canada
| | - Yohan Bossé
- Quebec Heart and Lung Institute Research Center, Quebec City, QC G1V4G5, Canada
- Department of Molecular Medicine, Laval University, Quebec City, QC G1V4G5, Canada
| | - Philippe Joubert
- Quebec Heart and Lung Institute Research Center, Quebec City, QC G1V4G5, Canada
- Department of Medical Biochemistry, Molecular Biology and Pathology, Laval University, Quebec City, QC G1V4G5, Canada
| |
Collapse
|
14
|
Chappell GA, Heintz MM, Haws LC. Transcriptomic analyses of livers from mice exposed to 1,4-dioxane for up to 90 days to assess potential mode(s) of action underlying liver tumor development. Curr Res Toxicol 2021; 2:30-41. [PMID: 34345848 PMCID: PMC8320614 DOI: 10.1016/j.crtox.2021.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 12/11/2022] Open
Abstract
1,4-Dioxane is a volatile organic compound with industrial and commercial applications as a solvent and in the manufacture of other chemicals. 1,4-Dioxane has been demonstrated to induce liver tumors in chronic rodent bioassays conducted at very high doses. The available evidence for 1,4-dioxane-induced liver tumors in rodents aligns with a threshold-dependent mode of action (MOA), with the underlying mechanism being less clear in the mouse than in rats. To gain a better understanding of the underlying molecular mechanisms related to liver tumor development in mice orally exposed to 1,4-dioxane, transcriptomics analysis was conducted on liver tissue collected from a 90-day drinking water study in female B6D2F1/Crl mice (Lafranconi et al., 2020). Using tissue samples from female mice exposed to 1,4-dioxane in the drinking water at concentrations of 0, 40, 200, 600, 2,000 or 6,000 ppm for 7, 28, and 90 days, transcriptomic analyses demonstrate minimal treatment effects on global gene expression at concentrations below 600 ppm. At higher concentrations, genes involved in phase II metabolism and mitotic cell cycle checkpoints were significantly upregulated. There was an overall lack of enrichment of genes related to DNA damage response. The increase in mitotic signaling is most prevalent in the livers of mice exposed to 1,4-dioxane at the highest concentrations for 90 days. This finding aligns with phenotypic changes reported by Lafranconi et al. (2020) after 90-days of exposure to 6,000 ppm 1,4-dioxane in the same tissues. The transcriptomics analysis further supports overarching study findings demonstrating a non-mutagenic, threshold-based, mitogenic MOA for 1,4-dioxane-induced liver tumors.
Collapse
Affiliation(s)
- G A Chappell
- ToxStrategies, Inc., Asheville, NC, United States
| | - M M Heintz
- ToxStrategies, Inc., Asheville, NC, United States
| | - L C Haws
- ToxStrategies, Inc., Austin, TX, United States
| |
Collapse
|
15
|
Pan X, Ma X. A Novel Six-Gene Signature for Prognosis Prediction in Ovarian Cancer. Front Genet 2020; 11:1006. [PMID: 33193589 PMCID: PMC7593580 DOI: 10.3389/fgene.2020.01006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 08/06/2020] [Indexed: 12/18/2022] Open
Abstract
Ovarian cancer (OC) is the most malignant tumor in the female reproductive tract. Although abundant molecular biomarkers have been identified, a robust and accurate gene expression signature is still essential to assist oncologists in evaluating the prognosis of OC patients. In this study, samples from 367 patients in The Cancer Genome Atlas (TCGA) database were subjected to mRNA expression profiling. Then, we used a gene set enrichment analysis (GSEA) to screen genes correlated with epithelial–mesenchymal transition (EMT) and assess their prognostic power with a Cox proportional regression model. Six genes (TGFBI, SFRP1, COL16A1, THY1, PPIB, BGN) associated with overall survival (OS) were used to construct a risk assessment model, after which the patients were divided into high-risk and low-risk groups. The six-gene signature was an independent prognostic biomarker of OS for OC patients based on the multivariate Cox regression analysis. In addition, the six-gene model was validated with samples from the Gene Expression Omnibus (GEO) database. In summary, we established a six-gene signature relevant to the prognosis of OC, which might become a therapeutic tool with clinical applications in the future.
Collapse
Affiliation(s)
- Xin Pan
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoxin Ma
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
16
|
Mezencev R, Auerbach SS. The sensitivity of transcriptomics BMD modeling to the methods used for microarray data normalization. PLoS One 2020; 15:e0232955. [PMID: 32413060 PMCID: PMC7228135 DOI: 10.1371/journal.pone.0232955] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 04/25/2020] [Indexed: 11/25/2022] Open
Abstract
Whole-genome expression data generated by microarray studies have shown promise for quantitative human health risk assessment. While numerous approaches have been developed to determine benchmark doses (BMDs) from probeset-level dose responses, sensitivity of the results to methods used for normalization of the data has not yet been systematically investigated. Normalization of microarray data converts raw hybridization signals to expression estimates that are expected to be proportional to the amounts of transcripts in the profiled specimens. Different approaches to normalization have been shown to greatly influence the results of some downstream analyses, including biological interpretation. In this study we evaluate the influence of microarray normalization methods on the transcriptomic BMDs. We demonstrate using in vivo data that the use of alternative pipelines for normalization of Affymetrix microarray data can have a considerable impact on the number of detected differentially expressed genes and pathways (processes) determined to be treatment responsive, which may lead to alternative interpretations of the data. In addition, we found that normalization can have a considerable effect (as much as ~30-fold in this study) on estimation of the minimum biological potency (transcriptomic point of departure). We argue for consideration of alternative normalization methods and their data-informed selection to most effectively interpret microarray data for use in human health risk assessment.
Collapse
Affiliation(s)
- Roman Mezencev
- Center for Public Health and Environmental Assessment, Office of Research and Development, US EPA, Washington DC, United States of America
| | - Scott S. Auerbach
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, Durham, NC, United States of America
| |
Collapse
|
17
|
Federico A, Serra A, Ha MK, Kohonen P, Choi JS, Liampa I, Nymark P, Sanabria N, Cattelani L, Fratello M, Kinaret PAS, Jagiello K, Puzyn T, Melagraki G, Gulumian M, Afantitis A, Sarimveis H, Yoon TH, Grafström R, Greco D. Transcriptomics in Toxicogenomics, Part II: Preprocessing and Differential Expression Analysis for High Quality Data. NANOMATERIALS 2020; 10:nano10050903. [PMID: 32397130 PMCID: PMC7279140 DOI: 10.3390/nano10050903] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/29/2020] [Accepted: 05/04/2020] [Indexed: 12/28/2022]
Abstract
Preprocessing of transcriptomics data plays a pivotal role in the development of toxicogenomics-driven tools for chemical toxicity assessment. The generation and exploitation of large volumes of molecular profiles, following an appropriate experimental design, allows the employment of toxicogenomics (TGx) approaches for a thorough characterisation of the mechanism of action (MOA) of different compounds. To date, a plethora of data preprocessing methodologies have been suggested. However, in most cases, building the optimal analytical workflow is not straightforward. A careful selection of the right tools must be carried out, since it will affect the downstream analyses and modelling approaches. Transcriptomics data preprocessing spans across multiple steps such as quality check, filtering, normalization, batch effect detection and correction. Currently, there is a lack of standard guidelines for data preprocessing in the TGx field. Defining the optimal tools and procedures to be employed in the transcriptomics data preprocessing will lead to the generation of homogeneous and unbiased data, allowing the development of more reliable, robust and accurate predictive models. In this review, we outline methods for the preprocessing of three main transcriptomic technologies including microarray, bulk RNA-Sequencing (RNA-Seq), and single cell RNA-Sequencing (scRNA-Seq). Moreover, we discuss the most common methods for the identification of differentially expressed genes and to perform a functional enrichment analysis. This review is the second part of a three-article series on Transcriptomics in Toxicogenomics.
Collapse
Affiliation(s)
- Antonio Federico
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland; (A.F.); (A.S.); (L.C.); (M.F.); (P.A.S.K.)
- BioMediTech Institute, Tampere University, FI-33014 Tampere, Finland
| | - Angela Serra
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland; (A.F.); (A.S.); (L.C.); (M.F.); (P.A.S.K.)
- BioMediTech Institute, Tampere University, FI-33014 Tampere, Finland
| | - My Kieu Ha
- Center for Next Generation Cytometry, Hanyang University, Seoul 04763, Korea; (M.K.H.); (J.-S.C.); (T.-H.Y.)
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
- Institute of Next Generation Material Design, Hanyang University, Seoul 04763, Korea
| | - Pekka Kohonen
- Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (P.K.); (P.N.); (R.G.)
- Division of Toxicology, Misvik Biology, 20520 Turku, Finland
| | - Jang-Sik Choi
- Center for Next Generation Cytometry, Hanyang University, Seoul 04763, Korea; (M.K.H.); (J.-S.C.); (T.-H.Y.)
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
- Institute of Next Generation Material Design, Hanyang University, Seoul 04763, Korea
| | - Irene Liampa
- School of Chemical Engineering, National Technical University of Athens, 157 80 Athens, Greece; (I.L.); (H.S.)
| | - Penny Nymark
- Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (P.K.); (P.N.); (R.G.)
- Division of Toxicology, Misvik Biology, 20520 Turku, Finland
| | - Natasha Sanabria
- National Institute for Occupational Health, Johannesburg 30333, South Africa; (N.S.); (M.G.)
| | - Luca Cattelani
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland; (A.F.); (A.S.); (L.C.); (M.F.); (P.A.S.K.)
- BioMediTech Institute, Tampere University, FI-33014 Tampere, Finland
| | - Michele Fratello
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland; (A.F.); (A.S.); (L.C.); (M.F.); (P.A.S.K.)
- BioMediTech Institute, Tampere University, FI-33014 Tampere, Finland
| | - Pia Anneli Sofia Kinaret
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland; (A.F.); (A.S.); (L.C.); (M.F.); (P.A.S.K.)
- BioMediTech Institute, Tampere University, FI-33014 Tampere, Finland
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
| | - Karolina Jagiello
- QSAR Lab Ltd., Aleja Grunwaldzka 190/102, 80-266 Gdansk, Poland; (K.J.); (T.P.)
- Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland
| | - Tomasz Puzyn
- QSAR Lab Ltd., Aleja Grunwaldzka 190/102, 80-266 Gdansk, Poland; (K.J.); (T.P.)
- Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland
| | - Georgia Melagraki
- Nanoinformatics Department, NovaMechanics Ltd., Nicosia 1065, Cyprus; (G.M.); (A.A.)
| | - Mary Gulumian
- National Institute for Occupational Health, Johannesburg 30333, South Africa; (N.S.); (M.G.)
- Haematology and Molecular Medicine Department, School of Pathology, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Antreas Afantitis
- Nanoinformatics Department, NovaMechanics Ltd., Nicosia 1065, Cyprus; (G.M.); (A.A.)
| | - Haralambos Sarimveis
- School of Chemical Engineering, National Technical University of Athens, 157 80 Athens, Greece; (I.L.); (H.S.)
| | - Tae-Hyun Yoon
- Center for Next Generation Cytometry, Hanyang University, Seoul 04763, Korea; (M.K.H.); (J.-S.C.); (T.-H.Y.)
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
- Institute of Next Generation Material Design, Hanyang University, Seoul 04763, Korea
| | - Roland Grafström
- Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (P.K.); (P.N.); (R.G.)
- Division of Toxicology, Misvik Biology, 20520 Turku, Finland
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland; (A.F.); (A.S.); (L.C.); (M.F.); (P.A.S.K.)
- BioMediTech Institute, Tampere University, FI-33014 Tampere, Finland
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
- Correspondence:
| |
Collapse
|
18
|
Serra A, Fratello M, Cattelani L, Liampa I, Melagraki G, Kohonen P, Nymark P, Federico A, Kinaret PAS, Jagiello K, Ha MK, Choi JS, Sanabria N, Gulumian M, Puzyn T, Yoon TH, Sarimveis H, Grafström R, Afantitis A, Greco D. Transcriptomics in Toxicogenomics, Part III: Data Modelling for Risk Assessment. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E708. [PMID: 32276469 PMCID: PMC7221955 DOI: 10.3390/nano10040708] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/30/2022]
Abstract
Transcriptomics data are relevant to address a number of challenges in Toxicogenomics (TGx). After careful planning of exposure conditions and data preprocessing, the TGx data can be used in predictive toxicology, where more advanced modelling techniques are applied. The large volume of molecular profiles produced by omics-based technologies allows the development and application of artificial intelligence (AI) methods in TGx. Indeed, the publicly available omics datasets are constantly increasing together with a plethora of different methods that are made available to facilitate their analysis, interpretation and the generation of accurate and stable predictive models. In this review, we present the state-of-the-art of data modelling applied to transcriptomics data in TGx. We show how the benchmark dose (BMD) analysis can be applied to TGx data. We review read across and adverse outcome pathways (AOP) modelling methodologies. We discuss how network-based approaches can be successfully employed to clarify the mechanism of action (MOA) or specific biomarkers of exposure. We also describe the main AI methodologies applied to TGx data to create predictive classification and regression models and we address current challenges. Finally, we present a short description of deep learning (DL) and data integration methodologies applied in these contexts. Modelling of TGx data represents a valuable tool for more accurate chemical safety assessment. This review is the third part of a three-article series on Transcriptomics in Toxicogenomics.
Collapse
Affiliation(s)
- Angela Serra
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland; (A.S.); (M.F.); (L.C.); (A.F.); (P.A.S.K.)
- BioMediTech Institute, Tampere University, FI-33014 Tampere, Finland
| | - Michele Fratello
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland; (A.S.); (M.F.); (L.C.); (A.F.); (P.A.S.K.)
- BioMediTech Institute, Tampere University, FI-33014 Tampere, Finland
| | - Luca Cattelani
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland; (A.S.); (M.F.); (L.C.); (A.F.); (P.A.S.K.)
- BioMediTech Institute, Tampere University, FI-33014 Tampere, Finland
| | - Irene Liampa
- School of Chemical Engineering, National Technical University of Athens, 157 80 Athens, Greece; (I.L.); (H.S.)
| | - Georgia Melagraki
- Nanoinformatics Department, NovaMechanics Ltd., Nicosia 1065, Cyprus; (G.M.); (A.A.)
| | - Pekka Kohonen
- Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (P.K.); (P.N.); (R.G.)
- Division of Toxicology, Misvik Biology, 20520 Turku, Finland
| | - Penny Nymark
- Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (P.K.); (P.N.); (R.G.)
- Division of Toxicology, Misvik Biology, 20520 Turku, Finland
| | - Antonio Federico
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland; (A.S.); (M.F.); (L.C.); (A.F.); (P.A.S.K.)
- BioMediTech Institute, Tampere University, FI-33014 Tampere, Finland
| | - Pia Anneli Sofia Kinaret
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland; (A.S.); (M.F.); (L.C.); (A.F.); (P.A.S.K.)
- BioMediTech Institute, Tampere University, FI-33014 Tampere, Finland
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
| | - Karolina Jagiello
- QSAR Lab Ltd., Aleja Grunwaldzka 190/102, 80-266 Gdansk, Poland; (K.J.); (T.P.)
- University of Gdansk, Faculty of Chemistry, Wita Stwosza 63, 80-308 Gdansk, Poland
| | - My Kieu Ha
- Center for Next Generation Cytometry, Hanyang University, Seoul 04763, Korea; (M.K.H.); (J.-S.C.); (T.-H.Y.)
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
- Institute of Next Generation Material Design, Hanyang University, Seoul 04763, Korea
| | - Jang-Sik Choi
- Center for Next Generation Cytometry, Hanyang University, Seoul 04763, Korea; (M.K.H.); (J.-S.C.); (T.-H.Y.)
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
- Institute of Next Generation Material Design, Hanyang University, Seoul 04763, Korea
| | - Natasha Sanabria
- National Institute for Occupational Health, Johannesburg 30333, South Africa; (N.S.); (M.G.)
| | - Mary Gulumian
- National Institute for Occupational Health, Johannesburg 30333, South Africa; (N.S.); (M.G.)
- Haematology and Molecular Medicine Department, School of Pathology, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Tomasz Puzyn
- QSAR Lab Ltd., Aleja Grunwaldzka 190/102, 80-266 Gdansk, Poland; (K.J.); (T.P.)
- University of Gdansk, Faculty of Chemistry, Wita Stwosza 63, 80-308 Gdansk, Poland
| | - Tae-Hyun Yoon
- Center for Next Generation Cytometry, Hanyang University, Seoul 04763, Korea; (M.K.H.); (J.-S.C.); (T.-H.Y.)
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
- Institute of Next Generation Material Design, Hanyang University, Seoul 04763, Korea
| | - Haralambos Sarimveis
- School of Chemical Engineering, National Technical University of Athens, 157 80 Athens, Greece; (I.L.); (H.S.)
| | - Roland Grafström
- Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (P.K.); (P.N.); (R.G.)
- Division of Toxicology, Misvik Biology, 20520 Turku, Finland
| | - Antreas Afantitis
- Nanoinformatics Department, NovaMechanics Ltd., Nicosia 1065, Cyprus; (G.M.); (A.A.)
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland; (A.S.); (M.F.); (L.C.); (A.F.); (P.A.S.K.)
- BioMediTech Institute, Tampere University, FI-33014 Tampere, Finland
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
19
|
Ewald JD, Soufan O, Crump D, Hecker M, Xia J, Basu N. EcoToxModules: Custom Gene Sets to Organize and Analyze Toxicogenomics Data from Ecological Species. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:4376-4387. [PMID: 32106671 DOI: 10.1021/acs.est.9b06607] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Traditional results from toxicogenomics studies are complex lists of significantly impacted genes or gene sets, which are challenging to synthesize down to actionable results with a clear interpretation. Here, we defined two sets of 21 custom gene sets, called the functional and statistical EcoToxModules, in fathead minnow (Pimephales promelas) to (1) re-cast predefined molecular pathways into a toxicological framework and (2) provide a data-driven, unsupervised grouping of genes impacted by exposure to environmental contaminants. The functional EcoToxModules were identified by re-organizing KEGG pathways into biological processes that are more relevant to ecotoxicology based on the input from expert scientists and regulators. The statistical EcoToxModules were identified using co-expression analysis of publicly available microarray data (n = 303 profiles) measured in livers of fathead minnows after exposure to 38 different conditions. Potential applications of the EcoToxModules were demonstrated with two case studies that represent exposure to a pure chemical and to environmental wastewater samples. In comparisons to differential expression and gene set analysis, we found that EcoToxModule responses were consistent with these traditional results. Additionally, they were easier to visualize and quantitatively compare across different conditions, which facilitated drawing conclusions about the relative toxicity of the exposures within each case study.
Collapse
Affiliation(s)
- Jessica D Ewald
- Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-Bellevue H9X 3V9, Canada
| | - Othman Soufan
- Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-Bellevue H9X 3V9, Canada
| | - Doug Crump
- Ecotoxicology and Wildlife Health Division, Environment and Climate Change Canada, National Wildlife Research Centre, Ottawa K1A 0H3, Canada
| | - Markus Hecker
- School of the Environment & Sustainability and Toxicology Centre, University of Saskatchewan, Saskatoon S7N 5B3, Canada
| | - Jianguo Xia
- Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-Bellevue H9X 3V9, Canada
| | - Niladri Basu
- Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-Bellevue H9X 3V9, Canada
| |
Collapse
|
20
|
Chappell GA, Thompson CM, Wolf JC, Cullen JM, Klaunig JE, Haws LC. Assessment of the Mode of Action Underlying the Effects of GenX in Mouse Liver and Implications for Assessing Human Health Risks. Toxicol Pathol 2020; 48:494-508. [PMID: 32138627 PMCID: PMC7153225 DOI: 10.1177/0192623320905803] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
GenX is an alternative to environmentally persistent long-chain perfluoroalkyl and polyfluoroalkyl substances. Mice exposed to GenX exhibit liver hypertrophy, elevated peroxisomal enzyme activity, and other apical endpoints consistent with peroxisome proliferators. To investigate the potential role of peroxisome proliferator-activated receptor alpha (PPARα) activation in mice, and other molecular signals potentially related to observed liver changes, RNA sequencing was conducted on paraffin-embedded liver sections from a 90-day subchronic toxicity study of GenX conducted in mice. Differentially expressed genes were identified for each treatment group, and gene set enrichment analysis was conducted using gene sets that represent biological processes and known canonical pathways. Peroxisome signaling and fatty acid metabolism were among the most significantly enriched gene sets in both sexes at 0.5 and 5 mg/kg GenX; no pathways were enriched at 0.1 mg/kg. Gene sets specific to the PPARα subtype were significantly enriched. These findings were phenotypically anchored to histopathological changes in the same tissue blocks: hypertrophy, mitoses, and apoptosis. In vitro PPARα transactivation assays indicated that GenX activates mouse PPARα. These results indicate that the liver changes observed in GenX-treated mice occur via a mode of action (MOA) involving PPARα, an important finding for human health risk assessment as this MOA has limited relevance to humans.
Collapse
Affiliation(s)
| | | | | | - John M. Cullen
- North Carolina State University College of Veterinary Medicine, Raleigh, NC, USA
| | - James E. Klaunig
- Indiana University, School of Public Health, Bloomington, IN, USA
| | | |
Collapse
|
21
|
Noyes PD, Friedman KP, Browne P, Haselman JT, Gilbert ME, Hornung MW, Barone S, Crofton KM, Laws SC, Stoker TE, Simmons SO, Tietge JE, Degitz SJ. Evaluating Chemicals for Thyroid Disruption: Opportunities and Challenges with in Vitro Testing and Adverse Outcome Pathway Approaches. ENVIRONMENTAL HEALTH PERSPECTIVES 2019; 127:95001. [PMID: 31487205 PMCID: PMC6791490 DOI: 10.1289/ehp5297] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 07/01/2019] [Accepted: 08/13/2019] [Indexed: 05/19/2023]
Abstract
BACKGROUND Extensive clinical and experimental research documents the potential for chemical disruption of thyroid hormone (TH) signaling through multiple molecular targets. Perturbation of TH signaling can lead to abnormal brain development, cognitive impairments, and other adverse outcomes in humans and wildlife. To increase chemical safety screening efficiency and reduce vertebrate animal testing, in vitro assays that identify chemical interactions with molecular targets of the thyroid system have been developed and implemented. OBJECTIVES We present an adverse outcome pathway (AOP) network to link data derived from in vitro assays that measure chemical interactions with thyroid molecular targets to downstream events and adverse outcomes traditionally derived from in vivo testing. We examine the role of new in vitro technologies, in the context of the AOP network, in facilitating consideration of several important regulatory and biological challenges in characterizing chemicals that exert effects through a thyroid mechanism. DISCUSSION There is a substantial body of knowledge describing chemical effects on molecular and physiological regulation of TH signaling and associated adverse outcomes. Until recently, few alternative nonanimal assays were available to interrogate chemical effects on TH signaling. With the development of these new tools, screening large libraries of chemicals for interactions with molecular targets of the thyroid is now possible. Measuring early chemical interactions with targets in the thyroid pathway provides a means of linking adverse outcomes, which may be influenced by many biological processes, to a thyroid mechanism. However, the use of in vitro assays beyond chemical screening is complicated by continuing limits in our knowledge of TH signaling in important life stages and tissues, such as during fetal brain development. Nonetheless, the thyroid AOP network provides an ideal tool for defining causal linkages of a chemical exerting thyroid-dependent effects and identifying research needs to quantify these effects in support of regulatory decision making. https://doi.org/10.1289/EHP5297.
Collapse
Affiliation(s)
- Pamela D Noyes
- National Center for Environmental Assessment, Office of Research and Development (ORD), U.S. Environmental Protection Agency (EPA), Washington, DC, USA
| | - Katie Paul Friedman
- National Center for Computational Toxicology, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Patience Browne
- Environment Health and Safety Division, Environment Directorate, Organisation for Economic Co-operation and Development (OECD), Paris, France
| | - Jonathan T Haselman
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory (NHEERL), ORD, U.S. EPA, Duluth, Minnesota, USA
| | - Mary E Gilbert
- Toxicity Assessment Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Michael W Hornung
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory (NHEERL), ORD, U.S. EPA, Duluth, Minnesota, USA
| | - Stan Barone
- Office of Pollution Prevention and Toxics, Office of Chemical Safety and Pollution Prevention, U.S. EPA, Washington, DC, USA
| | - Kevin M Crofton
- National Center for Computational Toxicology, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Susan C Laws
- Toxicity Assessment Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Tammy E Stoker
- Toxicity Assessment Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Steven O Simmons
- National Center for Computational Toxicology, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Joseph E Tietge
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory (NHEERL), ORD, U.S. EPA, Duluth, Minnesota, USA
| | - Sigmund J Degitz
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory (NHEERL), ORD, U.S. EPA, Duluth, Minnesota, USA
| |
Collapse
|
22
|
Liu Q, Zhang Y, Wang P, Liu J, Li B, Yu Y, Wu H, Kang R, Zhang X, Wang Z. Deciphering the scalene association among type-2 diabetes mellitus, prostate cancer, and chronic myeloid leukemia via enrichment analysis of disease-gene network. Cancer Med 2019; 8:2268-2277. [PMID: 30938105 PMCID: PMC6536925 DOI: 10.1002/cam4.1845] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 09/25/2018] [Accepted: 10/06/2018] [Indexed: 12/12/2022] Open
Abstract
The potential biological relationship between type‐2 diabetes mellitus (T2DM) has been focused in numerous studies. To investigate the molecular associations among T2DM, prostate cancer (PCa), and chronic myeloid leukemia (CML), using a biomolecular network enrichment analysis. We obtained a list of disease‐related genes and constructed disease networks. Then, GO enrichment analysis was performed to identify the significant functions and pathways of overlapping modules in the Database for Annotation, Visualization and Integrated Discovery (DAVID) database. More than 75% of these overlapping genes were found to be consistent with the findings of previous studies. In the three diseases, we found that Sarcoglycan delta (SGCD) and Rho family GTPase 3 (RND3) were the overlapping genes and identified negative regulation of apoptotic process and negative regulation of transcription from RNA polymerase II promoter RNA as the two overlapping biological functions. CML and PCa were the most closely related, with 34 overlapping genes, five overlapping modules, 27 overlapping biological functions, and nine overlapping pathways. There were 13 overlapping genes, one overlapping modules, four overlapping biological functions and one overlapping pathway (FoxO signaling pathway) were found in T2DM and CML.And T2DM and PCa were the least related pair in our study, with only six overlapping genes, five overlapping modules, and one overlapping biological function. SGCD and RND3 were the main gene‐to‐gene relationship among T2DM, CML, and PCa; apoptosis, development, and transcription from RNA polymerase II promote processes were the main functional connections among T2DM, CML, and PCa by network enrichment analysis. There is a “scalene” relationship among T2DM, CML, and PCa at gene, pathway, biological process, and module levels: CML and PCa were the most closely related, the second were T2DM and PCa, and T2DM and PCa were the least related pair in our study. Our study provides a new avenue for further studies on T2DM and cancers, which may promote the discovery and development of novel therapeutic and can be used to treat multiple diseases.
Collapse
Affiliation(s)
- Qiong Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yingying Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Pengqian Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jun Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bing Li
- Institute of Information on Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanan Yu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongli Wu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruixia Kang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoxu Zhang
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
23
|
Zhang Q, Li J, Middleton A, Bhattacharya S, Conolly RB. Bridging the Data Gap From in vitro Toxicity Testing to Chemical Safety Assessment Through Computational Modeling. Front Public Health 2018; 6:261. [PMID: 30255008 PMCID: PMC6141783 DOI: 10.3389/fpubh.2018.00261] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/21/2018] [Indexed: 12/18/2022] Open
Abstract
Chemical toxicity testing is moving steadily toward a human cell and organoid-based in vitro approach for reasons including scientific relevancy, efficiency, cost, and ethical rightfulness. Inferring human health risk from chemical exposure based on in vitro testing data is a challenging task, facing various data gaps along the way. This review identifies these gaps and makes a case for the in silico approach of computational dose-response and extrapolation modeling to address many of the challenges. Mathematical models that can mechanistically describe chemical toxicokinetics (TK) and toxicodynamics (TD), for both in vitro and in vivo conditions, are the founding pieces in this regard. Identifying toxicity pathways and in vitro point of departure (PoD) associated with adverse health outcomes requires an understanding of the molecular key events in the interacting transcriptome, proteome, and metabolome. Such an understanding will in turn help determine the sets of sensitive biomarkers to be measured in vitro and the scope of toxicity pathways to be modeled in silico. In vitro data reporting both pathway perturbation and chemical biokinetics in the culture medium serve to calibrate the toxicity pathway and virtual tissue models, which can then help predict PoDs in response to chemical dosimetry experienced by cells in vivo. Two types of in vitro to in vivo extrapolation (IVIVE) are needed. (1) For toxic effects involving systemic regulations, such as endocrine disruption, organism-level adverse outcome pathway (AOP) models are needed to extrapolate in vitro toxicity pathway perturbation to in vivo PoD. (2) Physiologically-based toxicokinetic (PBTK) modeling is needed to extrapolate in vitro PoD dose metrics into external doses for expected exposure scenarios. Linked PBTK and TD models can explore the parameter space to recapitulate human population variability in response to chemical insults. While challenges remain for applying these modeling tools to support in vitro toxicity testing, they open the door toward population-stratified and personalized risk assessment.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Jin Li
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, United Kingdom
| | - Alistair Middleton
- Unilever, Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, United Kingdom
| | - Sudin Bhattacharya
- Biomedical Engineering, Michigan State University, East Lansing, MI, United States
| | - Rory B Conolly
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, United States Environmental Protection Agency, Durham, NC, United States
| |
Collapse
|
24
|
Piñero J, Furlong LI, Sanz F. In silico models in drug development: where we are. Curr Opin Pharmacol 2018; 42:111-121. [PMID: 30205360 DOI: 10.1016/j.coph.2018.08.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/30/2018] [Accepted: 08/13/2018] [Indexed: 02/07/2023]
Abstract
The use and utility of computational models in drug development has significantly grown in the last decades, fostered by the availability of high throughput datasets and new data analysis strategies. These in silico approaches are demonstrating their ability to generate reliable predictions as well as new knowledge on the mode of action of drugs and the mechanisms underlying their side effects, altogether helping to reduce the costs of drug development. The aim of this review is to provide a panorama of developments in the field in the last two years.
Collapse
Affiliation(s)
- Janet Piñero
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra (UPF), Carrer Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Laura I Furlong
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra (UPF), Carrer Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Ferran Sanz
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra (UPF), Carrer Dr. Aiguader 88, 08003 Barcelona, Spain.
| |
Collapse
|
25
|
Qutob SS, Chauhan V, Kuo B, Williams A, Yauk CL, McNamee JP, Gollapudi B. The application of transcriptional benchmark dose modeling for deriving thresholds of effects associated with solar-simulated ultraviolet radiation exposure. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2018; 59:502-515. [PMID: 29761935 PMCID: PMC6099464 DOI: 10.1002/em.22196] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 03/02/2018] [Accepted: 03/16/2018] [Indexed: 06/08/2023]
Abstract
Considerable data has been generated to elucidate the transcriptional response of cells to ultraviolet radiation (UVR) exposure providing a mechanistic understanding of UVR-induced cellular responses. However, using these data to support standards development has been challenging. In this study, we apply benchmark dose (BMD) modeling of transcriptional data to derive thresholds of gene responsiveness following exposure to solar-simulated UVR. Human epidermal keratinocytes were exposed to three doses (10, 20, 150 kJ/m2 ) of solar simulated UVR and assessed for gene expression changes 6 and 24 hr postexposure. The dose-response curves for genes with p-fit values (≥ 0.1) were used to derive BMD values for genes and pathways. Gene BMDs were bi-modally distributed, with a peak at ∼16 kJ/m2 and ∼108 kJ/m2 UVR exposure. Genes/pathways within Mode 1 were involved in cell signaling and DNA damage response, while genes/pathways in the higher Mode 2 were associated with immune response and cancer development. The median value of each Mode coincides with the current human exposure limits for UVR and for the minimal erythemal dose, respectively. Such concordance implies that the use of transcriptional BMD data may represent a promising new approach for deriving thresholds of actinic effects. Environ. Mol. Mutagen. 59:502-515, 2018. © 2018 The Authors Environmental and Molecular Mutagenesis published by Wiley Periodicals, Inc. on behalf of Environmental Mutagen Society.
Collapse
Affiliation(s)
- Sami S. Qutob
- Consumer and Clinical Radiation Protection BureauHealth CanadaOttawaOntarioK1A 1C1Canada
| | - Vinita Chauhan
- Consumer and Clinical Radiation Protection BureauHealth CanadaOttawaOntarioK1A 1C1Canada
| | - Byron Kuo
- Environmental Health Science and Research Bureau, Health CanadaOttawaOntarioCanada
| | - Andrew Williams
- Environmental Health Science and Research Bureau, Health CanadaOttawaOntarioCanada
| | - Carole L. Yauk
- Environmental Health Science and Research Bureau, Health CanadaOttawaOntarioCanada
| | - James P. McNamee
- Consumer and Clinical Radiation Protection BureauHealth CanadaOttawaOntarioK1A 1C1Canada
| | | |
Collapse
|
26
|
Liu Z, Delavan B, Roberts R, Tong W. Transcriptional Responses Reveal Similarities Between Preclinical Rat Liver Testing Systems. Front Genet 2018; 9:74. [PMID: 29616076 PMCID: PMC5870427 DOI: 10.3389/fgene.2018.00074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 02/19/2018] [Indexed: 01/03/2023] Open
Abstract
Toxicogenomics (TGx) is an important tool to gain an enhanced understanding of toxicity at the molecular level. Previously, we developed a pair ranking (PRank) method to assess in vitro to in vivo extrapolation (IVIVE) using toxicogenomic datasets from the Open Toxicogenomics Project-Genomics Assisted Toxicity Evaluation System (TG-GATEs) database. With this method, we investiagted three important questions that were not addressed in our previous study: (1) is a 1-day in vivo short-term assay able to replace the 28-day standard and expensive toxicological assay? (2) are some biological processes more conservative across different preclinical testing systems than others? and (3) do these preclinical testing systems have the similar resolution in differentiating drugs by their therapeutic uses? For question 1, a high similarity was noted (PRank score = 0.90), indicating the potential utility of shorter term in vivo studies to predict outcome in longer term and more expensive in vivo model systems. There was a moderate similarity between rat primary hepatocytes and in vivo repeat-dose studies (PRank score = 0.71) but a low similarity (PRank score = 0.56) between rat primary hepatocytes and in vivo single dose studies. To address question 2, we limited the analysis to gene sets relevant to specific toxicogenomic pathways and we found that pathways such as lipid metabolism were consistently over-represented in all three assay systems. For question 3, all three preclinical assay systems could distinguish compounds from different therapeutic categories. This suggests that any noted differences in assay systems was biological process-dependent and furthermore that all three systems have utility in assessing drug responses within a certain drug class. In conclusion, this comparison of three commonly used rat TGx systems provides useful information in utility and application of TGx assays.
Collapse
Affiliation(s)
- Zhichao Liu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States
| | - Brian Delavan
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States.,Department of Biosciences, University of Arkansas at Little Rock, Little Rock, AR, United States
| | - Ruth Roberts
- ApconiX, Alderley Edge, United Kingdom.,Department of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Weida Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States
| |
Collapse
|
27
|
Vinken M, Knapen D, Vergauwen L, Hengstler JG, Angrish M, Whelan M. Adverse outcome pathways: a concise introduction for toxicologists. Arch Toxicol 2017; 91:3697-3707. [PMID: 28660287 DOI: 10.1007/s00204-017-2020-z] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 06/22/2017] [Indexed: 12/14/2022]
Abstract
Adverse outcome pathways (AOPs) are designed to provide a clear-cut mechanistic representation of critical toxicological effects that propagate over different layers of biological organization from the initial interaction of a chemical with a molecular target to an adverse outcome at the individual or population level. Adverse outcome pathways are currently gaining momentum, especially in view of their many potential applications as pragmatic tools in the fields of human toxicology, ecotoxicology, and risk assessment. A number of guidance documents, issued by the Organization for Economic Cooperation and Development, as well as landmark papers, outlining best practices to develop, assess and use AOPs, have been published in the last few years. The present paper provides a synopsis of the main principles related to the AOP framework for the toxicologist less familiar with this area, followed by two case studies relevant for human toxicology and ecotoxicology.
Collapse
Affiliation(s)
- Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| | - Dries Knapen
- Zebrafishlab, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Lucia Vergauwen
- Zebrafishlab, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium.,Systemic Physiological and Ecotoxicological Research (SPHERE), Department of Biology, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, 44139, Dortmund, Germany
| | - Michelle Angrish
- National Center for Environmental Assessment, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, NC, 27709, USA
| | - Maurice Whelan
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| |
Collapse
|