Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 385
Filter
1.
Environ Sci Technol ; 57(44): 16823-16833, 2023 11 07.
Article in English | MEDLINE | ID: mdl-37874250

ABSTRACT

Haloacetaldehydes (HALs) represent the third-largest category of disinfection byproducts (DBPs) in drinking water in terms of weight. As a subset of unregulated DBPs, only a few HALs have undergone assessment, yielding limited information regarding their genotoxicity mechanisms. Herein, we developed a simplified yeast-based toxicogenomics assay to evaluate the genotoxicity of five specific HALs. This assay recorded the protein expression profiles of eight Saccharomyces cerevisiae strains fused with green fluorescent protein, including all known DNA damage and repair pathways. High-resolution real-time pathway activation data and protein expression profiles in conjunction with clustering analysis revealed that the five HALs induced various DNA damage and repair pathways. Among these, chloroacetaldehyde and trichloroacetaldehyde were found to be positively associated with genotoxicity, while dichloroacetaldehyde, bromoacetaldehyde, and tribromoacetaldehyde displayed negative associations. The protein effect level index, which are molecular end points derived from a toxicogenomics assay, exhibited a statistically significant positive correlation with the results of traditional genotoxicity assays, such as the comet assay (rp = 0.830 and p < 0.001) and SOS/umu assay (rp = 0.786 and p = 0.004). This yeast-based toxicogenomics assay, which employs a minimal set of gene biomarkers, can be used for mechanistic genotoxicity screening and assessment of HALs and other chemical compounds. These results contribute to bridging the knowledge gap regarding the molecular mechanisms underlying the genotoxicity of HALs and enable the categorization of HALs based on their distinct DNA damage and repair mechanisms.


Subject(s)
Disinfectants , Water Pollutants, Chemical , Water Purification , Disinfection/methods , Saccharomyces cerevisiae/genetics , Toxicogenetics/methods , Water Purification/methods , DNA Damage , Water Pollutants, Chemical/analysis , Disinfectants/analysis , Disinfectants/chemistry
2.
Ecotoxicol Environ Saf ; 250: 114466, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36587411

ABSTRACT

BACKGROUND: Given the increasing exposure of humans to environmental chemicals and the limitations of conventional toxicity test, there is an urgent need to develop next-generation risk assessment methods. OBJECTIVES: This study aims to establish a novel computational system named Toxicogenomics Scoring System (TGSS) to predict the carcinogenicity of chemicals coupling chemical-gene interactions with multiple cancer transcriptomic datasets. METHODS: Chemical-related gene signatures were derived from chemical-gene interaction data from the Comparative Toxicogenomics Database (CTD). For each cancer type in TCGA, genes were ranked by their effects on tumorigenesis, which is based on the differential expression between tumor and normal samples. Next, we developed carcinogenicity scores (C-scores) using pre-ranked GSEA to quantify the correlation between chemical-related gene signatures and ranked gene lists. Then we established TGSS by systematically evaluating the C-scores in multiple chemical-tumor pairs. Furthermore, we examined the performance of our approach by ROC curves or prognostic analyses in TCGA and multiple independent cancer cohorts. RESULTS: Forty-six environmental chemicals were finally included in the study. C-score was calculated for each chemical-tumor pair. The C-scores of IARC Group 3 chemicals were significantly lower than those of chemicals in Group 1 (P-value = 0.02) and Group 2 (P-values = 7.49 ×10-5). ROC curves analysis indicated that C-score could distinguish "high-risk chemicals" from the other compounds (AUC = 0.67) with a specificity and sensitivity of 0.86 and 0.57. The results of survival analysis were also in line with the assessed carcinogenicity in TGSS for the chemicals in Group 1. Finally, consistent results were further validated in independent cancer cohorts. CONCLUSION: TGSS highlighted the great potential of integrating chemical-gene interactions with gene-cancer relationships to predict the carcinogenic risk of chemicals, which would be valuable for systems toxicology.


Subject(s)
Neoplasms , Toxicogenetics , Humans , Toxicogenetics/methods , Carcinogens/toxicity , Neoplasms/chemically induced , Neoplasms/genetics , Cell Transformation, Neoplastic , Risk Assessment
3.
Front Biosci (Landmark Ed) ; 27(10): 294, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36336867

ABSTRACT

Environmental toxicogenomics aims to collect, analyze and interpret data on changes in gene expression and protein activity resulting from exposure to toxic substances using high-performance omics technologies. Molecular profiling methods such as genomics, transcriptomics, proteomics, metabolomics, and bioinformatics techniques, permit the simultaneous analysis of a multitude of gene variants in an organism exposed to toxic agents to search for genes prone to damage, detect patterns and mechanisms of toxicity, and identify specific gene expression profiles that can provide biomarkers of exposure and risk. Compared to previous approaches to measuring molecular changes caused by toxicants, toxicogenomic technologies can improve environmental risk assessment while reducing animal studies. We discuss the prospects and limitations of converting omic datasets into valuable information, focusing on assessing the risks of mixed toxic substances to the environment and human health.


Subject(s)
Genomics , Toxicogenetics , Animals , Humans , Toxicogenetics/methods , Genomics/methods , Proteomics/methods , Computational Biology , Metabolomics
4.
Environ Sci Technol ; 56(22): 15960-15968, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36268973

ABSTRACT

Transcriptomics dose-response analysis (TDRA) has emerged as a promising approach for integrating toxicogenomics data into a risk assessment context; however, variability and uncertainty associated with experimental design are not well understood. Here, we evaluated n = 55 RNA-seq profiles derived from Japanese quail liver tissue following exposure to chlorpyrifos (0, 0.04, 0.1, 0.2, 0.4, 1, 2, 4, 10, 20, and 40 µg/g; n = 5 replicates per group) via egg injection. The full dataset was subsampled 637 times to generate smaller datasets with different dose ranges and spacing (designs A-E) and number of replicates (n = 2-5). TDRA of the 637 datasets revealed substantial variability in the gene and pathway benchmark doses, but relative stability in overall transcriptomic point-of-departure (tPOD) values when tPODs were calculated with the "pathway" and "mode" methods. Further, we found that tPOD values were more dependent on the dose range and spacing than on the number of replicates, suggesting that optimal experimental designs should use fewer replicates (n = 2 or 3) and more dose groups to reduce uncertainty in the results. Finally, tPOD values ranged by over ten times for all surveyed experimental designs and tPOD types, suggesting that tPODs should be interpreted as order-of-magnitude estimates.


Subject(s)
Coturnix , Transcriptome , Animals , Uncertainty , Dose-Response Relationship, Drug , Toxicogenetics/methods , Risk Assessment/methods
5.
Ecotoxicol Environ Saf ; 240: 113678, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35653977

ABSTRACT

This study compared the toxicity mechanisms of four X-ray-based iodinated contrast media (ICM) on Escherichia coli (E. coli) and yeast microarray assays, aiming to determine the diverse toxicity mechanisms among different exposed organisms and the relationship between toxicity and their physical and chemical characteristics. The conventional phenotypic endpoint cytotoxicity and the change in reactive oxygen species (ROS) level were employed in conjunction with toxicogenomics to quantify changes in the gene/protein biomarker level in the regulation of different damage/repair pathways. The results showed that molecular toxicity endpoints, named transcriptional effect level index (TELI) and protein effect level index (PELI) for E. coli and yeast, respectively, correlated well with the phenotypic endpoints. Temporal altered gene/protein expression profiles revealed dynamic and complex damage/toxic mechanisms. In particular, compared with E. coli cells, yeast cells exposed to ICM exhibited significantly higher stress intensity and diverse stress types, resulting in stress or damage to the organism. The toxic mechanisms of ICM are concentration/property-dependent and relevant to the cellular structure and defense systems in prokaryotes and eukaryotes. In particular, the toxicity of ionic ICM is higher than that of non-ionic ICM, and eukaryotes are more susceptible than prokaryotes to ICM exposure.


Subject(s)
Contrast Media , Escherichia coli , Contrast Media/toxicity , Escherichia coli/genetics , Saccharomyces cerevisiae/genetics , Toxicogenetics/methods , X-Rays
6.
Nihon Yakurigaku Zasshi ; 157(3): 200-206, 2022.
Article in Japanese | MEDLINE | ID: mdl-35491119

ABSTRACT

We are constructing the "Percellome Database" containing many transcriptomes of mice exposed to a series of chemicals to elucidate the molecular mechanism of toxicity and to develop toxicity prediction technology. Acute toxicity of a chemical can be predicted to a certain extent by searching the similarity of the transcriptomes obtained by the single-dose exposure experiments. In addition, we are analyzing the relation between the transcriptome and the epigenome i.e. histone modification and genomic DNA methylation to understand the molecular mechanism of the repeated dose toxicity. We are attempting to expand the scale and improve the efficiency of the analysis by introducing artificial intelligence technologies. This approach should maximize the use of toxicogenomics technology for optimizing the experimental protocols for repeated dose toxicity studies towards 3Rs principle, and optimizing the process of in silico toxicity prediction by combining the available big data.


Subject(s)
Artificial Intelligence , Transcriptome , Animals , Epigenesis, Genetic , Genomics , Mice , Toxicogenetics/methods
7.
Toxicol Sci ; 186(2): 242-259, 2022 03 28.
Article in English | MEDLINE | ID: mdl-34971401

ABSTRACT

Animal studies are a critical component in biomedical research, pharmaceutical product development, and regulatory submissions. There is a worldwide effort in toxicology toward "reducing, refining, and replacing" animal use. Here, we proposed a deep generative adversarial network (GAN)-based framework capable of deriving new animal results from existing animal studies without additional experiments. To prove the concept, we employed this Tox-GAN framework to generate both gene activities and expression profiles for multiple doses and treatment durations in toxicogenomics (TGx). Using the pre-existing rat liver TGx data from the Open Toxicogenomics Project-Genomics-Assisted Toxicity Evaluation System (Open TG-GATES), we generated Tox-GAN transcriptomic profiles with high similarity (0.997 ± 0.002 in intensity and 0.740 ± 0.082 in fold change) to the corresponding real gene expression profiles. Consequently, Tox-GAN showed an outstanding performance in 2 critical TGx applications, gaining a molecular understanding of underlying toxicological mechanisms and gene expression-based biomarker development. For the former, over 87% agreement in Gene Ontology was found between Tox-GAN results and real gene expression data. For the latter, the concordance of biomarkers between real and generated data was high in both predictive performance and biomarker genes. We also demonstrated that the Tox-GAN models constructed with the Open TG-GATES data were capable of generating transcriptomic profiles reported in DrugMatrix. Finally, we demonstrated potential utility for Tox-GAN in aiding chemical-based read-across. To the best of our knowledge, the proposed Tox-GAN model is novel in its ability to generate in vivo transcriptomic profiles at different treatment conditions from chemical structures. Overall, Tox-GAN holds great promise for generating high-quality toxicogenomic profiles without animal experimentation.


Subject(s)
Artificial Intelligence , Toxicogenetics , Animals , Biomarkers , Genomics , Rats , Toxicogenetics/methods , Transcriptome
8.
Int J Mol Sci ; 22(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638921

ABSTRACT

The mass production of graphene oxide (GO) unavoidably elevates the chance of human exposure, as well as the possibility of release into the environment with high stability, raising public concern as to its potential toxicological risks and the implications for humans and ecosystems. Therefore, a thorough assessment of GO toxicity, including its potential reliance on key physicochemical factors, which is lacking in the literature, is of high significance and importance. In this study, GO toxicity, and its dependence on oxidation level, elemental composition, and size, were comprehensively assessed. A newly established quantitative toxicogenomic-based toxicity testing approach, combined with conventional phenotypic bioassays, were employed. The toxicogenomic assay utilized a GFP-fused yeast reporter library covering key cellular toxicity pathways. The results reveal that, indeed, the elemental composition and size do exert impacts on GO toxicity, while the oxidation level exhibits no significant effects. The UV-treated GO, with significantly higher carbon-carbon groups and carboxyl groups, showed a higher toxicity level, especially in the protein and chemical stress categories. With the decrease in size, the toxicity level of the sonicated GOs tended to increase. It is proposed that the covering and subsequent internalization of GO sheets might be the main mode of action in yeast cells.


Subject(s)
Environmental Pollutants/toxicity , Graphite/toxicity , Nanostructures/toxicity , Toxicity Tests/methods , Toxicogenetics/methods , A549 Cells , Cluster Analysis , Comet Assay/methods , DNA Damage , Environmental Pollutants/chemistry , Graphite/chemistry , Humans , Microscopy, Electron, Scanning/methods , Nanostructures/chemistry , Nanostructures/ultrastructure , Oxidation-Reduction/drug effects , Photoelectron Spectroscopy/methods , Proteome/classification , Proteome/drug effects , Proteomics/methods , Reactive Oxygen Species/metabolism , Yeasts/cytology , Yeasts/drug effects , Yeasts/metabolism
9.
Arch Toxicol ; 95(12): 3745-3775, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34626214

ABSTRACT

Mechanism-based risk assessment is urged to advance and fully permeate into current safety assessment practices, possibly at early phases of drug safety testing. Toxicogenomics is a promising source of mechanisms-revealing data, but interpretative analysis tools specific for the testing systems (e.g. hepatocytes) are lacking. In this study, we present the TXG-MAPr webtool (available at https://txg-mapr.eu/WGCNA_PHH/TGGATEs_PHH/ ), an R-Shiny-based implementation of weighted gene co-expression network analysis (WGCNA) obtained from the Primary Human Hepatocytes (PHH) TG-GATEs dataset. The 398 gene co-expression networks (modules) were annotated with functional information (pathway enrichment, transcription factor) to reveal their mechanistic interpretation. Several well-known stress response pathways were captured in the modules, were perturbed by specific stressors and showed preservation in rat systems (rat primary hepatocytes and rat in vivo liver), with the exception of DNA damage and oxidative stress responses. A subset of 87 well-annotated and preserved modules was used to evaluate mechanisms of toxicity of endoplasmic reticulum (ER) stress and oxidative stress inducers, including cyclosporine A, tunicamycin and acetaminophen. In addition, module responses can be calculated from external datasets obtained with different hepatocyte cells and platforms, including targeted RNA-seq data, therefore, imputing biological responses from a limited gene set. As another application, donors' sensitivity towards tunicamycin was investigated with the TXG-MAPr, identifying higher basal level of intrinsic immune response in donors with pre-existing liver pathology. In conclusion, we demonstrated that gene co-expression analysis coupled to an interactive visualization environment, the TXG-MAPr, is a promising approach to achieve mechanistic relevant, cross-species and cross-platform evaluation of toxicogenomic data.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Hepatocytes/drug effects , Risk Assessment/methods , Toxicogenetics/methods , Acetaminophen/toxicity , Animals , Chemical and Drug Induced Liver Injury/genetics , Cyclosporine/toxicity , Datasets as Topic , Endoplasmic Reticulum Stress/drug effects , Gene Expression Profiling , Gene Regulatory Networks , Hepatocytes/pathology , Humans , Oxidative Stress/drug effects , Rats , Species Specificity , Tunicamycin/toxicity
10.
Methods Mol Biol ; 2326: 67-94, 2021.
Article in English | MEDLINE | ID: mdl-34097262

ABSTRACT

Gene expression analysis has been becoming a popular method for studying gene function and response to different environmental stresses, including toxin/pollution exposure. Selection of a suitable reference gene is critically important for gene expression analysis due to that wrong reference genes will cause misleading and even wrong conclusion. A good reference gene should be a more stable reference gene, particularly during the toxicant exposure treatment and/or other investigation condition. In this chapter, a step-by-step protocol is present for primer design, reverse transcription PCR, primer efficiency and specificity test, qRT-PCR, and the strategy for identifying most stable reference genes for toxicogenomic and gene expression analysis. The detailed method for determining the primer gene specificity and primer efficiency are also presented in this chapter. Low primer efficiency will affect the fold changes during gene expression analysis; however, it does not affect the conclusion, up- or downregulation. Choosing a wrong reference gene may result in wrong conclusion.


Subject(s)
Gene Expression Profiling/methods , Genomics/methods , Toxicity Tests/methods , Cell Culture Techniques/methods , Gene Expression/drug effects , Genes, Essential/drug effects , Humans , MCF-7 Cells , Polymerase Chain Reaction/methods , Toxicogenetics/methods
11.
Adv Sci (Weinh) ; 8(10): 2004588, 2021 05.
Article in English | MEDLINE | ID: mdl-34026454

ABSTRACT

Toxicogenomics opens novel opportunities for hazard assessment by utilizing computational methods to map molecular events and biological processes. In this study, the transcriptomic and immunopathological changes associated with airway exposure to a total of 28 engineered nanomaterials (ENM) are investigated. The ENM are selected to have different core (Ag, Au, TiO2, CuO, nanodiamond, and multiwalled carbon nanotubes) and surface chemistries (COOH, NH2, or polyethylene glycosylation (PEG)). Additionally, ENM with variations in either size (Au) or shape (TiO2) are included. Mice are exposed to 10 µg of ENM by oropharyngeal aspiration for 4 consecutive days, followed by extensive histological/cytological analyses and transcriptomic characterization of lung tissue. The results demonstrate that transcriptomic alterations are correlated with the inflammatory cell infiltrate in the lungs. Surface modification has varying effects on the airways with amination rendering the strongest inflammatory response, while PEGylation suppresses toxicity. However, toxicological responses are also dependent on ENM core chemistry. In addition to ENM-specific transcriptional changes, a subset of 50 shared differentially expressed genes is also highlighted that cluster these ENM according to their toxicity. This study provides the largest in vivo data set currently available and as such provides valuable information to be utilized in developing predictive models for ENM toxicity.


Subject(s)
Lung/drug effects , Nanostructures/toxicity , Toxicogenetics/methods , Animals , Female , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Models, Animal , Nanostructures/chemistry , Nanostructures/classification , Transcriptome
12.
Molecules ; 26(5)2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33807638

ABSTRACT

Silica nanoparticles are a class of molecules commonly used in drug or gene delivery systems that either facilitate the delivery of therapeutics to specific drug targets or enable the efficient delivery of constructed gene products into biological systems. Some in vivo or in vitro studies have demonstrated the toxic effects of silica nanoparticles. Despite the availability of risk management tools in response to the growing use of synthetic silica in commercial products, the molecular mechanism of toxicity induced by silica nanoparticles is not well characterized. The purpose of this study was to elucidate the effects of silica nanoparticle exposure in three types of cells including human aortic endothelial cells, mouse-derived macrophages, and A549 non-small cell lung cancer cells using toxicogenomic analysis. The results indicated that among all three cell types, the TNF and MAPK signaling pathways were the common pathways upregulated by silica nanoparticles. These findings may provide insight into the effects of silica nanoparticle exposure in the human body and the possible mechanism of toxicity.


Subject(s)
Gene Expression Regulation/drug effects , Nanoparticles/chemistry , Signal Transduction/drug effects , Silicon Dioxide/pharmacology , A549 Cells , Animals , Cell Survival/drug effects , Cells, Cultured , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Macrophages/cytology , Macrophages/drug effects , Mice , Nanoparticles/toxicity , RAW 264.7 Cells , Signal Transduction/genetics , Silicon Dioxide/chemistry , Silicon Dioxide/toxicity , Toxicogenetics/methods , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/metabolism
13.
NPJ Syst Biol Appl ; 7(1): 7, 2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33504769

ABSTRACT

The ToxCast in vitro screening program has provided concentration-response bioactivity data across more than a thousand assay endpoints for thousands of chemicals found in our environment and commerce. However, most ToxCast screening assays have evaluated individual biological targets in cancer cell lines lacking integrated physiological functionality (such as receptor signaling, metabolism). We evaluated differentiated HepaRGTM cells, a human liver-derived cell model understood to effectively model physiologically relevant hepatic signaling. Expression of 93 gene transcripts was measured by quantitative polymerase chain reaction using Fluidigm 96.96 dynamic arrays in response to 1060 chemicals tested in eight-point concentration-response. A Bayesian framework quantitatively modeled chemical-induced changes in gene expression via six transcription factors including: aryl hydrocarbon receptor, constitutive androstane receptor, pregnane X receptor, farnesoid X receptor, androgen receptor, and peroxisome proliferator-activated receptor alpha. For these chemicals the network model translates transcriptomic data into Bayesian inferences about molecular targets known to activate toxicological adverse outcome pathways. These data also provide new insights into the molecular signaling network of HepaRGTM cell cultures.


Subject(s)
Hepatocytes/drug effects , High-Throughput Screening Assays/methods , Toxicogenetics/methods , Bayes Theorem , Cell Culture Techniques , Cell Line , Humans , Liver/cytology , Small Molecule Libraries , Transcription Factors/drug effects , Transcriptome/genetics
14.
Brief Bioinform ; 22(1): 428-437, 2021 01 18.
Article in English | MEDLINE | ID: mdl-31838506

ABSTRACT

Identifying hepatotoxicity as early as possible is significant in drug development. In this study, we developed a drug-induced hepatotoxicity prediction model taking account of both the biological context and the computational efficacy based on toxicogenomics data. Specifically, we proposed a novel gene selection algorithm considering gene's participation, named BioCB, to choose the discriminative genes and make more efficient prediction. Then instead of using the raw gene expression levels to characterize each drug, we developed a two-dimensional biological process feature pattern map to represent each drug. Then we employed two strategies to handle the maps and identify the hepatotoxicity, the direct use of maps, named Two-dim branch, and vectorization of maps, named One-dim branch. The two strategies subsequently used the deep convolutional neural networks and LightGBM as predictors, respectively. Additionally, we here for the first time proposed a stacked vectorized gene matrix, which was more predictive than the raw gene matrix. Results validated on both in vivo and in vitro data from two public data sets, the TG-GATES and DrugMatrix, show that the proposed One-dim branch outperforms the deep framework, the Two-dim branch, and has achieved high accuracy and efficiency. The implementation of the proposed method is available at https://github.com/RanSuLab/Hepatotoxicity.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Drug Development/methods , Genomics/methods , Toxicogenetics/methods , Humans , Software
15.
Nucleic Acids Res ; 49(D1): D1138-D1143, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33068428

ABSTRACT

The public Comparative Toxicogenomics Database (CTD; http://ctdbase.org/) is an innovative digital ecosystem that relates toxicological information for chemicals, genes, phenotypes, diseases, and exposures to advance understanding about human health. Literature-based, manually curated interactions are integrated to create a knowledgebase that harmonizes cross-species heterogeneous data for chemical exposures and their biological repercussions. In this biennial update, we report a 20% increase in CTD curated content and now provide 45 million toxicogenomic relationships for over 16 300 chemicals, 51 300 genes, 5500 phenotypes, 7200 diseases and 163 000 exposure events, from 600 comparative species. Furthermore, we increase the functionality of chemical-phenotype content with new data-tabs on CTD Disease pages (to help fill in knowledge gaps for environmental health) and new phenotype search parameters (for Batch Query and Venn analysis tools). As well, we introduce new CTD Anatomy pages that allow users to uniquely explore and analyze chemical-phenotype interactions from an anatomical perspective. Finally, we have enhanced CTD Chemical pages with new literature-based chemical synonyms (to improve querying) and added 1600 amino acid-based compounds (to increase chemical landscape). Together, these updates continue to augment CTD as a powerful resource for generating testable hypotheses about the etiologies and molecular mechanisms underlying environmentally influenced diseases.


Subject(s)
Databases, Factual , Gene-Environment Interaction , Genome, Human/drug effects , Genomics/methods , Prescription Drugs/pharmacology , Xenobiotics/toxicity , Databases, Chemical , Databases, Genetic , Genotype , Humans , Internet , Knowledge Bases , Organ Specificity , Phenotype , Prescription Drugs/chemistry , Software , Toxicogenetics/methods , Xenobiotics/chemistry
16.
Toxicol Appl Pharmacol ; 406: 115237, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32920000

ABSTRACT

Improvement of COVID-19 clinical condition was seen in studies where combination of antiretroviral drugs, lopinavir and ritonavir, as well as immunomodulant antimalaric, chloroquine/hydroxychloroquine together with the macrolide-type antibiotic, azithromycin, was used for patient's treatment. Although these drugs are "old", their pharmacological and toxicological profile in SARS-CoV-2 - infected patients are still unknown. Thus, by using in silico toxicogenomic data-mining approach, we aimed to assess both risks and benefits of the COVID-19 treatment with the most promising candidate drugs combinations: lopinavir/ritonavir and chloroquine/hydroxychloroquine + azithromycin. The Comparative Toxicogenomics Database (CTD; http://CTD.mdibl.org), Cytoscape software (https://cytoscape.org) and ToppGene Suite portal (https://toppgene.cchmc.org) served as a foundation in our research. Our results have demonstrated that lopinavir/ritonavir increased the expression of the genes involved in immune response and lipid metabolism (IL6, ICAM1, CCL2, TNF, APOA1, etc.). Chloroquine/hydroxychloroquine + azithromycin interacted with 6 genes (CCL2, CTSB, CXCL8, IL1B, IL6 and TNF), whereas chloroquine and azithromycin affected two additional genes (BCL2L1 and CYP3A4), which might be a reason behind a greater number of consequential diseases. In contrast to lopinavir/ritonavir, chloroquine/hydroxychloroquine + azithromycin downregulated the expression of TNF and IL6. As expected, inflammation, cardiotoxicity, and dyslipidaemias were revealed as the main risks of lopinavir/ritonavir treatment, while chloroquine/hydroxychloroquine + azithromycin therapy was additionally linked to gastrointestinal and skin diseases. According to our results, these drug combinations should be administrated with caution to patients suffering from cardiovascular problems, autoimmune diseases, or acquired and hereditary lipid disorders.


Subject(s)
Betacoronavirus , Computer Simulation , Data Mining/methods , Toxicogenetics/methods , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Azithromycin/administration & dosage , Azithromycin/adverse effects , COVID-19 , Chloroquine/administration & dosage , Chloroquine/adverse effects , Coronavirus Infections/drug therapy , Coronavirus Infections/genetics , Databases, Genetic , Drug Therapy, Combination , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/genetics , Humans , Hydroxychloroquine/administration & dosage , Hydroxychloroquine/adverse effects , Lopinavir/administration & dosage , Lopinavir/adverse effects , Pandemics , Pneumonia, Viral/drug therapy , Pneumonia, Viral/genetics , Ritonavir/administration & dosage , Ritonavir/adverse effects , SARS-CoV-2 , COVID-19 Drug Treatment
17.
Toxicology ; 442: 152530, 2020 09.
Article in English | MEDLINE | ID: mdl-32599119

ABSTRACT

Kidney injury caused by disease, trauma, environmental exposures, or drugs may result in decreased renal function, chronic kidney disease, or acute kidney failure. Diagnosis of kidney injury using serum creatinine levels, a common clinical test, only identifies renal dysfunction after the kidneys have undergone severe damage. Other indicators sensitive to kidney injury, such as the level of urine kidney injury molecule-1 (KIM-1), lack the ability to differentiate between injury phenotypes. To address early detection as well as detailed categorization of kidney-injury phenotypes in preclinical animal or cellular studies, we previously identified eight sets (modules) of co-expressed genes uniquely associated with kidney histopathology. Here, we used mercuric chloride (HgCl2)-a model nephrotoxicant-to chemically induce kidney injuries as monitored by KIM-1 levels in Sprague Dawley rats at two doses (0.25 or 0.50 mg/kg) and two exposure lengths (10 or 34 h). We collected whole transcriptome RNA-seq data derived from five animals at each dose and time point to perform a toxicogenomics analysis. Consistent with documented injury phenotypes for HgCl2 toxicity, our kidney-injury-module approach identified the onset of necrosis and dilation as early as 10 h after a dose of 0.50 mg/kg that produced only mild injury as judged by urinary KIM-1 excretion. The results of these animal studies highlight the potential of the kidney-injury-module approach to provide a sensitive and histopathology-specific readout of renal toxicity.


Subject(s)
Kidney Diseases/chemically induced , Kidney Diseases/pathology , Mercuric Chloride/toxicity , Toxicogenetics/methods , Animals , Aspartate Aminotransferases/blood , Base Sequence , Biomarkers/urine , Body Weight/drug effects , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/urine , Gene Expression/drug effects , Male , Necrosis , Protein Folding/drug effects , Rats , Rats, Sprague-Dawley
18.
Toxicol In Vitro ; 66: 104877, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32387679

ABSTRACT

When considering toxic chemicals in the environment, a mechanistic, causal explanation of toxicity may be preferred over a statistical or machine learning-based prediction by itself. Elucidating a mechanism of toxicity is, however, a costly and time-consuming process that requires the participation of specialists from a variety of fields, often relying on animal models. We present an innovative mechanistic inference framework (MechSpy), which can be used as a hypothesis generation aid to narrow the scope of mechanistic toxicology analysis. MechSpy generates hypotheses of the most likely mechanisms of toxicity, by combining a semantically-interconnected knowledge representation of human biology, toxicology and biochemistry with gene expression time series on human tissue. Using vector representations of biological entities, MechSpy seeks enrichment in a manually curated list of high-level mechanisms of toxicity, represented as biochemically- and causally-linked ontology concepts. Besides predicting the canonical mechanism of toxicity for many well-studied compounds, we experimentally validated some of our predictions for other chemicals without an established mechanism of toxicity. This mechanistic inference framework is an advantageous tool for predictive toxicology, and the first of its kind to produce a mechanistic explanation for each prediction. MechSpy can be modified to include additional mechanisms of toxicity, and is generalizable to other types of mechanisms of human biology.


Subject(s)
Toxicogenetics/methods , Cell Line , Computational Biology/methods , Gene Expression , Genomics , Humans , Software
19.
Nucleic Acids Res ; 48(W1): W455-W462, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32421831

ABSTRACT

In the past few decades, major initiatives have been launched around the world to address chemical safety testing. These efforts aim to innovate and improve the efficacy of existing methods with the long-term goal of developing new risk assessment paradigms. The transcriptomic and toxicological profiling of mammalian cells has resulted in the creation of multiple toxicogenomic datasets and corresponding tools for analysis. To enable easy access and analysis of these valuable toxicogenomic data, we have developed ToxicoDB (toxicodb.ca), a free and open cloud-based platform integrating data from large in vitro toxicogenomic studies, including gene expression profiles of primary human and rat hepatocytes treated with 231 potential toxicants. To efficiently mine these complex toxicogenomic data, ToxicoDB provides users with harmonized chemical annotations, time- and dose-dependent plots of compounds across datasets, as well as the toxicity-related pathway analysis. The data in ToxicoDB have been generated using our open-source R package, ToxicoGx (github.com/bhklab/ToxicoGx). Altogether, ToxicoDB provides a streamlined process for mining highly organized, curated, and accessible toxicogenomic data that can be ultimately applied to preclinical toxicity studies and further our understanding of adverse outcomes.


Subject(s)
Databases, Genetic , Software , Toxicogenetics/methods , Acetaminophen/toxicity , Animals , Computer Graphics , DNA/biosynthesis , Data Mining , Gene Expression/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Nucleic Acid Synthesis Inhibitors/toxicity , Rats
20.
Gigascience ; 9(5)2020 05 01.
Article in English | MEDLINE | ID: mdl-32449777

ABSTRACT

BACKGROUND: Omics technologies have been widely applied in toxicology studies to investigate the effects of different substances on exposed biological systems. A classical toxicogenomic study consists in testing the effects of a compound at different dose levels and different time points. The main challenge consists in identifying the gene alteration patterns that are correlated to doses and time points. The majority of existing methods for toxicogenomics data analysis allow the study of the molecular alteration after the exposure (or treatment) at each time point individually. However, this kind of analysis cannot identify dynamic (time-dependent) events of dose responsiveness. RESULTS: We propose TinderMIX, an approach that simultaneously models the effects of time and dose on the transcriptome to investigate the course of molecular alterations exerted in response to the exposure. Starting from gene log fold-change, TinderMIX fits different integrated time and dose models to each gene, selects the optimal one, and computes its time and dose effect map; then a user-selected threshold is applied to identify the responsive area on each map and verify whether the gene shows a dynamic (time-dependent) and dose-dependent response; eventually, responsive genes are labelled according to the integrated time and dose point of departure. CONCLUSIONS: To showcase the TinderMIX method, we analysed 2 drugs from the Open TG-GATEs dataset, namely, cyclosporin A and thioacetamide. We first identified the dynamic dose-dependent mechanism of action of each drug and compared them. Our analysis highlights that different time- and dose-integrated point of departure recapitulates the toxicity potential of the compounds as well as their dynamic dose-dependent mechanism of action.


Subject(s)
Computational Biology/methods , Software , Toxicogenetics/methods , Algorithms , Dose-Response Relationship, Drug , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Pharmacogenomic Testing , Pharmacogenomic Variants
SELECTION OF CITATIONS
SEARCH DETAIL