1
|
Zhu H, Conley JM, Karavadhi S, LaVigne JE, Watts VJ, Sun H, Shen M, Hall MD, Ren H, Patnaik S. Discovery of novel and selective GPR17 antagonists as pharmacological tools for developing new therapeutic strategies in diabetes and obesity. Eur J Med Chem 2025; 295:117794. [PMID: 40460721 DOI: 10.1016/j.ejmech.2025.117794] [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] [Received: 03/24/2025] [Revised: 05/19/2025] [Accepted: 05/20/2025] [Indexed: 06/11/2025]
Abstract
G protein coupled receptors (GPCRs) are promising targets for diabetes and obesity therapy due to their roles in metabolism and excellent potential for pharmacological manipulation. We previously reported that Gpr17 ablation in the brain-gut axis leads to improved metabolic homeostasis, suggesting GPR17 antagonism could be developed for diabetes and obesity treatment. Here, we performed high throughput screening (HTS) and identified two new GPR17 antagonists (compound 978 and 527). Both compounds antagonized downstream Gαi/o, Gαq and β-arrestin signaling with high selectivity for GPR17, but not the closely related purinergic and cysteinyl leukotriene receptors. The molecular mechanisms of antagonism were revealed through Schild analysis, structure-activity relationship (SAR) studies and homology modeling. Compound 978, a competitive antagonist against the surrogate small molecule agonist MDL29,951 (MDL), and its analog (793) attenuated GPR17 signaling and promoted glucagon-like peptide-1 (GLP-1) secretion in enteroendocrine cells. In summary, we identified selective GPR17 antagonists through HTS, which represent promising pharmacological tools for developing new therapeutic strategies in diabetes and obesity.
Collapse
Affiliation(s)
- Hu Zhu
- Early Translational Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Jason M Conley
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, USA
| | - Surendra Karavadhi
- Early Translational Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Justin E LaVigne
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
| | - Val J Watts
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
| | - Hongmao Sun
- Early Translational Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Min Shen
- Early Translational Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Matthew D Hall
- Early Translational Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Hongxia Ren
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, USA; Center for Diabetes and Metabolic Diseases, USA; Stark Neurosciences Research Institute, USA; Department of Anatomy, Cell Biology & Physiology, USA; Department of Biochemistry & Molecular Biology, USA; Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Samarjit Patnaik
- Early Translational Branch, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA.
| |
Collapse
|
2
|
Clayton J, Shi L, Robertson MJ, Skiniotis G, Michaelides M, Stavitskaya L, Shen J. A putative binding model of nitazene derivatives at the μ-opioid receptor. Neuropharmacology 2025; 273:110437. [PMID: 40185362 DOI: 10.1016/j.neuropharm.2025.110437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/09/2025] [Accepted: 03/28/2025] [Indexed: 04/07/2025]
Abstract
Nitazenes are a class of novel synthetic opioids with exceptionally high potency. Currently, an experimental structure of μOR-opioid receptor (μOR) in complex with a nitazene is lacking. Here we used a suite of computational tools, including consensus docking, conventional molecular dynamics (MD) and metadynamics simulations, to investigate the μOR binding modes of nitro-containing meto-, eto-, proto-, buto-, and isotonitazenes and nitro-less analogs, metodes-, etodes-, and protodesnitazenes. Docking generated three binding modes, whereby the nitro-substituted or unsubstituted benzimidazole group extends into SP1 (subpocket 1 between transmembrane helix or TM 2 and 3), SP2 (subpocket 2 between TM1, TM2, and TM7) or SP3 (subpocket 3 between TM5 and TM6). Simulations suggest that etonitazene and likely also other nitazenes favor the SP2-binding mode. Comparison to the experimental structures of μOR in complex with BU72, fentanyl, and mitragynine pseudoindoxyl (MP) allows us to propose a putative model for μOR-ligand recognition in which ligand can access hydrophobic SP1 or hydrophilic SP2, mediated by the conformational change of Gln1242.60. Interestingly, in addition to water-mediated hydrogen bonds, the nitro group in nitazenes forms a π-hole interaction with the conserved Tyr751.39. Our computational analysis provides new insights into the mechanism of μOR-opioid recognition, paving the way for investigations of the structure-activity relationships of nitazenes.
Collapse
Affiliation(s)
- Joseph Clayton
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, 20993, USA; Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, NIH/DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Michael J Robertson
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Georgios Skiniotis
- Department of Molecular and Cellular Physiology, Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Michael Michaelides
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Section, Neuroimaging Research Branch, National Institute on Drug Abuse, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Lidiya Stavitskaya
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, 20993, USA.
| | - Jana Shen
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA.
| |
Collapse
|
3
|
Antoniou M, Papavasileiou KD, Tsoumanis A, Melagraki G, Afantitis A. Predicting peroxisome proliferator-activated receptor gamma potency of small molecules: a synergistic consensus model and deep learning binding affinity approach powered by Enalos Cloud Platform. Mol Divers 2025:10.1007/s11030-025-11230-6. [PMID: 40515966 DOI: 10.1007/s11030-025-11230-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/16/2025] [Indexed: 06/16/2025]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) antagonists play a critical role in regulating glucose and lipid metabolism, making them promising candidates for antidiabetic therapies. To support the ongoing search of such compounds, this study introduces two advanced in silico models for predicting the binding affinity and biological activity of small molecules targeting PPARγ. A neural network was developed to classify compounds as strong or weak binders based on molecular docking scores. Additionally, a consensus model combining Random Forest, Support Vector Machine, and k-Nearest Neighbours algorithms was implemented to predict the antagonistic activity of small molecules. Both models were rigorously validated according to the Organisation for Economic Co-operation and Development (OECD) guidelines, to ensure generalisability and sufficient efficiency in detecting the minority class (active antagonists). Mechanistic insights into how key molecular descriptors influence PPARγ activity were discussed in a posteriori interpretation. A case study involving 34 prioritised per- and polyfluoroalkyl substances (PFAS) were screened with the developed workflows to demonstrate their practical application. The models, integrated into user-friendly web applications via the Enalos Cloud Platform, enable accessible and efficient virtual screening, supporting the discovery of PPARγ modulators.
Collapse
Affiliation(s)
- Maria Antoniou
- Department of ChemoInformatics, NovaMechanics Ltd, 1070, Nicosia, Cyprus
- Computation-Based Science and Technology Research Centre, The Cyprus Institute, 2121, Nicosia, Cyprus
- Entelos Institute, 6059, Larnaca, State, Cyprus
| | - Konstantinos D Papavasileiou
- Entelos Institute, 6059, Larnaca, State, Cyprus
- Department of ChemoInformatics, NovaMechanics MIKE, 18545, Piraeus, Greece
| | - Antreas Tsoumanis
- Department of ChemoInformatics, NovaMechanics Ltd, 1070, Nicosia, Cyprus
- Entelos Institute, 6059, Larnaca, State, Cyprus
- Department of ChemoInformatics, NovaMechanics MIKE, 18545, Piraeus, Greece
| | - Georgia Melagraki
- Division of Physical Sciences Applications, Hellenic Military Academy, 16672, Vari, Greece
| | - Antreas Afantitis
- Department of ChemoInformatics, NovaMechanics Ltd, 1070, Nicosia, Cyprus.
- Entelos Institute, 6059, Larnaca, State, Cyprus.
- Department of ChemoInformatics, NovaMechanics MIKE, 18545, Piraeus, Greece.
| |
Collapse
|
4
|
Parham F, Eccles KM, Rider CV, Sakamuru S, Xia M, Huang R, Tice RR, Dinse GE, DeVito MJ. Lessons learned from evaluating defined chemical mixtures in a high-throughput estrogen receptor assay system. Toxicol Sci 2025; 205:191-204. [PMID: 39972627 PMCID: PMC12038247 DOI: 10.1093/toxsci/kfaf020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025] Open
Abstract
In this article, we provide a proof of concept evaluating the utility of the U.S. Tox21 high-throughput screening approach to assess the hazard of chemical mixtures using 2 estrogen receptor (ER) assays. A subset of chemicals identified in Phase I of the Tox21 program as active in the ER agonist assay were used to design mixtures for testing in Phase II. Individual chemicals and mixtures were evaluated in 2 cell-based ER alpha (ERα) activation assays: One incorporating a transfected ligand-binding domain in an ERα β-lactamase reporter cell line (ER-bla) and the full-length endogenous receptor in the MCF7 cell line with a luciferase reporter gene (ER-luc). Concentration-response data from individual chemicals were used to predict the joint effect based on mixtures modeling methods and were compared with observed mixtures data to assess model fit. The models tended to overpredict mixture responses in the ER-bla assay, whereas predictions were closer to observed responses in the ER-luc assay, indicating that a full-length endogenous ER is a preferred model for high-throughput mixture analysis. Lessons learned from this research include the importance of analyzing the individual chemicals used for predictions and the mixtures in the same experimental paradigm to minimize variation, developing methods for imputing missing values from incomplete concentration-response curves, and establishing criteria to determine when inactive chemicals should be omitted from mixture predictions.
Collapse
Affiliation(s)
- Fred Parham
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Kristin M Eccles
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Cynthia V Rider
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Srilatha Sakamuru
- National Center for Advancing Translational Sciences, Bethesda, MD 20850, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences, Bethesda, MD 20850, USA
| | - Ruili Huang
- National Center for Advancing Translational Sciences, Bethesda, MD 20850, USA
| | | | | | - Michael J DeVito
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27709, USA
| |
Collapse
|
5
|
Clayton J, Shi L, Robertson MJ, Skiniotis G, Michaelides M, Stavitskaya L, Shen J. A Putative Binding Model of Nitazene Derivatives at the μ-Opioid Receptor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.03.616560. [PMID: 39990498 PMCID: PMC11844390 DOI: 10.1101/2024.10.03.616560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Nitazenes are a class of novel synthetic opioids with exceptionally high potency. Currently, an experimental structure of μOR-opioid receptor (μOR) in complex with a nitazene is lacking. Here we used a suite of computational tools, including consensus docking, conventional molecular dynamics (MD) and metadynamics simulations, to investigate the μOR binding modes of nitro-containing meto-, eto-, proto-, buto-, and isotonitazenes and nitro-less analogs, metodes-, etodes-, and protodesnitazenes. Docking generated three binding modes, whereby the nitro-substituted or unsubstituted benzimidazole group extends into SP1 (subpocket 1 between transmembrane helix or TM 2 and 3), SP2 (subpocket 2 between TM1, TM2, and TM7) or SP3 (subpocket 3 between TM5 and TM6). Simulations suggest that etonitazene and likely also other nitazenes favor the SP2-binding mode. Comparison to the experimental structures of μOR in complex with BU72, fentanyl, and mitragynine pseudoindoxyl (MP) allows us to propose a putative model for μOR-ligand recognition in which ligand can access hydrophobic SP1 or hydrophilic SP2, mediated by the conformational change of Gln1242.60. Interestingly, in addition to water-mediated hydrogen bonds, the nitro group in nitazenes forms a π-hole interaction with the conserved Tyr751.39. Our computational analysis provides new insights into the mechanism of μOR-opioid recognition, paving the way for investigations of the structure-activity relationships of nitazenes.
Collapse
Affiliation(s)
- Joseph Clayton
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, NIH/DHHS, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Michael J Robertson
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Georgios Skiniotis
- Department of Molecular and Cellular Physiology, Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael Michaelides
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Section, Neuroimaging Research Branch, National Institute on Drug Abuse, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Lidiya Stavitskaya
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Jana Shen
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| |
Collapse
|
6
|
Richard AM, Tao D, LeClair CA, Leister W, Tretyakov KV, White E, Lewis KC, Sefler A, Shinn P, Collins BJ, Nguyen DT, Ye L, Zhao T, Xu T, Williams AJ, Waidyanatha S, Thomas RS, Tice R, Simeonov A, Huang R. Analytical Quality Evaluation of the Tox21 Compound Library. Chem Res Toxicol 2025; 38:15-41. [PMID: 39829241 PMCID: PMC11752516 DOI: 10.1021/acs.chemrestox.4c00330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025]
Abstract
The analytical quality of compounds subjected to high-throughput screening (HTS) impacts accurate interpretation of assay results, with poor quality samples potentially leading to false negatives or positives. The Tox21 "10K" library consists of over 8900 unique compounds, spanning a diverse landscape of environmental and pharmaceutical chemicals, posing opportunities and challenges for analytical quality control (QC) determinations. Tox21 sample plates stored in DMSO at ambient conditions for 0 (T0) and/or 4 months (T4), totaling more than 13K unique sample identifiers (Tox21 IDs), were subjected to various analyses, including liquid and gas chromatography mass spectrometry (LC-MS, GC-MS) and nuclear magnetic resonance (NMR). Results for each sample at T0 or T4 underwent expert review and, where possible, a QC grade conveying purity, identity, and concentration was assigned. Herein, we relate details of the methods applied and report on the original (v0) Tox21 ID level results. Thirteen QC grades were condensed to 5 quality scores to aid global analysis, resulting in reinterpretation and improvement of >700 sample grades. Of the 92% T0 samples successfully graded, 76% exceeded 90% purity. For 76% of samples that were also tested at T4, 89% showed no evidence of sample loss or degradation. Prioritized quality bins were used to summarize thousands of replicate sample-level QC results to a compound-level QC score to support structure-based analyses. ToxPrint chemotype analysis identified structural features enriched in unstable compounds, as well as in high and low quality T0 subsets. Predicted vapor pressure was weakly correlated with low-concentration QC indicators, reflecting likely entanglement with method amenability and quality issues. Finally, an ongoing EPA effort to re-evaluate the original QC spectra is generating insights that will further modify QC grades. Tox21 QC spectra and results will be made available in a new public QC browser, facilitating further evaluation to support HTS interpretation and modeling applications.
Collapse
Affiliation(s)
- Ann M. Richard
- Center for
Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina 27711, United States
| | - Dingyin Tao
- Division
of Preclinical Innovation, National Center for Advancing Translational
Sciences (NCATS), National Institutes of
Health (NIH), Rockville, Maryland 20850, United States
| | - Christopher A. LeClair
- Division
of Preclinical Innovation, National Center for Advancing Translational
Sciences (NCATS), National Institutes of
Health (NIH), Rockville, Maryland 20850, United States
| | - William Leister
- Division
of Preclinical Innovation, National Center for Advancing Translational
Sciences (NCATS), National Institutes of
Health (NIH), Rockville, Maryland 20850, United States
| | - Kirill V. Tretyakov
- Biomolecular
Measurement Division, National Institute
of Standards and Technology (NIST), Gaithersburg, Maryland 20899, United States
| | - Edward White
- Biomolecular
Measurement Division, National Institute
of Standards and Technology (NIST), Gaithersburg, Maryland 20899, United States
| | - Ken C. Lewis
- OpAns, Durham, North Carolina 27713, United States
| | | | - Paul Shinn
- Division
of Preclinical Innovation, National Center for Advancing Translational
Sciences (NCATS), National Institutes of
Health (NIH), Rockville, Maryland 20850, United States
| | - Bradley J. Collins
- Division
of Translational Toxicology (DTT), National
Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, United States
| | - Dac-Trung Nguyen
- Division
of Preclinical Innovation, National Center for Advancing Translational
Sciences (NCATS), National Institutes of
Health (NIH), Rockville, Maryland 20850, United States
| | - Lin Ye
- Division
of Preclinical Innovation, National Center for Advancing Translational
Sciences (NCATS), National Institutes of
Health (NIH), Rockville, Maryland 20850, United States
| | - Tongan Zhao
- Division
of Preclinical Innovation, National Center for Advancing Translational
Sciences (NCATS), National Institutes of
Health (NIH), Rockville, Maryland 20850, United States
| | - Tuan Xu
- Division
of Preclinical Innovation, National Center for Advancing Translational
Sciences (NCATS), National Institutes of
Health (NIH), Rockville, Maryland 20850, United States
| | - Antony J. Williams
- Center for
Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina 27711, United States
| | - Suramya Waidyanatha
- Division
of Translational Toxicology (DTT), National
Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, United States
| | - Russell S. Thomas
- Center for
Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina 27711, United States
| | - Raymond Tice
- Division
of Translational Toxicology (DTT), National
Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, United States
| | - Anton Simeonov
- Division
of Preclinical Innovation, National Center for Advancing Translational
Sciences (NCATS), National Institutes of
Health (NIH), Rockville, Maryland 20850, United States
| | - Ruili Huang
- Division
of Preclinical Innovation, National Center for Advancing Translational
Sciences (NCATS), National Institutes of
Health (NIH), Rockville, Maryland 20850, United States
| |
Collapse
|
7
|
Lynch C, Margolis R, Niebler J, Travers J, Sakamuru S, Zhao T, Klumpp-Thomas C, Huang R, Xia M. Identification of human pregnane X receptor antagonists utilizing a high-throughput screening platform. Front Pharmacol 2024; 15:1448744. [PMID: 39508053 PMCID: PMC11537999 DOI: 10.3389/fphar.2024.1448744] [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: 06/13/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Pregnane X receptor (PXR) is a xenobiotic-sensing nuclear receptor with a well-established role in regulating drug metabolism and clearance. Recent studies have shown that PXR is involved in cell proliferation, apoptosis, immune response, and energy homeostasis. It is important to identify compounds that may modulate PXR activity to prevent drug-drug interactions, distinguish chemicals which could potentially generate toxicity, and identify compounds for further development towards therapeutic usage. In this study, we have screened the National Center for Advancing Translational Sciences (NCATS) Pharmacologically Active Chemical Toolbox (NPACT) library, which consists of 5,099 unique pharmacologically active synthetic and naturally derived small molecules to identify PXR antagonists. Ninety-four compounds were identified as potential PXR antagonists through a primary screen and 66 were confirmed in a confirmation study. Of these compounds, twenty potential PXR antagonists, including gamma-secretase modulator 2 (GSM2) and fusidic acid, were selected for further study based on their efficacy, potency, and novelty. Their PXR inhibition abilities were assessed by examining their effects on cytrochrome P450 (CYP) 3A4 mRNA expression using metabolically competent HepaRG cells. Additionally, a pharmacological inhibition assay using various concentrations of rifampicin as a stimulator was performed in HepG2-CYP3A4-hPXR cells to confirm the activity of the 20 selected compounds against PXR. Finally, HepaRG cells were used to confirm PXR antagonism by verification of a concentration-dependent decrease of CYP3A4 when co-treated with the known PXR agonist, rifampicin. Additionally, the potent actives were further investigated using molecular docking to find the potential interactions of the novel ligands with the active sites of hPXR. To our knowledge from the current study, GSM2 and fusidic acid have been identified as novel PXR antagonists, which provides useful information for further investigation regarding possible drug-drug interactions, as well as the detection of potential therapeutic effects or other toxic consequences.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
8
|
Zhao Y, Fent K. Endogenous hormones matters in evaluation of endocrine disruptive effects mediated by nuclear receptors. ECO-ENVIRONMENT & HEALTH 2024; 3:257-259. [PMID: 39220231 PMCID: PMC11364016 DOI: 10.1016/j.eehl.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/07/2024] [Accepted: 04/21/2024] [Indexed: 09/04/2024]
Abstract
Image 1.
Collapse
Affiliation(s)
- Yanbin Zhao
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Karl Fent
- ETH Zürich, Institute of Biogeochemistry and Pollution Dynamics, Department of Environmental Systems Science, CH-8092 Zürich, Switzerland
| |
Collapse
|
9
|
Yang S, Zhang L, Khan K, Travers J, Huang R, Jovanovic VM, Veeramachaneni R, Sakamuru S, Tristan CA, Davis EE, Klumpp-Thomas C, Witt KL, Simeonov A, Shaw ND, Xia M. Identification of Environmental Compounds That May Trigger Early Female Puberty by Activating Human GnRHR and KISS1R. Endocrinology 2024; 165:bqae103. [PMID: 39254333 PMCID: PMC11384912 DOI: 10.1210/endocr/bqae103] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Indexed: 09/11/2024]
Abstract
There has been an alarming trend toward earlier puberty in girls, suggesting the influence of an environmental factor(s). As the reactivation of the reproductive axis during puberty is thought to be mediated by the hypothalamic neuropeptides kisspeptin and gonadotropin-releasing hormone (GnRH), we asked whether an environmental compound might activate the kisspeptin (KISS1R) or GnRH receptor (GnRHR). We used GnRHR or KISS1R-expressing HEK293 cells to screen the Tox21 10K compound library, a compendium of pharmaceuticals and environmental compounds, for GnRHR and KISS1R activation. Agonists were identified using Ca2+ flux and phosphorylated extracellularly regulated kinase (p-ERK) detection assays. Follow-up studies included measurement of genes known to be upregulated upon receptor activation using relevant murine or human cell lines and molecular docking simulation. Musk ambrette was identified as a KISS1R agonist, and treatment with musk ambrette led to increased expression of Gnrh1 in murine and human hypothalamic cells and expansion of GnRH neuronal area in developing zebrafish larvae. Molecular docking demonstrated that musk ambrette interacts with the His309, Gln122, and Gln123 residues of the KISS1R. A group of cholinergic agonists with structures similar to methacholine was identified as GnRHR agonists. When applied to murine gonadotrope cells, these agonists upregulated Fos, Jun, and/or Egr1. Molecular docking revealed a potential interaction between GnRHR and 5 agonists, with Asn305 constituting the most conservative GnRHR binding site. In summary, using a Tox21 10K compound library screen combined with cellular, molecular, and structural biology techniques, we have identified novel environmental agents that may activate the human KISS1R or GnRHR.
Collapse
Affiliation(s)
- Shu Yang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Li Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kamal Khan
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago, IL 60611, USA
| | - Jameson Travers
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ruili Huang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vukasin M Jovanovic
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rithvik Veeramachaneni
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Srilatha Sakamuru
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Carlos A Tristan
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Erica E Davis
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago, IL 60611, USA
- Department of Pediatrics, Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Carleen Klumpp-Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kristine L Witt
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Anton Simeonov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Natalie D Shaw
- Pediatric Neuroendocrinology Group, Clinical Research Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709 USA
| | - Menghang Xia
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
10
|
Tong ZB, Huang R, Braisted J, Chu PH, Simeonov A, Gerhold DL. 3D-Suspension culture platform for high throughput screening of neurotoxic chemicals using LUHMES dopaminergic neurons. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100143. [PMID: 38280460 PMCID: PMC11056300 DOI: 10.1016/j.slasd.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/21/2023] [Accepted: 01/22/2024] [Indexed: 01/29/2024]
Abstract
Three-dimensional (3D) cell culture in vitro promises to improve representation of neuron physiology in vivo. This inspired development of a 3D culture platform for LUHMES (Lund Human Mesencephalic) dopaminergic neurons for high-throughput screening (HTS) of chemicals for neurotoxicity. Three culture platforms, adhesion (2D-monolayer), 3D-suspension, and 3D-shaken, were compared to monitor mRNA expression of seven neuronal marker genes, DCX, DRD2, ENO2, NEUROD4, SYN1, TH, and TUBB3. These seven marker genes reached similar maxima in all three formats, with the two 3D platforms showing similar kinetics, whereas several markers peaked earlier in 2D adhesion compared to both 3D culture platforms. The differentiated LUHMES (dLUHMES) neurons treated with ziram, methylmercury or thiram dynamically increased expression of metallothionein biomarker genes MT1G, MT1E and MT2A at 6 h. These gene expression increases were generally more dynamic in 2D adhesion cultures than in 3D cultures, but were generally comparable between 3D-suspension and 3D-u plate (low binding) platforms. Finally, we adapted 3D-suspension culture of dLUHMES and neural stem cells to 1536 well plates with a HTS cytotoxicity assay. This HTS assay revealed that cytotoxicity IC50 values were not significantly different between adhesion and 3D-suspension platforms for 31 of 34 (91%) neurotoxicants tested, whereas IC50 values were significantly different for at least two toxicants. In summary, the 3D-suspension culture platform for LUHMES dopaminergic neurons supported full differentiation and reproducible assay results, enabling quantitative HTS (qHTS) for cytotoxicity in 1536 well format with a Robust Z' score of 0.68.
Collapse
Affiliation(s)
- Zhi-Bin Tong
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States
| | - Ruili Huang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States
| | - John Braisted
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States
| | - Pei-Hsuan Chu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States
| | - Anton Simeonov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States
| | - David L Gerhold
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States.
| |
Collapse
|
11
|
Ardenkjær-Skinnerup J, Saar D, Petersen PSS, Pedersen M, Svingen T, Kragelund BB, Hadrup N, Ravn-Haren G, Emanuelli B, Brown KA, Vogel U. PPARγ antagonists induce aromatase transcription in adipose tissue cultures. Biochem Pharmacol 2024; 222:116095. [PMID: 38423186 DOI: 10.1016/j.bcp.2024.116095] [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] [Received: 10/31/2023] [Revised: 01/11/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024]
Abstract
Aromatase is the rate-limiting enzyme in the biosynthesis of estrogens and a key risk factor for hormone receptor-positive breast cancer. In postmenopausal women, estrogens synthesized in adipose tissue promotes the growth of estrogen receptor positive breast cancers. Activation of peroxisome proliferator-activated receptor gamma (PPARγ) in adipose stromal cells (ASCs) leads to decreased expression of aromatase and differentiation of ASCs into adipocytes. Environmental chemicals can act as antagonists of PPARγ and disrupt its function. This study aimed to test the hypothesis that PPARγ antagonists can promote breast cancer by stimulating aromatase expression in human adipose tissue. Primary cells and explants from human adipose tissue as well as A41hWAT, C3H10T1/2, and H295R cell lines were used to investigate PPARγ antagonist-stimulated effects on adipogenesis, aromatase expression, and estrogen biosynthesis. Selected antagonists inhibited adipocyte differentiation, preventing the adipogenesis-associated downregulation of aromatase. NMR spectroscopy confirmed direct interaction between the potent antagonist DEHPA and PPARγ, inhibiting agonist binding. Short-term exposure of ASCs to PPARγ antagonists upregulated aromatase only in differentiated cells, and a similar effect could be observed in human breast adipose tissue explants. Overexpression of PPARG with or without agonist treatment reduced aromatase expression in ASCs. The data suggest that environmental PPARγ antagonists regulate aromatase expression in adipose tissue through two mechanisms. The first is indirect and involves inhibition of adipogenesis, while the second occurs more acutely.
Collapse
Affiliation(s)
- Jacob Ardenkjær-Skinnerup
- The National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark; The National Research Centre for the Working Environment, Copenhagen Ø, Denmark
| | - Daniel Saar
- REPIN and Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen N, Denmark
| | - Patricia S S Petersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen N, Denmark
| | - Mikael Pedersen
- The National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Terje Svingen
- The National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Birthe B Kragelund
- REPIN and Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen N, Denmark
| | - Niels Hadrup
- The National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark; The National Research Centre for the Working Environment, Copenhagen Ø, Denmark
| | - Gitte Ravn-Haren
- The National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Brice Emanuelli
- The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen N, Denmark
| | - Kristy A Brown
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA; Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA.
| | - Ulla Vogel
- The National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark; The National Research Centre for the Working Environment, Copenhagen Ø, Denmark.
| |
Collapse
|
12
|
Ardenkjær-Skinnerup J, Nissen ACVE, Nikolov NG, Hadrup N, Ravn-Haren G, Wedebye EB, Vogel U. Orthogonal assay and QSAR modelling of Tox21 PPARγ antagonist in vitro high-throughput screening assay. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 105:104347. [PMID: 38143042 DOI: 10.1016/j.etap.2023.104347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
Disruption of signalling mediated by the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) is associated with risk of cancer, metabolic diseases, and endocrine disruption. The purpose of this study was to identify environmental chemicals acting as PPARγ antagonists. Data from the Tox21 PPARγ antagonism assay were replicated using a reporter system in HEK293 cells. Two quantitative structure-activity relationship (QSAR) models were developed, and five REACH-registered substances predicted positive were tested in vitro. Reporter assay results were consistent with Tox21 data since all conflicting results could be explained by assay interference. QSAR models showed good predictive performance, and follow-up experiments revealed two PPARγ antagonists out of three non-interfering chemicals. In conclusion, the developed QSAR models and follow-up experiments are important steps in the discovery of potential endocrine- and metabolism-disrupting chemicals.
Collapse
Affiliation(s)
- Jacob Ardenkjær-Skinnerup
- The National Food Institute, Technical University of Denmark, Kemitorvet 202, 2800 Kongens Lyngby, Denmark; The National Research Centre for the Working Environment, Lersø Parkallé 105, 2100 Copenhagen Ø, Denmark
| | | | - Nikolai Georgiev Nikolov
- The National Food Institute, Technical University of Denmark, Kemitorvet 202, 2800 Kongens Lyngby, Denmark
| | - Niels Hadrup
- The National Food Institute, Technical University of Denmark, Kemitorvet 202, 2800 Kongens Lyngby, Denmark; The National Research Centre for the Working Environment, Lersø Parkallé 105, 2100 Copenhagen Ø, Denmark
| | - Gitte Ravn-Haren
- The National Food Institute, Technical University of Denmark, Kemitorvet 202, 2800 Kongens Lyngby, Denmark
| | - Eva Bay Wedebye
- The National Food Institute, Technical University of Denmark, Kemitorvet 202, 2800 Kongens Lyngby, Denmark
| | - Ulla Vogel
- The National Food Institute, Technical University of Denmark, Kemitorvet 202, 2800 Kongens Lyngby, Denmark; The National Research Centre for the Working Environment, Lersø Parkallé 105, 2100 Copenhagen Ø, Denmark.
| |
Collapse
|
13
|
Escher BI, Binnington MJ, König M, Lei YD, Wania F. Mixture effect assessment applying in vitro bioassays to in-tissue silicone extracts of traditional foods prepared from beluga whale blubber. ENVIRONMENTAL SCIENCE. PROCESSES & IMPACTS 2023; 25:1759-1770. [PMID: 37254953 DOI: 10.1039/d3em00076a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
We complement an earlier study on the nutrient and environmental contaminant levels in Arctic beluga whale traditional foods by mixture effect assessment using in vitro bioassays. Mixtures were extracted by in-tissue sampling of raw blubber and several traditional food preparations including Muktuk and Uqsuq using silicone (polydimethylsiloxane, PDMS) as sampler. PDMS extracts persistent and degradable neutral organic chemicals of a wide range of hydrophobicity with defined lipid-PDMS partition ratios. The solvent extracts of PDMS were dosed in various reporter gene assays based on human cell lines. Cytotoxicity was consistent across all cell lines and was a good indicator of overall chemical burden. No hormone-like effects on the estrogen receptor, the progesterone receptor and the glucocorticoid receptor were observed but a few samples activated the androgen receptor, albeit with low potency. The peroxisome-proliferator activated receptor (PPARγ) was the most sensitive endpoint followed by activation of oxidative stress response and activation of the arylhydrocarbon (AhR) receptor. The detected pollutants only explained a small fraction of the experimental mixture effects, indicating additional bioactive pollutants. The effect levels of the extracted mixtures were higher than those observed in blubber extracts of dugongs living off the shore of Australia. Roasting over an open fire or food preparation near a smokehouse led to increased PAH levels that were reflected in increased oxidative stress response and activation of the AhR. So far in vitro assays have only been used to quantify persistent dioxin-like chemicals in food and feed but this pilot study demonstrates a much broader potential for food safety evaluations complementing chemical analytical monitoring.
Collapse
Affiliation(s)
- Beate I Escher
- Department of Cell Toxicology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany.
- Environmental Toxicology, Department of Geosciences, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Matthew J Binnington
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, Ontario, M1C 1A4, Canada.
| | - Maria König
- Department of Cell Toxicology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany.
| | - Ying D Lei
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, Ontario, M1C 1A4, Canada.
| | - Frank Wania
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, Ontario, M1C 1A4, Canada.
| |
Collapse
|
14
|
Safari M, Scotto L, Litman T, Petrukhin LA, Zhu H, Shen M, Robey RW, Hall MD, Fojo T, Bates SE. Novel Therapeutic Strategies Exploiting the Unique Properties of Neuroendocrine Neoplasms. Cancers (Basel) 2023; 15:4960. [PMID: 37894327 PMCID: PMC10605125 DOI: 10.3390/cancers15204960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Over the last few decades of treatment, the outcomes for at least some subsets of neuroendocrine neoplasms (NENs) have improved. However, the identification of new vulnerabilities for this heterogeneous group of cancers remains a priority. METHODS Using two libraries of compounds selected for potential repurposing, we identified the inhibitors of nicotinamide phosphoribosyltransferase (NAMPT) and histone deacetylases (HDAC) as the agents with the highest activity. We validated the hits in an expanded set of neuroendocrine cell lines and examined the mechanisms of action. RESULTS In Kelly, NH-6, and NCI-H82, which are two neuroblastoma and one small cell lung cancer cell lines, respectively, metabolic studies suggested that cell death following NAMPT inhibition is the result of a reduction in basal oxidative phosphorylation and energy production. NAMPT is the rate-limiting enzyme in the production of NAD+, and in the three cell lines, NAMPT inhibition led to a marked reduction in the ATP and NAD+ levels and the catalytic activity of the citric acid cycle. Moreover, comparative analysis of the mRNA expression in drug-sensitive and -insensitive cell lines found less dependency of the latter on oxidative phosphorylation for their energy requirement. Further, the analysis of HDAC and NAMPT inhibitors administered in combination found marked activity using low sub-lethal concentrations of both agents, suggesting a synergistic effect. CONCLUSION These data suggest NAMPT inhibitors alone or in combination with HDAC inhibitors could be particularly effective in the treatment of neuroendocrine neoplasms.
Collapse
Affiliation(s)
- Maryam Safari
- Division of Hematology/Oncology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Luigi Scotto
- Division of Hematology/Oncology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Thomas Litman
- Department of Immunology and Microbiology, University of Copenhagen, 1172 Copenhagen, Denmark
| | - Lubov A. Petrukhin
- Division of Hematology/Oncology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Hu Zhu
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, MD 20892, USA
| | - Min Shen
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, MD 20892, USA
| | - Robert W. Robey
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew D. Hall
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, MD 20892, USA
| | - Tito Fojo
- Division of Hematology/Oncology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
- James J. Peters Bronx Veterans Affairs Medical Center, Bronx, NY 10468, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Susan E. Bates
- Division of Hematology/Oncology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
- James J. Peters Bronx Veterans Affairs Medical Center, Bronx, NY 10468, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
15
|
Ruan T, Li P, Wang H, Li T, Jiang G. Identification and Prioritization of Environmental Organic Pollutants: From an Analytical and Toxicological Perspective. Chem Rev 2023; 123:10584-10640. [PMID: 37531601 DOI: 10.1021/acs.chemrev.3c00056] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
Exposure to environmental organic pollutants has triggered significant ecological impacts and adverse health outcomes, which have been received substantial and increasing attention. The contribution of unidentified chemical components is considered as the most significant knowledge gap in understanding the combined effects of pollutant mixtures. To address this issue, remarkable analytical breakthroughs have recently been made. In this review, the basic principles on recognition of environmental organic pollutants are overviewed. Complementary analytical methodologies (i.e., quantitative structure-activity relationship prediction, mass spectrometric nontarget screening, and effect-directed analysis) and experimental platforms are briefly described. The stages of technique development and/or essential parts of the analytical workflow for each of the methodologies are then reviewed. Finally, plausible technique paths and applications of the future nontarget screening methods, interdisciplinary techniques for achieving toxicant identification, and burgeoning strategies on risk assessment of chemical cocktails are discussed.
Collapse
Affiliation(s)
- Ting Ruan
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Pengyang Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haotian Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tingyu Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
16
|
Stanojević M, Sollner Dolenc M, Vračko M. Development of in silico classification models for binding affinity to the glucocorticoid receptor. CHEMOSPHERE 2023:139147. [PMID: 37301514 DOI: 10.1016/j.chemosphere.2023.139147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 04/12/2023] [Accepted: 06/04/2023] [Indexed: 06/12/2023]
Abstract
The endocrine disrupting properties of chemicals acting through the glucocorticoid receptor (GR) have attracted considerable interest. Since there are few data for most chemicals on their endocrine properties in silico approaches seem to be the most appropriate tool for screening and prioritizing chemicals for planning further experiments. In this work, we developed classification models for binding affinity to the glucocorticoid receptor using the counterpropagation artificial neural network method. We considered two series of 142 and 182 compounds and their binding affinity to the glucocorticoid receptor as agonists and antagonists, respectively. The compounds belong to different chemical classes. The compounds were represented by a set of descriptors calculated with the DRAGON program. The clustering structure of sets was studied with standard principal component method. A weak separation between binders and non-binders was found. Another classification model was developed using the counterpropagation artificial neural network method (CPANN). The final classification models developed were well balanced and showed a high level of accuracy, with 85.7% of GR agonist and 78.9% of GR antagonist correctly assigned in leave-one-out cross-validation.
Collapse
Affiliation(s)
- Mark Stanojević
- Bisafe Doo, V Kladeh 11c, 1000, Ljubljana, Slovenia; University of Ljubljana, Faculty of Pharmacy, Aškerčeva Cesta 7, 1000, Ljubljana, Slovenia.
| | - Marija Sollner Dolenc
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva Cesta 7, 1000, Ljubljana, Slovenia.
| | - Marjan Vračko
- National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia.
| |
Collapse
|
17
|
Stanojević M, Sollner Dolenc M, Vračko M. Predictive Models for Compound Binding to Androgen and Estrogen Receptors Based on Counter-Propagation Artificial Neural Networks. TOXICS 2023; 11:486. [PMID: 37368586 DOI: 10.3390/toxics11060486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023]
Abstract
Endocrine-disrupting chemicals (EDCs) are exogenous substances that interfere with the normal function of the human endocrine system. These chemicals can affect specific nuclear receptors, such as androgen receptors (ARs) or estrogen receptors (ER) α and β, which play a crucial role in regulating complex physiological processes in humans. It is now more crucial than ever to identify EDCs and reduce exposure to them. For screening and prioritizing chemicals for further experimentation, the use of artificial neural networks (ANN), which allow the modeling of complicated, nonlinear relationships, is most appropriate. We developed six models that predict the binding of a compound to ARs, ERα, or ERβ as agonists or antagonists, using counter-propagation artificial neural networks (CPANN). Models were trained on a dataset of structurally diverse compounds, and activity data were obtained from the CompTox Chemicals Dashboard. Leave-one-out (LOO) tests were performed to validate the models. The results showed that the models had excellent performance with prediction accuracy ranging from 94% to 100%. Therefore, the models can predict the binding affinity of an unknown compound to the selected nuclear receptor based solely on its chemical structure. As such, they represent important alternatives for the safety prioritization of chemicals.
Collapse
Affiliation(s)
- Mark Stanojević
- BiSafe d.o.o., 1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | | | - Marjan Vračko
- National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| |
Collapse
|
18
|
Yasgar A, Bougie D, Eastman RT, Huang R, Itkin M, Kouznetsova J, Lynch C, McKnight C, Miller M, Ngan DK, Peryea T, Shah P, Shinn P, Xia M, Xu X, Zakharov AV, Simeonov A. Quantitative Bioactivity Signatures of Dietary Supplements and Natural Products. ACS Pharmacol Transl Sci 2023; 6:683-701. [PMID: 37200814 PMCID: PMC10186358 DOI: 10.1021/acsptsci.2c00194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Indexed: 05/20/2023]
Abstract
Dietary supplements and natural products are often marketed as safe and effective alternatives to conventional drugs, but their safety and efficacy are not well regulated. To address the lack of scientific data in these areas, we assembled a collection of Dietary Supplements and Natural Products (DSNP), as well as Traditional Chinese Medicinal (TCM) plant extracts. These collections were then profiled in a series of in vitro high-throughput screening assays, including a liver cytochrome p450 enzyme panel, CAR/PXR signaling pathways, and P-glycoprotein (P-gp) transporter assay activities. This pipeline facilitated the interrogation of natural product-drug interaction (NaPDI) through prominent metabolizing pathways. In addition, we compared the activity profiles of the DSNP/TCM substances with those of an approved drug collection (the NCATS Pharmaceutical Collection or NPC). Many of the approved drugs have well-annotated mechanisms of action (MOAs), while the MOAs for most of the DSNP and TCM samples remain unknown. Based on the premise that compounds with similar activity profiles tend to share similar targets or MOA, we clustered the library activity profiles to identify overlap with the NPC to predict the MOAs of the DSNP/TCM substances. Our results suggest that many of these substances may have significant bioactivity and potential toxicity, and they provide a starting point for further research on their clinical relevance.
Collapse
Affiliation(s)
- Adam Yasgar
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Danielle Bougie
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Richard T Eastman
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Misha Itkin
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Jennifer Kouznetsova
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Caitlin Lynch
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Crystal McKnight
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Mitch Miller
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Deborah K Ngan
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Tyler Peryea
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Pranav Shah
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Paul Shinn
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Xin Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Alexey V Zakharov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| |
Collapse
|
19
|
Xu T, Kabir M, Sakamuru S, Shah P, Padilha E, Ngan DK, Xia M, Xu X, Simeonov A, Huang R. Predictive Models for Human Cytochrome P450 3A7 Selective Inhibitors and Substrates. J Chem Inf Model 2023; 63:846-855. [PMID: 36719788 PMCID: PMC10664139 DOI: 10.1021/acs.jcim.2c01516] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Inappropriate use of prescription drugs is potentially more harmful in fetuses/neonates than in adults. Cytochrome P450 (CYP) 3A subfamily undergoes developmental changes in expression, such as a transition from CYP3A7 to CYP3A4 shortly after birth, which provides a potential way to distinguish medication effects on fetuses/neonates and adults. The purpose of this study was to build first-in-class predictive models for both inhibitors and substrates of CYP3A7/CYP3A4 using chemical structure analysis. Three metrics were used to evaluate model performance: area under the receiver operating characteristic curve (AUC-ROC), balanced accuracy (BA), and Matthews correlation coefficient (MCC). The performance varied for each CYP3A7/CYP3A4 inhibitor/substrate model depending on the data set type, model type, rebalancing method, and specific feature set. For the active inhibitor/substrate data set, the optimal models achieved AUC-ROC values ranging from 0.77 ± 0.01 to 0.84 ± 0.01. For the selective inhibitor/substrate data set, the optimal models achieved AUC-ROC values ranging from 0.72 ± 0.02 to 0.79 ± 0.04. The predictive power of the optimal models was validated by compounds with known potencies as CYP3A7/CYP3A4 inhibitors or substrates. In addition, we identified structural features significant for CYP3A7/CYP3A4 selective or common inhibitors and substrates. In summary, the top performing models can be further applied as a tool to rapidly evaluate the safety and efficacy of new drugs separately for fetuses/neonates and adults. The significant structural features could guide the design of new therapeutic drugs as well as aid in the optimization of existing medicine for fetuses/neonates.
Collapse
Affiliation(s)
- Tuan Xu
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Md Kabir
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
- The Graduate School of Biomedical Sciences, Departments of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Srilatha Sakamuru
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Pranav Shah
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Elias Padilha
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Deborah K. Ngan
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Menghang Xia
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Xin Xu
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Anton Simeonov
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Ruili Huang
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| |
Collapse
|
20
|
Melnikov F, Anger LT, Hasselgren C. Toward Quantitative Models in Safety Assessment: A Case Study to Show Impact of Dose-Response Inference on hERG Inhibition Models. Int J Mol Sci 2022; 24:ijms24010635. [PMID: 36614078 PMCID: PMC9820331 DOI: 10.3390/ijms24010635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 12/31/2022] Open
Abstract
Due to challenges with historical data and the diversity of assay formats, in silico models for safety-related endpoints are often based on discretized data instead of the data on a natural continuous scale. Models for discretized endpoints have limitations in usage and interpretation that can impact compound design. Here, we present a consistent data inference approach, exemplified on two data sets of Ether-à-go-go-Related Gene (hERG) K+ inhibition data, for dose-response and screening experiments that are generally applicable for in vitro assays. hERG inhibition has been associated with severe cardiac effects and is one of the more prominent safety targets assessed in drug development, using a wide array of in vitro and in silico screening methods. In this study, the IC50 for hERG inhibition is estimated from diverse historical proprietary data. The IC50 derived from a two-point proprietary screening data set demonstrated high correlation (R = 0.98, MAE = 0.08) with IC50s derived from six-point dose-response curves. Similar IC50 estimation accuracy was obtained on a public thallium flux assay data set (R = 0.90, MAE = 0.2). The IC50 data were used to develop a robust quantitative model. The model's MAE (0.47) and R2 (0.46) were on par with literature statistics and approached assay reproducibility. Using a continuous model has high value for pharmaceutical projects, as it enables rank ordering of compounds and evaluation of compounds against project-specific inhibition thresholds. This data inference approach can be widely applicable to assays with quantitative readouts and has the potential to impact experimental design and improve model performance, interpretation, and acceptance across many standard safety endpoints.
Collapse
|
21
|
Confirmation of high-throughput screening data and novel mechanistic insights into FXR-xenobiotic interactions by orthogonal assays. Curr Res Toxicol 2022; 3:100092. [PMID: 36353521 PMCID: PMC9637864 DOI: 10.1016/j.crtox.2022.100092] [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: 05/25/2022] [Revised: 10/06/2022] [Accepted: 10/26/2022] [Indexed: 11/05/2022] Open
Abstract
Toxicology in the 21st Century (Tox21) is a federal collaboration employing a high-throughput robotic screening system to test 10,000 environmental chemicals. One of the primary goals of the program is prioritizing toxicity evaluations through in vitro high-throughput screening (HTS) assays for large numbers of chemicals already in commercial use for which little or no toxicity data is available. Within the Tox21 screening program, disruption in nuclear receptor (NR) signaling represents a particular area of interest. Given the role of NR's in modulating a wide range of biological processes, alterations of their activity can have profound biological impacts. Farnesoid X receptor (FXR) is a member of the nuclear receptor superfamily that has demonstrated importance in bile acid homeostasis, glucose metabolism, lipid homeostasis and hepatic regeneration. In this study, we re-evaluated 24 FXR agonists and antagonists identified through Tox21 using select orthogonal assays. In transient transactivation assays, 7/8 putative agonists and 4/4 putative inactive compounds were confirmed. Likewise, we confirmed 9/12 antagonists tested. Using a mammalian two hybrid approach we demonstrate that both FXR agonists and antagonists facilitate FXRα-coregulator interactions suggesting that differential coregulator recruitment may mediate activation/repression of FXRα mediated transcription. Additionally, we tested the ability of select FXR agonists and antagonists to facilitate hepatic transcription of FXR gene targets Shp and Bsep in a teleost (Medaka) model. Through application of in vitro cell-based assays, in silico modeling and in vivo gene expressions, we demonstrated the molecular complexity of FXR:ligand interactions and confirmed the ability of diverse ligands to modulate FXRα, facilitate differential coregulator recruitment and activate/repress receptor-mediated transcription. Overall, we suggest a multiplicative approach to assessment of nuclear receptor function may facilitate a greater understanding of the biological and mechanistic complexities of nuclear receptor activities and further our ability to interpret broad HTS outcomes.
Collapse
Key Words
- Bsep, bile salt export pump
- CDCA, chenodeoxycholic acid
- DMSO, dimethyl sulfoxide
- EPA, U.S. Environmental Protection Agency
- FXR, Farnesoid X receptor
- Farnesoid X receptor
- High-throughput screening
- M2H, mammalian two-hybrid
- Medaka
- RXR, retinoid X receptor
- Shp, small heterodimer partner
- Teleost models
- Tox21, Toxicology in the 21st Century
- ToxCast
- qHTS, quantitative high-throughput screening
Collapse
|
22
|
Yang R, Liu S, Yin N, Zhang Y, Faiola F. Tox21-Based Comparative Analyses for the Identification of Potential Toxic Effects of Environmental Pollutants. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:14668-14679. [PMID: 36178254 DOI: 10.1021/acs.est.2c04467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Chemical pollution has become a prominent environmental problem. In recent years, quantitative high-throughput screening (qHTS) assays have been developed for the fast assessment of chemicals' toxic effects. Toxicology in the 21st Century (Tox21) is a well-known and continuously developing qHTS project. Recent reports utilizing Tox21 data have mainly focused on setting up mathematical models for in vivo toxicity predictions, with less attention to intuitive qHTS data visualization. In this study, we attempted to reveal and summarize the toxic effects of environmental pollutants by analyzing and visualizing Tox21 qHTS data. Via PubMed text mining, toxicity/structure clustering, and manual classification, we detected a total of 158 chemicals of environmental concern (COECs) from the Tox21 library that we classified into 13 COEC groups based on structure and activity similarities. By visualizing these COEC groups' bioactivities, we demonstrated that COECs frequently displayed androgen and progesterone antagonistic effects, xenobiotic receptor agonistic roles, and mitochondrial toxicity. We also revealed many other potential targets of the 13 COEC groups, which were not well illustrated yet, and that current Tox21 assays may not correctly classify known teratogens. In conclusion, we provide a feasible method to intuitively understand qHTS data.
Collapse
Affiliation(s)
- Renjun Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuyu Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
- Wellcome Trust/CRUK Gurdon Institute, Department of Pathology, University of Cambridge, Cambridge CB2 1QN, U.K
| | - Nuoya Yin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Zhang
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Francesco Faiola
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
23
|
Shi Z, Xia M, Xiao S, Zhang Q. Identification of nonmonotonic concentration-responses in Tox21 high-throughput screening estrogen receptor assays. Toxicol Appl Pharmacol 2022; 452:116206. [PMID: 35988584 PMCID: PMC9452481 DOI: 10.1016/j.taap.2022.116206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 10/15/2022]
Abstract
Environmental endocrine-disrupting chemicals (EDCs) interfere with the metabolism and actions of endogenous hormones. It has been well documented in numerous in vivo and in vitro studies that EDCs can exhibit nonmonotonic dose response (NMDR) behaviors. Not conforming to the conventional linear or linear-no-threshold response paradigm, these NMDR relationships pose practical challenges to the risk assessment of EDCs. In the meantime, the endocrine signaling pathways and biological mechanisms underpinning NMDR remain incompletely understood. The US Tox21 program has conducted in vitro cell-based high-throughput screening assays for estrogen receptors (ER), androgen receptors, and other nuclear receptors, and screened the 10 K-compound library for potential endocrine activities. Using 15 concentrations across several orders of magnitude of concentration range and run in both agonist and antagonist modes, these Tox21 assay datasets contain valuable quantitative information that can be explored to evaluate the nonlinear effects of EDCs and may infer potential mechanisms. In this study we analyzed the concentration-response curves (CRCs) in all 8 Tox21 ERα and ERβ assays by developing clustering and classification algorithms customized to the datasets to identify various shapes of CRCs. After excluding NMDR curves likely caused by cytotoxicity, luciferase inhibition, or autofluorescence, hundreds of compounds were identified to exhibit Bell or U-shaped CRCs. Bell-shaped CRCs are about 7 times more frequent than U-shaped ones in the Tox21 ER assays. Many compounds exhibit NMDR in at least one assay, and some EDCs well-known for their NMDRs in the literature were also identified, suggesting their nonmonotonic effects may originate at cellular levels involving transcriptional ER signaling. The developed computational methods for NMDR identification in ER assays can be adapted and applied to other high-throughput bioassays.
Collapse
Affiliation(s)
- Zhenzhen Shi
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences, NIH, Bethesda, MD, USA
| | - Shuo Xiao
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, USA
| | - Qiang Zhang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| |
Collapse
|
24
|
Song WS, Koh DH, Kim EY. Orthogonal assay for validation of Tox21 PPARγ data and applicability to in silico prediction model. Toxicol In Vitro 2022; 84:105445. [PMID: 35863590 DOI: 10.1016/j.tiv.2022.105445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/01/2022] [Accepted: 07/13/2022] [Indexed: 11/28/2022]
Abstract
High-throughput screening data from the Tox21 database is used for prioritizing hazardous chemicals and building in silico-based toxicity prediction models. One of the Tox21 dataset, peroxisome proliferator-activated receptor-gamma (PPARγ), a nuclear receptor superfamily, identified various endpoints in HEK293 cells. PPARγ mediates various toxic effects when its receptors are activated or inhibited by ligands such as thiazolidinedione and GW9662. In this study, an orthogonal assay was constructed to verify the effectiveness of the Tox21 PPARγ data, and the effect of highly reliable data on in silico model construction was investigated. The orthogonal assay was a reporter gene assay based on the PPARγ ligand binding domain in CV-1 cells. Only 39% of agonists and 55% of antagonists had similar responses in CV-1 and HEK293 cells. Thus, the effectiveness of Tox21 data on PPARγ may vary depending on the cell line. However, in silico PLS-DA analysis with only high-reliability data (i.e., the same response in both cell lines), yielded more accurate prediction of the activity of potential chemical ligands, despite the small number of samples. Thus, obtaining reliable chemical screening data for PPARγ through orthogonal analysis, even for only limited chemicals, supports the construction of highly predictive in silico models with improved screening efficiency.
Collapse
Affiliation(s)
- Woo-Seon Song
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea
| | - Dong-Hee Koh
- Department of Biology, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea
| | - Eun-Young Kim
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea; Department of Biology, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea.
| |
Collapse
|
25
|
Sakamuru S, Huang R, Xia M. Use of Tox21 Screening Data to Evaluate the COVID-19 Drug Candidates for Their Potential Toxic Effects and Related Pathways. Front Pharmacol 2022; 13:935399. [PMID: 35910344 PMCID: PMC9333127 DOI: 10.3389/fphar.2022.935399] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/16/2022] [Indexed: 12/15/2022] Open
Abstract
Currently, various potential therapeutic agents for coronavirus disease-2019 (COVID-19), a global pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), are being investigated worldwide mainly through the drug repurposing approach. Several anti-viral, anti-bacterial, anti-malarial, and anti-inflammatory drugs were employed in randomized trials and observational studies for developing new therapeutics for COVID-19. Although an increasing number of repurposed drugs have shown anti-SARS-CoV-2 activities in vitro, so far only remdesivir has been approved by the US FDA to treat COVID-19, and several other drugs approved for Emergency Use Authorization, including sotrovimab, tocilizumab, baricitinib, paxlovid, molnupiravir, and other potential strategies to develop safe and effective therapeutics for SARS-CoV-2 infection are still underway. Many drugs employed as anti-viral may exert unwanted side effects (i.e., toxicity) via unknown mechanisms. To quickly assess these drugs for their potential toxicological effects and mechanisms, we used the Tox21 in vitro assay datasets generated from screening ∼10,000 compounds consisting of approved drugs and environmental chemicals against multiple cellular targets and pathways. Here we summarize the toxicological profiles of small molecule drugs that are currently under clinical trials for the treatment of COVID-19 based on their in vitro activities against various targets and cellular signaling pathways.
Collapse
|
26
|
Kabir M, Padilha EC, Shah P, Huang R, Sakamuru S, Gonzalez E, Ye L, Hu X, Henderson MJ, Xia M, Xu X. Identification of Selective CYP3A7 and CYP3A4 Substrates and Inhibitors Using a High-Throughput Screening Platform. Front Pharmacol 2022; 13:899536. [PMID: 35847040 PMCID: PMC9283723 DOI: 10.3389/fphar.2022.899536] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/27/2022] [Indexed: 11/26/2022] Open
Abstract
Cytochrome P450 (CYP) 3A7 is one of the major xenobiotic metabolizing enzymes in human embryonic, fetal, and newborn liver. CYP3A7 expression has also been observed in a subset of the adult population, including pregnant women, as well as in various cancer patients. The characterization of CYP3A7 is not as extensive as other CYPs, and health authorities have yet to provide guidance towards DDI assessment. To identify potential CYP3A7-specific molecules, we used a P450-Glo CYP3A7 enzyme assay to screen a library of ∼5,000 compounds, including FDA-approved drugs and drug-like molecules, and compared these screening data with that from a P450-Glo CYP3A4 assay. Additionally, a subset of 1,000 randomly selected compounds were tested in a metabolic stability assay. By combining the data from the qHTS P450-Glo and metabolic stability assays, we identified several chemical features important for CYP3A7 selectivity. Halometasone was chosen for further evaluation as a potential CYP3A7-selective inhibitor using molecular docking. From the metabolic stability assay, we identified twenty-two CYP3A7-selective substrates over CYP3A4 in supersome setting. Our data shows that CYP3A7 has ligand promiscuity, much like CYP3A4. Furthermore, we have established a large, high-quality dataset that can be used in predictive modeling for future drug metabolism and interaction studies.
Collapse
Affiliation(s)
- Md Kabir
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
- Department of Pharmacology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Elias C. Padilha
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
| | - Pranav Shah
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
| | - Ruili Huang
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
| | - Srilatha Sakamuru
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
| | - Eric Gonzalez
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
- Novartis Institutes for BioMedical Research, Cambridge, MA, United States
| | - Lin Ye
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
| | - Xin Hu
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
| | - Mark J. Henderson
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
| | - Menghang Xia
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
- *Correspondence: Menghang Xia, ; Xin Xu,
| | - Xin Xu
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
- *Correspondence: Menghang Xia, ; Xin Xu,
| |
Collapse
|
27
|
Gui B, Wang C, Xu X, Li C, Zhao Y, Su L. Identification of active or inactive agonists of tumor suppressor protein based on Tox21 library. Toxicology 2022; 474:153224. [PMID: 35659517 DOI: 10.1016/j.tox.2022.153224] [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] [Received: 02/15/2022] [Revised: 05/15/2022] [Accepted: 05/25/2022] [Indexed: 11/18/2022]
Abstract
Exposure of cells to xenobiotic human-made products can lead to genotoxicity and cause DNA damage. It is an urgent need to quickly identify the chemicals that cause DNA damage, and their toxicity should be predicted. In this study, recursive partitioning (RP), binary logistic regression, and one machine learning approach, namely, random forest (RF) classifier, were used to predict the active and inactive compounds of a total 5036 data based on the assay conducted by a β-lactamase reporter gene under control of the p53 response element (p53RE) from Tox21 library. Results show that the binary logistic regression model with a threshold of 0.5 has a high accuracy rate (83%) to distinguish active and inactive compounds. The RF classifier method has satisfactory results, with an accuracy rate (84.38%) approximately higher than that of binary logistic regression. The models established can identify compounds that induce DNA damage and activate p53, and provide a scientific basis for the risk assessment of organic chemicals in the environment.
Collapse
Affiliation(s)
- Bingxin Gui
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, 2555 Jingyue Street, Changchun 130117 Jilin, PR China
| | - Chen Wang
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, 2555 Jingyue Street, Changchun 130117 Jilin, PR China
| | - Xiaotian Xu
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, 2555 Jingyue Street, Changchun 130117 Jilin, PR China
| | - Chao Li
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, 2555 Jingyue Street, Changchun 130117 Jilin, PR China
| | - Yuanhui Zhao
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, 2555 Jingyue Street, Changchun 130117 Jilin, PR China
| | - Limin Su
- State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, School of Environment, Northeast Normal University, 2555 Jingyue Street, Changchun 130117 Jilin, PR China.
| |
Collapse
|
28
|
Insights into the Endocrine Disrupting Activity of Emerging Non-Phthalate Alternate Plasticizers against Thyroid Hormone Receptor: A Structural Perspective. TOXICS 2022; 10:toxics10050263. [PMID: 35622676 PMCID: PMC9145736 DOI: 10.3390/toxics10050263] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/02/2022] [Accepted: 05/17/2022] [Indexed: 11/16/2022]
Abstract
Many endocrine-disrupting chemicals (EDCs) have a ubiquitous presence in our environment due to anthropogenic activity. These EDCs can disrupt hormone signaling in the human and animal body systems including the very important hypothalamic-pituitary-thyroid (HPT) axis causing adverse health effects. Thyroxine (T4) and triiodothyronine (T3) are hormones of the HPT axis which are essential for regulation of metabolism, heart rate, body temperature, growth, development, etc. In this study, potential endocrine-disrupting activity of the most common phthalate plasticizer, DEHP, and emerging non-phthalate alternate plasticizers, DINCH, ATBC, and DEHA against thyroid hormone receptor (TRα) were characterized. The structural binding characterization of indicated ligands was performed against the TRα ligand binding site employing Schrodinger’s induced fit docking (IFD) approach. The molecular simulations of interactions of the ligands against the residues lining a TRα binding pocket, including bonding interactions, binding energy, docking score, and IFD score were analyzed. In addition, the structural binding characterization of TRα native ligand, T3, was also done for comparative analysis. The results revealed that all ligands were placed stably in the TRα ligand-binding pocket. The binding energy values were highest for DINCH, followed by ATBC, and were higher than the values estimated for TRα native ligand, T3, whereas the values for DEHA and DEHP were similar and comparable to that of T3. This study suggested that all the indicated plasticizers have the potential for thyroid hormone disruption with two alternate plasticizers, DINCH and ATBC, exhibiting higher potential for thyroid dysfunction compared to DEHA and DEHP.
Collapse
|
29
|
Pípal M, Novák J, Rafajová A, Smutná M, Hilscherová K. Teratogenicity of retinoids detected in surface waters in zebrafish embryos and its predictability by in vitro assays. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 246:106151. [PMID: 35390581 DOI: 10.1016/j.aquatox.2022.106151] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/08/2022] [Accepted: 03/25/2022] [Indexed: 06/14/2023]
Abstract
Retinoids are newly detected compounds in aquatic ecosystems associated with cyanobacterial water blooms. Their potential health risks are only scarcely described despite numerous detections of all-trans retinoic acid (ATRA) and its derivatives in the environment. Besides the known teratogen ATRA there is only little or no information about their potency and namely their effects in vivo. We characterize ATRA and 8 other retinoids reported to occur in the environment for their bioactivity and teratogenicity using four in vitro reporter gene assays and zebrafish (Danio rerio) embryotoxicity assay. Our results document the ability of these compounds to interfere with retinoid signalling and cause teratogenicity at environmentally relevant levels with EC50 values at nM (hundreds of ng/L) levels and teratogenic indexes ranging from 2.8 (9cis retinoic acid) to 15.8 (retinal). The relative potency of individual compounds for teratogenicity ranged from 0.059 (retinal) to 0.96 (5,6-epoxy ATRA) when compared to ATRA. An environmentally relevant mixture of retinoids was tested showing good predictability of teratogenicity from the in vitro activities and additive toxicity of the mixture. The high teratogenicity of the newly described compounds associated with cyanobacteria presents a concern for developmental stages due to high conservation of the retinoid signalling across vertebrates.
Collapse
Affiliation(s)
- Marek Pípal
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Kamenice, Brno 62500 , Czech Republic
| | - Jiří Novák
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Kamenice, Brno 62500 , Czech Republic
| | - Aneta Rafajová
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Kamenice, Brno 62500 , Czech Republic
| | - Marie Smutná
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Kamenice, Brno 62500 , Czech Republic
| | - Klára Hilscherová
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Kamenice, Brno 62500 , Czech Republic.
| |
Collapse
|
30
|
Ngan DK, Xu T, Xia M, Zheng W, Huang R. Repurposing drugs as COVID-19 therapies: a toxicity evaluation. Drug Discov Today 2022; 27:1983-1993. [PMID: 35395401 PMCID: PMC8983078 DOI: 10.1016/j.drudis.2022.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 02/17/2022] [Accepted: 04/01/2022] [Indexed: 12/24/2022]
Abstract
Drug repurposing is an appealing method to address the Coronavirus 2019 (COVID-19) pandemic because of the low cost and efficiency. We analyzed our in-house database of approved drug screens and compared their activity profiles with results from a severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) cytopathic effect (CPE) assay. The activity profiles of the human ether-à-go-go-related gene (hERG), phospholipidosis (PLD), and many cytotoxicity screens were found significantly correlated with anti-SARS-CoV-2 activity. hERG inhibition is a nonspecific off-target effect that has contributed to promiscuous drug interactions, whereas drug-induced PLD is an undesirable effect linked to hERG blockers. Thus, this study identifies preferred drug candidates as well as chemical structures that should be avoided because of their potential to induce toxicity. Lastly, we highlight the hERG liability of anti-SARS-CoV-2 drugs currently enrolled in clinical trials.
Collapse
Affiliation(s)
- Deborah K Ngan
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Tuan Xu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Menghang Xia
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Wei Zheng
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Ruili Huang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA.
| |
Collapse
|
31
|
Robitaille J, Denslow ND, Escher BI, Kurita-Oyamada HG, Marlatt V, Martyniuk CJ, Navarro-Martín L, Prosser R, Sanderson T, Yargeau V, Langlois VS. Towards regulation of Endocrine Disrupting chemicals (EDCs) in water resources using bioassays - A guide to developing a testing strategy. ENVIRONMENTAL RESEARCH 2022; 205:112483. [PMID: 34863984 DOI: 10.1016/j.envres.2021.112483] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 06/13/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are found in every environmental medium and are chemically diverse. Their presence in water resources can negatively impact the health of both human and wildlife. Currently, there are no mandatory screening mandates or regulations for EDC levels in complex water samples globally. Bioassays, which allow quantifying in vivo or in vitro biological effects of chemicals are used commonly to assess acute toxicity in water. The existing OECD framework to identify single-compound EDCs offers a set of bioassays that are validated for the Estrogen-, Androgen-, and Thyroid hormones, and for Steroidogenesis pathways (EATS). In this review, we discussed bioassays that could be potentially used to screen EDCs in water resources, including in vivo and in vitro bioassays using invertebrates, fish, amphibians, and/or mammalians species. Strengths and weaknesses of samples preparation for complex water samples are discussed. We also review how to calculate the Effect-Based Trigger values, which could serve as thresholds to determine if a given water sample poses a risk based on existing quality standards. This work aims to assist governments and regulatory agencies in developing a testing strategy towards regulation of EDCs in water resources worldwide. The main recommendations include 1) opting for internationally validated cell reporter in vitro bioassays to reduce animal use & cost; 2) testing for cell viability (a critical parameter) when using in vitro bioassays; and 3) evaluating the recovery of the water sample preparation method selected. This review also highlights future research avenues for the EDC screening revolution (e.g., 3D tissue culture, transgenic animals, OMICs, and Adverse Outcome Pathways (AOPs)).
Collapse
Affiliation(s)
- Julie Robitaille
- Centre Eau Terre Environnement, Institut National de La Recherche Scientifique (INRS), Quebec City, QC, Canada
| | | | - Beate I Escher
- Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany; Eberhard Karls University Tübingen, Tübingen, Germany
| | | | - Vicki Marlatt
- Simon Fraser University, Burnaby, British Columbia, Canada
| | | | - Laia Navarro-Martín
- Institute of Environmental Assessment and Water Research (IDAEA-CSIC), Barcelona, Spain
| | | | - Thomas Sanderson
- Centre Armand-Frappier Santé Biotechnologie, INRS, Laval, QC, Canada
| | | | - Valerie S Langlois
- Centre Eau Terre Environnement, Institut National de La Recherche Scientifique (INRS), Quebec City, QC, Canada.
| |
Collapse
|
32
|
Huang R. A Quantitative High-Throughput Screening Data Analysis Pipeline for Activity Profiling. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2474:133-145. [PMID: 35294762 DOI: 10.1007/978-1-0716-2213-1_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The U.S. Tox21 program has developed in vitro assays to test large collections of environmental chemicals in a quantitative high-throughput screening (qHTS) format, using triplicate 15-dose titrations to generate over 100 million data points to date. Counterscreens are also employed to minimize interferences from non-target-specific assay artifacts, such as compound autofluorescence and cytotoxicity. New data analysis approaches are needed to integrate these data and characterize the activities observed from these assays. Here, we describe a complete analysis pipeline that evaluates these qHTS data for technical quality in terms of signal reproducibility. We integrate signals from repeated assay runs, primary readouts and counterscreens to produce a final call on on-target compound activity.
Collapse
Affiliation(s)
- Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
33
|
Evaluation of an imaging-based in vitro screening platform for estrogenic activity with OECD reference chemicals. Toxicol In Vitro 2022; 81:105348. [DOI: 10.1016/j.tiv.2022.105348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 02/23/2022] [Accepted: 03/14/2022] [Indexed: 11/24/2022]
|
34
|
Krishna S, Borrel A, Huang R, Zhao J, Xia M, Kleinstreuer N. High-Throughput Chemical Screening and Structure-Based Models to Predict hERG Inhibition. BIOLOGY 2022; 11:209. [PMID: 35205076 PMCID: PMC8869358 DOI: 10.3390/biology11020209] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 12/23/2022]
Abstract
Chemical inhibition of the human ether-a -go-go-related gene (hERG) potassium channel leads to a prolonged QT interval that can contribute to severe cardiotoxicity. The adverse effects of hERG inhibition are one of the principal causes of drug attrition in clinical and pre-clinical development. Preliminary studies have demonstrated that a wide range of environmental chemicals and toxicants may also inhibit the hERG channel and contribute to the pathophysiology of cardiovascular (CV) diseases. As part of the US federal Tox21 program, the National Center for Advancing Translational Science (NCATS) applied a quantitative high throughput screening (qHTS) approach to screen the Tox21 library of 10,000 compounds (~7871 unique chemicals) at 14 concentrations in triplicate to identify chemicals perturbing hERG activity in the U2OS cell line thallium flux assay platform. The qHTS cell-based thallium influx assay provided a robust and reliable dataset to evaluate the ability of thousands of drugs and environmental chemicals to inhibit hERG channel protein, and the use of chemical structure-based clustering and chemotype enrichment analysis facilitated the identification of molecular features that are likely responsible for the observed hERG activity. We employed several machine-learning approaches to develop QSAR prediction models for the assessment of hERG liabilities for drug-like and environmental chemicals. The training set was compiled by integrating hERG bioactivity data from the ChEMBL database with the Tox21 qHTS thallium flux assay data. The best results were obtained with the random forest method (~92.6% balanced accuracy). The data and scripts used to generate hERG prediction models are provided in an open-access format as key in vitro and in silico tools that can be applied in a translational toxicology pipeline for drug development and environmental chemical screening.
Collapse
Affiliation(s)
- Shagun Krishna
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences (NIEHS), Research Triangle, NC 27560, USA;
| | | | - Ruili Huang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), Bethesda, MD 20892-4874, USA; (R.H.); (J.Z.); (M.X.)
| | - Jinghua Zhao
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), Bethesda, MD 20892-4874, USA; (R.H.); (J.Z.); (M.X.)
| | - Menghang Xia
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), Bethesda, MD 20892-4874, USA; (R.H.); (J.Z.); (M.X.)
| | - Nicole Kleinstreuer
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences (NIEHS), Research Triangle, NC 27560, USA;
| |
Collapse
|
35
|
Hsieh JH. Accounting for Artifacts in High-Throughput Toxicity Assays. Methods Mol Biol 2022; 2474:155-167. [PMID: 35294764 DOI: 10.1007/978-1-0716-2213-1_15] [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: 06/14/2023]
Abstract
Compound activity identification is the primary goal in high throughput screening (HTS) assays. However, assay artifacts including both systematic (e.g., compound autofluorescence) and nonsystematic (e.g., noise) complicate activity interpretation. In addition, other than the traditional potency parameter, half-maximal effect concentration [EC50], additional activity parameters (e.g., point-of-departure [POD] and weighted area-under-the-curve [wAUC]) could be derived from HTS data for activity profiling. A data analysis pipeline has been developed to handle the artifacts, and to provide compound activity characterization with either binary or continuous metrics. This chapter outlines the steps in the pipeline using Tox21 estrogen receptor (ER) β-lactamase assays, including the formats to identify either agonists or antagonists, as well as the counterscreen assays for identifying artifacts as examples. The steps can be applied to other lower throughput assays with concentration-response data.
Collapse
Affiliation(s)
- Jui-Hua Hsieh
- National Institute of Environmental Health Sciences, Durham, NC, USA.
| |
Collapse
|
36
|
Hunt JP, Galiardi J, Free TJ, Yang SO, Poole D, Zhao EL, Andersen JL, Wood DW, Bundy BC. Mechanistic discoveries and simulation-guided assay optimization of portable hormone biosensors with cell-free protein synthesis. Biotechnol J 2021; 17:e2100152. [PMID: 34761537 DOI: 10.1002/biot.202100152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 10/19/2021] [Accepted: 11/08/2021] [Indexed: 01/10/2023]
Abstract
Nuclear receptors (NRs) influence nearly every system of the body and our lives depend on correct NR signaling. Thus, a key environmental and pharmaceutical quest is to identify and detect chemicals which interact with nuclear hormone receptors, including endocrine disrupting chemicals (EDCs), therapeutic receptor modulators, and natural hormones. Previously reported biosensors of nuclear hormone receptor ligands facilitated rapid detection of NR ligands using cell-free protein synthesis (CFPS). In this work, the advantages of CFPS are further leveraged and combined with kinetic analysis, autoradiography, and western blot to elucidate the molecular mechanism of this biosensor. Additionally, mathematical simulations of enzyme kinetics are used to optimize the biosensor assay, ultimately lengthening its readable window by five-fold and improving sensor signal strength by two-fold. This approach enabled the creation of an on-demand thyroid hormone biosensor with an observable color-change readout. This mathematical and experimental approach provides insight for engineering rapid and field-deployable CFPS biosensors and promises to improve methods for detecting natural hormones, therapeutic receptor modulators, and EDCs.
Collapse
Affiliation(s)
- John Porter Hunt
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| | - Jackelyn Galiardi
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Tyler J Free
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| | - Seung Ook Yang
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| | - Daniel Poole
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Emily Long Zhao
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| | - Joshua L Andersen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - David W Wood
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Bradley C Bundy
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| |
Collapse
|
37
|
Xu T, Zheng W, Huang R. High-throughput screening assays for SARS-CoV-2 drug development: Current status and future directions. Drug Discov Today 2021; 26:2439-2444. [PMID: 34048893 PMCID: PMC8146264 DOI: 10.1016/j.drudis.2021.05.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/16/2021] [Accepted: 05/19/2021] [Indexed: 02/08/2023]
Abstract
In response to the ongoing coronavirus disease 2019 (COVID-19) pandemic, a panel of assays has been developed and applied to screen collections of approved and investigational drugs for anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) activity in a quantitative high-throughput screening (qHTS) format. In this review, we applied data-driven approaches to evaluate the ability of each assay to identify potential anti-SARS-CoV-2 leads. Multitarget assays were found to show advantages in terms of accuracy and efficiency over single-target assays, whereas target-specific assays were more suitable for investigating compound mechanisms of action. Moreover, strict filtering with counter screens might be more detrimental than beneficial in identifying true positives. Thus, developing novel HTS assays acting simultaneously against multiple targets in the SARS-CoV-2 life cycle will benefit anti-COVID-19 drug discovery.
Collapse
|
38
|
Ciallella HL, Russo DP, Aleksunes LM, Grimm FA, Zhu H. Revealing Adverse Outcome Pathways from Public High-Throughput Screening Data to Evaluate New Toxicants by a Knowledge-Based Deep Neural Network Approach. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2021; 55:10875-10887. [PMID: 34304572 PMCID: PMC8713073 DOI: 10.1021/acs.est.1c02656] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Traditional experimental testing to identify endocrine disruptors that enhance estrogenic signaling relies on expensive and labor-intensive experiments. We sought to design a knowledge-based deep neural network (k-DNN) approach to reveal and organize public high-throughput screening data for compounds with nuclear estrogen receptor α and β (ERα and ERβ) binding potentials. The target activity was rodent uterotrophic bioactivity driven by ERα/ERβ activations. After training, the resultant network successfully inferred critical relationships among ERα/ERβ target bioassays, shown as weights of 6521 edges between 1071 neurons. The resultant network uses an adverse outcome pathway (AOP) framework to mimic the signaling pathway initiated by ERα and identify compounds that mimic endogenous estrogens (i.e., estrogen mimetics). The k-DNN can predict estrogen mimetics by activating neurons representing several events in the ERα/ERβ signaling pathway. Therefore, this virtual pathway model, starting from a compound's chemistry initiating ERα activation and ending with rodent uterotrophic bioactivity, can efficiently and accurately prioritize new estrogen mimetics (AUC = 0.864-0.927). This k-DNN method is a potential universal computational toxicology strategy to utilize public high-throughput screening data to characterize hazards and prioritize potentially toxic compounds.
Collapse
Affiliation(s)
- Heather L Ciallella
- Center for Computational and Integrative Biology, Rutgers University Camden, Camden, New Jersey 08103, United States
| | - Daniel P Russo
- Center for Computational and Integrative Biology, Rutgers University Camden, Camden, New Jersey 08103, United States
- Department of Chemistry, Rutgers University Camden, Camden, New Jersey 08102, United States
| | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, United States
| | - Fabian A Grimm
- ExxonMobil Biomedical Sciences, Inc., Annandale, New Jersey 08801, United States
| | - Hao Zhu
- Center for Computational and Integrative Biology, Rutgers University Camden, Camden, New Jersey 08103, United States
- Department of Chemistry, Rutgers University Camden, Camden, New Jersey 08102, United States
| |
Collapse
|
39
|
Human Pluripotent Stem Cells for High-Throughput Drug Screening and Characterization of Small Molecules. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2454:811-827. [PMID: 34128205 DOI: 10.1007/7651_2021_394] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Human pluripotent stem cells (hPSCs), such as induced pluripotent stem cells (iPSCs), hold great promise for drug discovery, toxicology studies, and regenerative medicine. Here, we describe standardized protocols and experimental procedures that combine automated cell culture for scalable production of hPSCs with quantitative high-throughput screening (qHTS) in miniaturized 384-well plates. As a proof of principle, we established dose-response assessments and determined optimal concentrations of 12 small molecule compounds that are commonly used in the stem cell field. Multi-parametric analysis of readouts from diverse assays including cell viability, mitochondrial membrane potential, plasma membrane integrity, and ATP production was used to distinguish normal biological responses from cellular stress induced by small molecule treatment. Collectively, the establishment of integrated workflows for cell manufacturing, qHTS, high-content imaging, and data analysis provides an end-to-end platform for industrial-scale projects and should leverage the drug discovery process using hPSC-derived cell types.
Collapse
|
40
|
Sakamuru S, Zhao J, Xia M, Hong H, Simeonov A, Vaisman I, Huang R. Predictive Models to Identify Small Molecule Activators and Inhibitors of Opioid Receptors. J Chem Inf Model 2021; 61:2675-2685. [PMID: 34047186 DOI: 10.1021/acs.jcim.1c00439] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Opioid receptors (OPRs) are the main targets for the treatment of pain and related disorders. The opiate compounds that activate these receptors are effective analgesics but their use leads to adverse effects, and they often are highly addictive drugs of abuse. There is an urgent need for alternative chemicals that are analgesics and to reduce/avoid the unwanted effects in order to relieve the public health crisis of opioid addiction. Here, we aim to develop computational models to predict the OPR activity of small molecule compounds based on chemical structures and apply these models to identify novel OPR active compounds. We used four different machine learning algorithms to build models based on quantitative high throughput screening (qHTS) data sets of three OPRs in both agonist and antagonist modes. The best performing models were applied to virtually screen a large collection of compounds. The model predicted active compounds were experimentally validated using the same qHTS assays that generated the training data. Random forest was the best classifier with the highest performance metrics, and the mu OPR (OPRM)-agonist model achieved the best performance measured by AUC-ROC (0.88) and MCC (0.7) values. The model predicted actives resulted in hit rates ranging from 2.3% (delta OPR-agonist) to 15.8% (OPRM-agonist) after experimental confirmation. Compared to the original assay hit rate, all models enriched the hit rate by ≥2-fold. Our approach produced robust OPR prediction models that can be applied to prioritize compounds from large libraries for further experimental validation. The models identified several novel potent compounds as activators/inhibitors of OPRs that were confirmed experimentally. The potent hits were further investigated using molecular docking to find the interactions of the novel ligands in the active site of the corresponding OPR.
Collapse
Affiliation(s)
- Srilatha Sakamuru
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States.,Bioinformatics and Computational Biology, School of Systems Biology, College of Science, George Mason University, Manassas, Virginia 20110, United States
| | - Jinghua Zhao
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Menghang Xia
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Huixiao Hong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079, United States
| | - Anton Simeonov
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Iosif Vaisman
- Bioinformatics and Computational Biology, School of Systems Biology, College of Science, George Mason University, Manassas, Virginia 20110, United States
| | - Ruili Huang
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| |
Collapse
|
41
|
Duijndam B, Goudriaan A, van den Hoorn T, van der Stel W, Le Dévédec S, Bouwman P, van der Laan JW, van de Water B. Physiologically Relevant Estrogen Receptor Alpha Pathway Reporters for Single-Cell Imaging-Based Carcinogenic Hazard Assessment of Estrogenic Compounds. Toxicol Sci 2021; 181:187-198. [PMID: 33769548 PMCID: PMC8163057 DOI: 10.1093/toxsci/kfab037] [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] [Indexed: 11/13/2022] Open
Abstract
Estrogen receptor alpha (ERα) belongs to the nuclear hormone receptor family of ligand-inducible transcription factors and regulates gene networks in biological processes such as cell growth and proliferation. Disruption of these networks by chemical compounds with estrogenic activity can result in adverse outcomes such as unscheduled cell proliferation, ultimately culminating in tumor formation. To distinguish disruptive activation from normal physiological responses, it is essential to quantify relationships between different key events leading to a particular adverse outcome. For this purpose, we established fluorescent protein MCF7 reporter cell lines for ERα-induced proliferation by bacterial artificial chromosome-based tagging of 3 ERα target genes: GREB1, PGR, and TFF1. These target genes are inducible by the non-genotoxic carcinogen and ERα agonist 17β-estradiol in an ERα-dependent manner and are essential for ERα-dependent cell-cycle progression and proliferation. The 3 GFP reporter cell lines were characterized in detail and showed different activation dynamics upon exposure to 17β-estradiol. In addition, they demonstrated specific activation in response to other established reference estrogenic compounds of different potencies, with similar sensitivities as validated OECD test methods. This study shows that these fluorescent reporter cell lines can be used to monitor the spatial and temporal dynamics of ERα pathway activation at the single-cell level for more mechanistic insight, thereby allowing a detailed assessment of the potential carcinogenic activity of estrogenic compounds in humans.
Collapse
Affiliation(s)
- Britt Duijndam
- Division of Drug Discovery & Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden 2333CC, The Netherlands.,Section on Pharmacology, Toxicology and Kinetics, Medicines Evaluation Board, Utrecht 3531AH, The Netherlands
| | - Annabel Goudriaan
- Division of Drug Discovery & Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden 2333CC, The Netherlands
| | - Tineke van den Hoorn
- Section on Pharmacology, Toxicology and Kinetics, Medicines Evaluation Board, Utrecht 3531AH, The Netherlands
| | - Wanda van der Stel
- Division of Drug Discovery & Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden 2333CC, The Netherlands
| | - Sylvia Le Dévédec
- Division of Drug Discovery & Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden 2333CC, The Netherlands
| | - Peter Bouwman
- Division of Drug Discovery & Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden 2333CC, The Netherlands
| | - Jan Willem van der Laan
- Section on Pharmacology, Toxicology and Kinetics, Medicines Evaluation Board, Utrecht 3531AH, The Netherlands
| | - Bob van de Water
- Division of Drug Discovery & Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden 2333CC, The Netherlands
| |
Collapse
|
42
|
Chen Y, Tristan CA, Chen L, Jovanovic VM, Malley C, Chu PH, Ryu S, Deng T, Ormanoglu P, Tao D, Fang Y, Slamecka J, Hong H, LeClair CA, Michael S, Austin CP, Simeonov A, Singeç I. A versatile polypharmacology platform promotes cytoprotection and viability of human pluripotent and differentiated cells. Nat Methods 2021; 18:528-541. [PMID: 33941937 PMCID: PMC8314867 DOI: 10.1038/s41592-021-01126-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 03/22/2021] [Indexed: 12/27/2022]
Abstract
Clinical translation of human pluripotent stem cells (hPSCs) requires advanced strategies that ensure safe and robust long-term growth and functional differentiation. Pluripotent cells are capable of extensive self-renewal, yet remain highly sensitive to environmental perturbations in vitro, posing challenges to their therapeutic use. Here, we deployed innovative high-throughput screening strategies to identify a small molecule cocktail that dramatically improves viability of hPSCs and their differentiated progeny. The combination of Chroman 1, Emricasan, Polyamines, and Trans-ISRIB (CEPT) enhanced cell survival of genetically stable hPSCs by simultaneously blocking several stress mechanisms that otherwise compromise cell structure and function. CEPT provided strong improvements for several key applications in stem cell research, including routine cell passaging, cryopreservation of pluripotent and differentiated cells, embryoid body (EB) and organoid formation, single-cell cloning, and genome editing. Thus, CEPT represents a unique polypharmacology strategy for comprehensive cytoprotection, providing a new rationale for efficient and safe utilization of hPSCs. Conferring cell fitness by multi-target drug combinations may become a common approach in cryobiology, drug development, and regenerative medicine.
Collapse
Affiliation(s)
- Yu Chen
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Carlos A Tristan
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Lu Chen
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Vukasin M Jovanovic
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Claire Malley
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Pei-Hsuan Chu
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Seungmi Ryu
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Tao Deng
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Pinar Ormanoglu
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Dingyin Tao
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Yuhong Fang
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Jaroslav Slamecka
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Hyenjong Hong
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Christopher A LeClair
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Sam Michael
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Christopher P Austin
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Anton Simeonov
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA
| | - Ilyas Singeç
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD, USA.
| |
Collapse
|
43
|
Abstract
Toxicity analysis is a major challenge in drug design and discovery. Recently significant progress has been made through machine learning due to its accuracy, efficiency, and lower cost. US Toxicology in the 21st Century (Tox21) screened a large library of compounds, including approximately 12 000 environmental chemicals and drugs, for different mechanisms responsible for eliciting toxic effects. The Tox21 Data Challenge offered a platform to evaluate different computational methods for toxicity predictions. Inspired by the success of multiscale weighted colored graph (MWCG) theory in protein-ligand binding affinity predictions, we consider MWCG theory for toxicity analysis. In the present work, we develop a geometric graph learning toxicity (GGL-Tox) model by integrating MWCG features and the gradient boosting decision tree (GBDT) algorithm. The benchmark tests of the Tox21 Data Challenge are employed to demonstrate the utility and usefulness of the proposed GGL-Tox model. An extensive comparison with other state-of-the-art models indicates that GGL-Tox is an accurate and efficient model for toxicity analysis and prediction.
Collapse
Affiliation(s)
- Jian Jiang
- Research Center of Nonlinear Science, College of Mathematics and Computer Science, Engineering Research Center of Hubei Province for Clothing Information, Wuhan Textile University, Wuhan 430200, P R. China
| | - Rui Wang
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
44
|
Li S, Zhao J, Huang R, Travers J, Klumpp-Thomas C, Yu W, MacKerell AD, Sakamuru S, Ooka M, Xue F, Sipes NS, Hsieh JH, Ryan K, Simeonov A, Santillo MF, Xia M. Profiling the Tox21 Chemical Collection for Acetylcholinesterase Inhibition. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:47008. [PMID: 33844597 PMCID: PMC8041433 DOI: 10.1289/ehp6993] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/01/2021] [Accepted: 03/09/2021] [Indexed: 05/26/2023]
Abstract
BACKGROUND Inhibition of acetylcholinesterase (AChE), a biomarker of organophosphorous and carbamate exposure in environmental and occupational human health, has been commonly used to identify potential safety liabilities. So far, many environmental chemicals, including drug candidates, food additives, and industrial chemicals, have not been thoroughly evaluated for their inhibitory effects on AChE activity. AChE inhibitors can have therapeutic applications (e.g., tacrine and donepezil) or neurotoxic consequences (e.g., insecticides and nerve agents). OBJECTIVES The objective of the current study was to identify environmental chemicals that inhibit AChE activity using in vitro and in silico models. METHODS To identify AChE inhibitors rapidly and efficiently, we have screened the Toxicology in the 21st Century (Tox21) 10K compound library in a quantitative high-throughput screening (qHTS) platform by using the homogenous cell-based AChE inhibition assay and enzyme-based AChE inhibition assays (with or without microsomes). AChE inhibitors identified from the primary screening were further tested in monolayer or spheroid formed by SH-SY5Y and neural stem cell models. The inhibition and binding modes of these identified compounds were studied with time-dependent enzyme-based AChE inhibition assay and molecular docking, respectively. RESULTS A group of known AChE inhibitors, such as donepezil, ambenonium dichloride, and tacrine hydrochloride, as well as many previously unreported AChE inhibitors, such as chelerythrine chloride and cilostazol, were identified in this study. Many of these compounds, such as pyrazophos, phosalone, and triazophos, needed metabolic activation. This study identified both reversible (e.g., donepezil and tacrine) and irreversible inhibitors (e.g., chlorpyrifos and bromophos-ethyl). Molecular docking analyses were performed to explain the relative inhibitory potency of selected compounds. CONCLUSIONS Our tiered qHTS approach allowed us to generate a robust and reliable data set to evaluate large sets of environmental compounds for their AChE inhibitory activity. https://doi.org/10.1289/EHP6993.
Collapse
Affiliation(s)
- Shuaizhang Li
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Jinghua Zhao
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Ruili Huang
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Jameson Travers
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Carleen Klumpp-Thomas
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Wenbo Yu
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland, USA
| | | | - Srilatha Sakamuru
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Masato Ooka
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Fengtian Xue
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland, USA
| | - Nisha S. Sipes
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Jui-Hua Hsieh
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Kristen Ryan
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Anton Simeonov
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Michael F. Santillo
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, Maryland, USA
| | - Menghang Xia
- Division for Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| |
Collapse
|
45
|
Zhu H, Chen CZ, Sakamuru S, Zhao J, Ngan DK, Simeonov A, Hall MD, Xia M, Zheng W, Huang R. Mining of high throughput screening database reveals AP-1 and autophagy pathways as potential targets for COVID-19 therapeutics. Sci Rep 2021; 11:6725. [PMID: 33762619 PMCID: PMC7990955 DOI: 10.1038/s41598-021-86110-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
The recent global pandemic of the Coronavirus disease 2019 (COVID-19) caused by the new coronavirus SARS-CoV-2 presents an urgent need for the development of new therapeutic candidates. Many efforts have been devoted to screening existing drug libraries with the hope to repurpose approved drugs as potential treatments for COVID-19. However, the antiviral mechanisms of action of the drugs found active in these phenotypic screens remain largely unknown. In an effort to deconvolute the viral targets in pursuit of more effective anti-COVID-19 drug development, we mined our in-house database of approved drug screens against 994 assays and compared their activity profiles with the drug activity profile in a cytopathic effect (CPE) assay of SARS-CoV-2. We found that the autophagy and AP-1 signaling pathway activity profiles are significantly correlated with the anti-SARS-CoV-2 activity profile. In addition, a class of neurology/psychiatry drugs was found to be significantly enriched with anti-SARS-CoV-2 activity. Taken together, these results provide new insights into SARS-CoV-2 infection and potential targets for COVID-19 therapeutics, which can be further validated by in vivo animal studies and human clinical trials.
Collapse
Affiliation(s)
- Hu Zhu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), DPI/NCATS, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Catherine Z Chen
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), DPI/NCATS, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Srilatha Sakamuru
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), DPI/NCATS, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Jinghua Zhao
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), DPI/NCATS, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Deborah K Ngan
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), DPI/NCATS, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Anton Simeonov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), DPI/NCATS, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Mathew D Hall
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), DPI/NCATS, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Menghang Xia
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), DPI/NCATS, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Wei Zheng
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), DPI/NCATS, 9800 Medical Center Drive, Rockville, MD, 20850, USA
| | - Ruili Huang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), DPI/NCATS, 9800 Medical Center Drive, Rockville, MD, 20850, USA.
| |
Collapse
|
46
|
Richard AM, Huang R, Waidyanatha S, Shinn P, Collins BJ, Thillainadarajah I, Grulke CM, Williams AJ, Lougee RR, Judson RS, Houck KA, Shobair M, Yang C, Rathman JF, Yasgar A, Fitzpatrick SC, Simeonov A, Thomas RS, Crofton KM, Paules RS, Bucher JR, Austin CP, Kavlock RJ, Tice RR. The Tox21 10K Compound Library: Collaborative Chemistry Advancing Toxicology. Chem Res Toxicol 2021. [PMID: 33140634 DOI: 10.1021/acs.chemrestox.0c0026410.1021/acs.chemrestox.0c00264.s003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Since 2009, the Tox21 project has screened ∼8500 chemicals in more than 70 high-throughput assays, generating upward of 100 million data points, with all data publicly available through partner websites at the United States Environmental Protection Agency (EPA), National Center for Advancing Translational Sciences (NCATS), and National Toxicology Program (NTP). Underpinning this public effort is the largest compound library ever constructed specifically for improving understanding of the chemical basis of toxicity across research and regulatory domains. Each Tox21 federal partner brought specialized resources and capabilities to the partnership, including three approximately equal-sized compound libraries. All Tox21 data generated to date have resulted from a confluence of ideas, technologies, and expertise used to design, screen, and analyze the Tox21 10K library. The different programmatic objectives of the partners led to three distinct, overlapping compound libraries that, when combined, not only covered a diversity of chemical structures, use-categories, and properties but also incorporated many types of compound replicates. The history of development of the Tox21 "10K" chemical library and data workflows implemented to ensure quality chemical annotations and allow for various reproducibility assessments are described. Cheminformatics profiling demonstrates how the three partner libraries complement one another to expand the reach of each individual library, as reflected in coverage of regulatory lists, predicted toxicity end points, and physicochemical properties. ToxPrint chemotypes (CTs) and enrichment approaches further demonstrate how the combined partner libraries amplify structure-activity patterns that would otherwise not be detected. Finally, CT enrichments are used to probe global patterns of activity in combined ToxCast and Tox21 activity data sets relative to test-set size and chemical versus biological end point diversity, illustrating the power of CT approaches to discern patterns in chemical-activity data sets. These results support a central premise of the Tox21 program: A collaborative merging of programmatically distinct compound libraries would yield greater rewards than could be achieved separately.
Collapse
Affiliation(s)
- Ann M Richard
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Suramya Waidyanatha
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, United States
| | - Paul Shinn
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Bradley J Collins
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, United States
| | - Inthirany Thillainadarajah
- Senior Environmental Employment Program, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Christopher M Grulke
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Antony J Williams
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Ryan R Lougee
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
- Oak Ridge Institute for Science and Education, United States Department of Energy, Oak Ridge, Tennessee 37830, United States
| | - Richard S Judson
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Keith A Houck
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Mahmoud Shobair
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Chihae Yang
- Altamira, LLC, Columbus, Ohio 43235, United States
- Molecular Networks, GmbH, Erlangen 90411, Germany
| | - James F Rathman
- Altamira, LLC, Columbus, Ohio 43235, United States
- Molecular Networks, GmbH, Erlangen 90411, Germany
| | - Adam Yasgar
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Suzanne C Fitzpatrick
- Center for Food Safety and Applied Nutrition, United States Food and Drug Administration, College Park, Maryland 20740, United States
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Russell S Thomas
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Kevin M Crofton
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
- R3Fellows, LLC, Durham, North Carolina 27701, United States
| | - Richard S Paules
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, United States
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, United States
| | - John R Bucher
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, United States
| | - Christopher P Austin
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Robert J Kavlock
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
- Kavlock Consulting, LLC, Washington, DC 20001, United States
| | - Raymond R Tice
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, United States
- RTice Consulting, Hillsborough, North Carolina 27278, United States
| |
Collapse
|
47
|
Richard AM, Huang R, Waidyanatha S, Shinn P, Collins BJ, Thillainadarajah I, Grulke CM, Williams AJ, Lougee RR, Judson RS, Houck KA, Shobair M, Yang C, Rathman JF, Yasgar A, Fitzpatrick SC, Simeonov A, Thomas RS, Crofton KM, Paules RS, Bucher JR, Austin CP, Kavlock RJ, Tice RR. The Tox21 10K Compound Library: Collaborative Chemistry Advancing Toxicology. Chem Res Toxicol 2021; 34:189-216. [PMID: 33140634 PMCID: PMC7887805 DOI: 10.1021/acs.chemrestox.0c00264] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Indexed: 12/13/2022]
Abstract
Since 2009, the Tox21 project has screened ∼8500 chemicals in more than 70 high-throughput assays, generating upward of 100 million data points, with all data publicly available through partner websites at the United States Environmental Protection Agency (EPA), National Center for Advancing Translational Sciences (NCATS), and National Toxicology Program (NTP). Underpinning this public effort is the largest compound library ever constructed specifically for improving understanding of the chemical basis of toxicity across research and regulatory domains. Each Tox21 federal partner brought specialized resources and capabilities to the partnership, including three approximately equal-sized compound libraries. All Tox21 data generated to date have resulted from a confluence of ideas, technologies, and expertise used to design, screen, and analyze the Tox21 10K library. The different programmatic objectives of the partners led to three distinct, overlapping compound libraries that, when combined, not only covered a diversity of chemical structures, use-categories, and properties but also incorporated many types of compound replicates. The history of development of the Tox21 "10K" chemical library and data workflows implemented to ensure quality chemical annotations and allow for various reproducibility assessments are described. Cheminformatics profiling demonstrates how the three partner libraries complement one another to expand the reach of each individual library, as reflected in coverage of regulatory lists, predicted toxicity end points, and physicochemical properties. ToxPrint chemotypes (CTs) and enrichment approaches further demonstrate how the combined partner libraries amplify structure-activity patterns that would otherwise not be detected. Finally, CT enrichments are used to probe global patterns of activity in combined ToxCast and Tox21 activity data sets relative to test-set size and chemical versus biological end point diversity, illustrating the power of CT approaches to discern patterns in chemical-activity data sets. These results support a central premise of the Tox21 program: A collaborative merging of programmatically distinct compound libraries would yield greater rewards than could be achieved separately.
Collapse
Affiliation(s)
- Ann M. Richard
- Center
for Computational Toxicology and Exposure, Office of Research and
Development, United States Environmental
Protection Agency, Research
Triangle Park, North Carolina 27711, United States
| | - Ruili Huang
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Suramya Waidyanatha
- Division
of the National Toxicology Program, National
Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, United States
| | - Paul Shinn
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Bradley J. Collins
- Division
of the National Toxicology Program, National
Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, United States
| | - Inthirany Thillainadarajah
- Senior
Environmental Employment Program, United
States Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Christopher M. Grulke
- Center
for Computational Toxicology and Exposure, Office of Research and
Development, United States Environmental
Protection Agency, Research
Triangle Park, North Carolina 27711, United States
| | - Antony J. Williams
- Center
for Computational Toxicology and Exposure, Office of Research and
Development, United States Environmental
Protection Agency, Research
Triangle Park, North Carolina 27711, United States
| | - Ryan R. Lougee
- Center
for Computational Toxicology and Exposure, Office of Research and
Development, United States Environmental
Protection Agency, Research
Triangle Park, North Carolina 27711, United States
- Oak
Ridge Institute for Science and Education, United States Department
of Energy, Oak Ridge, Tennessee 37830, United States
| | - Richard S. Judson
- Center
for Computational Toxicology and Exposure, Office of Research and
Development, United States Environmental
Protection Agency, Research
Triangle Park, North Carolina 27711, United States
| | - Keith A. Houck
- Center
for Computational Toxicology and Exposure, Office of Research and
Development, United States Environmental
Protection Agency, Research
Triangle Park, North Carolina 27711, United States
| | - Mahmoud Shobair
- Center
for Computational Toxicology and Exposure, Office of Research and
Development, United States Environmental
Protection Agency, Research
Triangle Park, North Carolina 27711, United States
| | - Chihae Yang
- Altamira,
LLC, Columbus, Ohio 43235, United States
- Molecular Networks, GmbH, Erlangen 90411, Germany
| | - James F. Rathman
- Altamira,
LLC, Columbus, Ohio 43235, United States
- Molecular Networks, GmbH, Erlangen 90411, Germany
| | - Adam Yasgar
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Suzanne C. Fitzpatrick
- Center
for Food Safety and Applied Nutrition, United
States Food and Drug Administration, College Park, Maryland 20740, United States
| | - Anton Simeonov
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Russell S. Thomas
- Center
for Computational Toxicology and Exposure, Office of Research and
Development, United States Environmental
Protection Agency, Research
Triangle Park, North Carolina 27711, United States
| | - Kevin M. Crofton
- Center
for Computational Toxicology and Exposure, Office of Research and
Development, United States Environmental
Protection Agency, Research
Triangle Park, North Carolina 27711, United States
- R3Fellows,
LLC, Durham, North Carolina 27701, United States
| | - Richard S. Paules
- Division
of the National Toxicology Program, National
Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, United States
- Division
of the National Toxicology Program, National
Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, United States
| | - John R. Bucher
- Division
of the National Toxicology Program, National
Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, United States
| | - Christopher P. Austin
- National
Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Robert J. Kavlock
- Center
for Computational Toxicology and Exposure, Office of Research and
Development, United States Environmental
Protection Agency, Research
Triangle Park, North Carolina 27711, United States
- Kavlock
Consulting, LLC, Washington, DC 20001, United States
| | - Raymond R. Tice
- Division
of the National Toxicology Program, National
Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, United States
- RTice Consulting, Hillsborough, North Carolina 27278, United States
| |
Collapse
|
48
|
Xu T, Wu L, Xia M, Simeonov A, Huang R. Systematic Identification of Molecular Targets and Pathways Related to Human Organ Level Toxicity. Chem Res Toxicol 2020; 34:412-421. [PMID: 33251791 DOI: 10.1021/acs.chemrestox.0c00305] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The mechanisms leading to organ level toxicities are poorly understood. In this study, we applied an integrated approach to deduce the molecular targets and biological pathways involved in chemically induced toxicity for eight common human organ level toxicity end points (carcinogenicity, cardiotoxicity, developmental toxicity, hepatotoxicity, nephrotoxicity, neurotoxicity, reproductive toxicity, and skin toxicity). Integrated analysis of in vitro assay data, molecular targets and pathway annotations from the literature, and toxicity-molecular target associations derived from text mining, combined with machine learning techniques, were used to generate molecular targets for each of the organ level toxicity end points. A total of 1516 toxicity-related genes were identified and subsequently analyzed for biological pathway coverage, resulting in 206 significant pathways (p-value <0.05), ranging from 3 (e.g., developmental toxicity) to 101 (e.g., skin toxicity) for each toxicity end point. This study presents a systematic and comprehensive analysis of molecular targets and pathways related to various in vivo toxicity end points. These molecular targets and pathways could aid in understanding the biological mechanisms of toxicity and serve as a guide for the design of suitable in vitro assays for more efficient toxicity testing. In addition, these results are complementary to the existing adverse outcome pathway (AOP) framework and can be used to aid in the development of novel AOPs. Our results provide abundant testable hypotheses for further experimental validation.
Collapse
Affiliation(s)
- Tuan Xu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Leihong Wu
- National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Menghang Xia
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Anton Simeonov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| | - Ruili Huang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland 20850, United States
| |
Collapse
|
49
|
Zhang S, Khan WA, Su L, Zhang X, Li C, Qin W, Zhao Y. Predicting oxidative stress induced by organic chemicals by using quantitative Structure-Activity relationship methods. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 201:110817. [PMID: 32512417 DOI: 10.1016/j.ecoenv.2020.110817] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 06/11/2023]
Abstract
Cellular exposure to xenobiotic human-made products will lead to oxidative stress that gives rise to DNA damage, as well as chemical or mechanical damage. Distinguishing the chemicals that will induce oxidative stress and predicting their toxicity is necessary. In the present study, 4270 compounds in the ARE-bla assay were investigated to predict active and inactive compounds by using simple algorithms, namely, recursive partitioning (RP) and binomial logistic regression, and to develop the quantitative structure-activity relationship (QSAR) models of chemicals that activate the ARE pathway to induce oxidative stress and exert toxic effects on cells. A decision tree based on scaffold-based fragments obtained through RP analysis showed the best identification accuracy. However, the overall identification accuracy of this model for active compounds was unsatisfactory due to limited fragments. Furthermore, a binomial logistic regression model was developed from 638 active compounds and 3632 inactive chemicals. The model with a cutoff of 0.15 could predict chemicals that were active or inactive with the prediction accuracy of 69.1%. Its area under the receiver operating characteristic (ROC) curve metric (AUROC) was 0.762, which indicated the acceptable predictive ability of this model. The parameters nBM (number of multiple bonds) and H% (percentage of H atom) played dominant roles in the prediction of the activity (inactive or active) of chemicals. A global QSAR model was developed to predict the toxicity of active chemicals. However, the model displayed an unsatisfactory result with R2 = 0.316 and R2ext = 0.090. Active chemicals were then classified on the basis of structure. A total of 79 compounds with carbon chains could be predicted with acceptable performance by using a QSAR model with six descriptors (R2 = 0.722, R2ext = 0.798, Q2Loo = 0.654, Q2Boot = 0.755, Q2ext = 0.721). The simple models established here contribute to efforts on identification compounds inducing oxidative stress and provide the scientific basis for risk assessment to organisms in the environment.
Collapse
Affiliation(s)
- Shengnan Zhang
- School of Environment, And State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, Northeast Normal University, 2555 Jingyue Street, Changchun, 130117, Jilin, PR China
| | - Waqas Amin Khan
- School of Environment, And State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, Northeast Normal University, 2555 Jingyue Street, Changchun, 130117, Jilin, PR China
| | - Limin Su
- School of Environment, And State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, Northeast Normal University, 2555 Jingyue Street, Changchun, 130117, Jilin, PR China.
| | - Xuehua Zhang
- School of Water Conservancy and Environment Engineering, Changchun Institute of Technology, Changchun, 130012, Jilin, PR China
| | - Chao Li
- School of Environment, And State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, Northeast Normal University, 2555 Jingyue Street, Changchun, 130117, Jilin, PR China
| | - Weichao Qin
- School of Environment, And State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, Northeast Normal University, 2555 Jingyue Street, Changchun, 130117, Jilin, PR China
| | - Yuanhui Zhao
- School of Environment, And State Environmental Protection Key Laboratory of Wetland Ecology and Vegetation Restoration, Northeast Normal University, 2555 Jingyue Street, Changchun, 130117, Jilin, PR China
| |
Collapse
|
50
|
Matsuzaka Y, Uesawa Y. Molecular Image-Based Prediction Models of Nuclear Receptor Agonists and Antagonists Using the DeepSnap-Deep Learning Approach with the Tox21 10K Library. Molecules 2020; 25:molecules25122764. [PMID: 32549344 PMCID: PMC7356846 DOI: 10.3390/molecules25122764] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/06/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023] Open
Abstract
The interaction of nuclear receptors (NRs) with chemical compounds can cause dysregulation of endocrine signaling pathways, leading to adverse health outcomes due to the disruption of natural hormones. Thus, identifying possible ligands of NRs is a crucial task for understanding the adverse outcome pathway (AOP) for human toxicity as well as the development of novel drugs. However, the experimental assessment of novel ligands remains expensive and time-consuming. Therefore, an in silico approach with a wide range of applications instead of experimental examination is highly desirable. The recently developed novel molecular image-based deep learning (DL) method, DeepSnap-DL, can produce multiple snapshots from three-dimensional (3D) chemical structures and has achieved high performance in the prediction of chemicals for toxicological evaluation. In this study, we used DeepSnap-DL to construct prediction models of 35 agonist and antagonist allosteric modulators of NRs for chemicals derived from the Tox21 10K library. We demonstrate the high performance of DeepSnap-DL in constructing prediction models. These findings may aid in interpreting the key molecular events of toxicity and support the development of new fields of machine learning to identify environmental chemicals with the potential to interact with NR signaling pathways.
Collapse
|