1
|
Takeshita JI, Goto Y, Yamamoto S, Sasaki T, Yoshinari K. Comprehensive analysis of the toxicity-related findings from repeated-dose subacute toxicity studies of industrial chemicals in male rats. Crit Rev Toxicol 2024; 54:996-1010. [PMID: 39636601 DOI: 10.1080/10408444.2024.2427221] [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: 07/06/2024] [Revised: 11/01/2024] [Accepted: 11/01/2024] [Indexed: 12/07/2024]
Abstract
The demand for alternatives to animal testing has increased, but there has been no significant progress in developing alternatives for repeated-dose toxicity tests despite their importance in chemical risk assessment. A comprehensive analysis of existing toxicity studies is the first step toward understanding toxicity and developing alternatives. However, such an analysis has yet to be performed for industrial chemicals. Therefore, we collected and organized publicly available repeated-dose subacute toxicity studies in male rats and constructed a database consisting of more than 2000 toxicity studies with about 500 toxicity-related findings. We then analyzed the no observed effect and lowest observed effect levels, toxicity-related findings, and organ categories in the database. The analyses revealed commonly and uncommonly observed toxicity-related findings and organ categories, as well as toxicity-related findings and organ categories with low and high median lowest observed effect levels. In addition, we extracted the toxicity studies registered in the Japanese and European chemical regulatory systems and conducted the same analysis for these datasets as the entire database. The results suggest that commonly observed toxicity-related findings were similar, but some toxicity-related findings differed in the frequency of observations between the two datasets.
Collapse
Affiliation(s)
- Jun-Ichi Takeshita
- Research Institute of Science for Safety and Sustainability, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yoshitaka Goto
- Sustainability Consulting Division 2, Mizuho Research & Technologies, Ltd., Tokyo, Japan
| | - Shinji Yamamoto
- Business Headquarters 2nd Segment, Research Institute of Systems Planning, Inc., Tokyo, Japan
| | - Takamitsu Sasaki
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kouichi Yoshinari
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
2
|
Matsushita K, Toyoda T, Akane H, Morikawa T, Ogawa K. Role of CD44 expressed in renal tubules during maladaptive repair in renal fibrogenesis in an allopurinol-induced rat model of chronic kidney disease. J Appl Toxicol 2024; 44:455-469. [PMID: 37876353 DOI: 10.1002/jat.4554] [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] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/30/2023] [Accepted: 10/01/2023] [Indexed: 10/26/2023]
Abstract
The kidney is a major target organ for the adverse effects of pharmaceuticals; renal tubular epithelial cells (TECs) are particularly vulnerable to drug-induced toxicity. TECs have regenerative capacity; however, maladaptive repair of TECs after injury leads to renal fibrosis, resulting in chronic kidney disease (CKD). We previously reported the specific expression of CD44 in failed-repair TECs of rat CKD model induced by ischemia reperfusion injury. Here, we investigated the pathophysiological role of CD44 in renal fibrogenesis in allopurinol-treated rat CKD model. Dilated or atrophic TECs expressing CD44 in fibrotic areas were collected by laser microdissection and subjected to microarray analysis. Gene ontology showed that extracellular matrix (ECM)-related genes were upregulated and differentiation-related genes were downregulated in dilated/atrophic TECs. Ingenuity Pathway Analysis identified CD44 as an upstream regulator of fibrosis-related genes, including Fn1, which encodes fibronectin. Immunohistochemistry demonstrated that dilated/atrophic TECs expressing CD44 showed decreases in differentiation markers of TECs and clear expression of mesenchymal markers during basement membrane attachment. In situ hybridization revealed an increase in Fn1 mRNA in the cytoplasm of dilated/atrophic TECs, whereas fibronectin was localized in the stroma around these TECs, supporting the production/secretion of ECM by dilated/atrophic TECs. Overall, these data indicated that dilated/atrophic TECs underwent a partial epithelial-mesenchymal transition (pEMT) and that CD44 promoted renal fibrogenesis via induction of ECM production in failed-repair TECs exhibiting pEMT. CD44 was detected in the urine and serum of APL-treated rats, which may reflect the expression of CD44 in the kidney.
Collapse
Affiliation(s)
- Kohei Matsushita
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Takeshi Toyoda
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Hirotoshi Akane
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Tomomi Morikawa
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Kumiko Ogawa
- Division of Pathology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| |
Collapse
|
3
|
Pannala VR, Wallqvist A. High-Throughput Transcriptomics Differentiates Toxic versus Non-Toxic Chemical Exposures Using a Rat Liver Model. Int J Mol Sci 2023; 24:17425. [PMID: 38139254 PMCID: PMC10743995 DOI: 10.3390/ijms242417425] [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] [Received: 11/14/2023] [Revised: 12/07/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023] Open
Abstract
To address the challenge of limited throughput with traditional toxicity testing, a newly developed high-throughput transcriptomics (HTT) platform, together with a 5-day in vivo rat model, offers an alternative approach to estimate chemical exposures and provide reasonable estimates of toxicological endpoints. This study contains an HTT analysis of 18 environmental chemicals with known liver toxicity. They were evaluated using male Sprague Dawley rats exposed to various concentrations daily for five consecutive days via oral gavage, with data collected on the sixth day. Here, we further explored the 5-day rat model to identify potential gene signatures that can differentiate between toxic and non-toxic liver responses and provide us with a potential histopathological endpoint of chemical exposure. We identified a distinct gene expression pattern that differentiated non-hepatotoxic compounds from hepatotoxic compounds in a dose-dependent manner, and an analysis of the significantly altered common genes indicated that toxic chemicals predominantly upregulated most of the genes and several pathways in amino acid and lipid metabolism. Finally, our liver injury module analysis revealed that several liver-toxic compounds showed similarities in the key injury phenotypes of cellular inflammation and proliferation, indicating potential molecular initiating processes that may lead to a specific end-stage liver disease.
Collapse
Affiliation(s)
- Venkat R. Pannala
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Frederick, MD 21702, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Frederick, MD 21702, USA
| |
Collapse
|
4
|
Kimura Y, Nakazawa S, Nishigori K, Mori Y, Ichihara J, Yoshioka Y. Ultra-high-field pharmacological functional MRI of dopamine D1 receptor-related interventions in anesthetized rats. Pharmacol Res Perspect 2023; 11:e01055. [PMID: 36807574 PMCID: PMC9939738 DOI: 10.1002/prp2.1055] [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: 06/03/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 02/22/2023] Open
Abstract
The dopamine D1 receptor (D1R) is associated with schizophrenia, Parkinson's disease, and attention deficit hyperactivity disorder. Although the receptor is considered a therapeutic target for these diseases, its neurophysiological function has not been fully elucidated. Pharmacological functional MRI (phfMRI) has been used to evaluate regional brain hemodynamic changes induced by neurovascular coupling resulting from pharmacological interventions, thus phfMRI studies can be used to help understand the neurophysiological function of specific receptors. Herein, the blood oxygenation level-dependent (BOLD) signal changes associated with D1R action in anesthetized rats was investigated by using a preclinical ultra-high-field 11.7-T MRI scanner. PhfMRI was performed before and after administration of the D1-like receptor agonist (SKF82958), antagonist (SCH39166), or physiological saline subcutaneously. Compared to saline, the D1-agonist induced a BOLD signal increase in the striatum, thalamus, prefrontal cortex, and cerebellum. At the same time, the D1-antagonist reduced the BOLD signal in the striatum, thalamus, and cerebellum by evaluating temporal profiles. PhfMRI detected D1R-related BOLD signal changes in the brain regions associated with high expression of D1R. We also measured the early expression of c-fos at the mRNA level to evaluate the effects of SKF82958 and isoflurane anesthesia on neuronal activity. Regardless of the presence of isoflurane anesthesia, c-fos expression level was increased in the region where positive BOLD responses were observed with administration of SKF82958. These findings demonstrated that phfMRI could be used to identify the effects of direct D1 blockade on physiological brain functions and also for neurophysiological assessment of dopamine receptor functions in living animals.
Collapse
Affiliation(s)
- Yuka Kimura
- Drug Development Research LaboratoriesSumitomo Dainippon Pharma Co LtdOsakaJapan
- Graduate School of Science and Technology, Division of Information ScienceNara Institute of Science and Technology (NAIST)IkomaJapan
- Present address:
Platform Technology Research UnitSumitomo Pharma Co LtdOsakaJapan
| | - Shunsuke Nakazawa
- Drug Development Research LaboratoriesSumitomo Dainippon Pharma Co LtdOsakaJapan
- Present address:
Global Corporate StrategySumitomo Pharma Co LtdOsakaJapan
| | - Kantaro Nishigori
- Drug Development Research LaboratoriesSumitomo Dainippon Pharma Co LtdOsakaJapan
- Present address:
Platform Technology Research UnitSumitomo Pharma Co LtdOsakaJapan
| | - Yuki Mori
- Center for Information and Neural Networks (CiNet), National Institute of Information and Communications TechnologyOsaka UniversityOsakaJapan
- Biofunctional Imaging Laboratory, Immunology Frontier Research Center (IFReC)Osaka UniversityOsakaJapan
- Present address:
Center for Translational NeuromedicineUniversity of CopenhagenCopenhagen NDenmark
| | - Junji Ichihara
- Drug Development Research LaboratoriesSumitomo Dainippon Pharma Co LtdOsakaJapan
- Present address:
Bioscience Research LaboratorySumitomo Chemical Co LtdOsakaJapan
| | - Yoshichika Yoshioka
- Center for Information and Neural Networks (CiNet), National Institute of Information and Communications TechnologyOsaka UniversityOsakaJapan
- Biofunctional Imaging Laboratory, Immunology Frontier Research Center (IFReC)Osaka UniversityOsakaJapan
- Present address:
Graduate School of Frontier BiosciencesOsaka UniversityOsakaJapan
| |
Collapse
|
5
|
Su R, He H, Sun C, Wang X, Liu X. Prediction of drug-induced hepatotoxicity based on histopathological whole slide images. Methods 2023; 212:31-38. [PMID: 36706825 DOI: 10.1016/j.ymeth.2023.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 12/30/2022] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Liver is an important metabolic organ in human body and is sensitive to toxic chemicals or drugs. Adverse reactions caused by drug hepatotoxicity will damage the liver and hepatotoxicity is the leading cause of removal of approved drugs from the market. Therefore, it is of great significance to identify liver toxicity as early as possible in the drug development process. In this study, we developed a predictive model for drug hepatotoxicity based on histopathological whole slide images (WSI) which are the by-product of drug experiments and have received little attention. To better represent the WSIs, we constructed a graph representation for each WSI by dividing it into small patches, taking sampled patches as nodes and calculating the correlation coefficients between node features as the edges of the graph structure. Then a WSI-level graph convolutional network (GCN) was built to effectively extract the node information of the graph and predict the toxicity. In addition, we introduced a gated attention global context vector (gaGCV) to combine the global context to make node features to contain more comprehensive information. The results validated on rat liver in vivo data from the Open TG-GATES show that the use of WSI for the prediction of toxicity is feasible and effective.
Collapse
Affiliation(s)
- Ran Su
- School of Computer Software, College of Intelligence and Computing, Tianjin University, China
| | - Hao He
- School of Computer Software, College of Intelligence and Computing, Tianjin University, China
| | | | - Xiaomin Wang
- National Clinical Research Center for Infectious Diseases, Shenzhen, Guangdong, China.
| | - Xiaofeng Liu
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| |
Collapse
|
6
|
Su R, Yang H, Wei L, Chen S, Zou Q. A multi-label learning model for predicting drug-induced pathology in multi-organ based on toxicogenomics data. PLoS Comput Biol 2022; 18:e1010402. [PMID: 36070305 PMCID: PMC9451100 DOI: 10.1371/journal.pcbi.1010402] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022] Open
Abstract
Drug-induced toxicity damages the health and is one of the key factors causing drug withdrawal from the market. It is of great significance to identify drug-induced target-organ toxicity, especially the detailed pathological findings, which are crucial for toxicity assessment, in the early stage of drug development process. A large variety of studies have devoted to identify drug toxicity. However, most of them are limited to single organ or only binary toxicity. Here we proposed a novel multi-label learning model named Att-RethinkNet, for predicting drug-induced pathological findings targeted on liver and kidney based on toxicogenomics data. The Att-RethinkNet is equipped with a memory structure and can effectively use the label association information. Besides, attention mechanism is embedded to focus on the important features and obtain better feature presentation. Our Att-RethinkNet is applicable in multiple organs and takes account the compound type, dose, and administration time, so it is more comprehensive and generalized. And more importantly, it predicts multiple pathological findings at the same time, instead of predicting each pathology separately as the previous model did. To demonstrate the effectiveness of the proposed model, we compared the proposed method with a series of state-of-the-arts methods. Our model shows competitive performance and can predict potential hepatotoxicity and nephrotoxicity in a more accurate and reliable way. The implementation of the proposed method is available at https://github.com/RanSuLab/Drug-Toxicity-Prediction-MultiLabel.
Collapse
Affiliation(s)
- Ran Su
- School of Computer Science and Technology, College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Haitang Yang
- School of Computer Science and Technology, College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Leyi Wei
- School of Software, Shandong University, Jinan, Shandong, China
| | - Siqi Chen
- School of Computer Science and Technology, College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Quan Zou
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, Zhejiang, China
| |
Collapse
|
7
|
House JS, Grimm FA, Klaren WD, Dalzell A, Kuchi S, Zhang SD, Lenz K, Boogaard PJ, Ketelslegers HB, Gant TW, Rusyn I, Wright FA. Grouping of UVCB substances with dose-response transcriptomics data from human cell-based assays. ALTEX 2022; 39:388–404. [PMID: 35288757 PMCID: PMC9344966 DOI: 10.14573/altex.2107051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 02/22/2022] [Indexed: 12/18/2022]
Abstract
The application of in vitro biological assays as new approach methodologies (NAMs) to support grouping of UVCB (unknown or variable composition, complex reaction products, and biological materials) substances has recently been demonstrated. In addition to cell-based phenotyping as NAMs, in vitro transcriptomic profiling is used to gain deeper mechanistic understanding of biological responses to chemicals and to support grouping and read-across. However, the value of gene expression profiling for characterizing complex substances like UVCBs has not been explored. Using 141 petroleum substance extracts, we performed dose-response transcriptomic profiling in human induced pluripotent stem cell (iPSC)-derived hepatocytes, cardiomyocytes, neurons, and endothelial cells, as well as cell lines MCF7 and A375. The goal was to determine whether transcriptomic data can be used to group these UVCBs and to further characterize the molecular basis for in vitro biological responses. We found distinct transcriptional responses for petroleum substances by manufacturing class. Pathway enrichment informed interpretation of effects of substances and UVCB petroleum-class. Transcriptional activity was strongly correlated with concentration of polycyclic aromatic compounds (PAC), especially in iPSC-derived hepatocytes. Supervised analysis using transcriptomics, alone or in combination with bioactivity data collected on these same substances/cells, suggest that transcriptomics data provide useful mechanistic information, but only modest additional value for grouping. Overall, these results further demonstrate the value of NAMs for grouping of UVCBs, identify informative cell lines, and provide data that could be used for justifying selection of substances for further testing that may be required for registration.
Collapse
Affiliation(s)
- John S House
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA.,Biostatistics & Computational Biology Branch, National Institute of Environmental Health Sciences, RTP, NC, USA
| | - Fabian A Grimm
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - William D Klaren
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA.,current address: ToxStrategies, Inc., Asheville, NC, USA
| | - Abigail Dalzell
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Harwell Science Campus, Oxon, UK
| | - Srikeerthana Kuchi
- Northern Ireland Centre for Stratified Medicine, Ulster University, L/Derry, Northern Ireland, UK.,current address: MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, UK
| | - Shu-Dong Zhang
- Northern Ireland Centre for Stratified Medicine, Ulster University, L/Derry, Northern Ireland, UK
| | - Klaus Lenz
- SYNCOM Forschungs und Entwicklungsberatung GmbH, Ganderkesee, Germany
| | | | | | - Timothy W Gant
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Harwell Science Campus, Oxon, UK
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Fred A Wright
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
8
|
Ellison C, Hewitt M, Przybylak K. In Silico Models for Hepatotoxicity. Methods Mol Biol 2022; 2425:355-392. [PMID: 35188639 DOI: 10.1007/978-1-0716-1960-5_14] [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
In this chapter, we review the state of the art of predicting human hepatotoxicity using in silico techniques. There has been significant progress in this area over the past 20 years but there are still some challenges ahead. Principally, these challenges are our partial understanding of a very complex biochemical system and our ability to emulate that in a predictive capacity. Here, we provide an overview of the published modeling approaches in this area to date and discuss their design, strengths and weaknesses. It is interesting to note the diversity in modeling approaches, whether they be statistical algorithms or evidenced-based approaches including structural alerts and pharmacophore models. Irrespective of modeling approach, it appears a common theme of access to appropriate, relevant, and high-quality data is a limitation to all and is likely to continue to be the focus of future research.
Collapse
Affiliation(s)
- Claire Ellison
- Human and Natural Sciences Directorate, School of Science, Engineering and Environment, University of Salford, Manchester, UK
| | - Mark Hewitt
- School of Pharmacy, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, UK.
| | | |
Collapse
|
9
|
Chen X, Roberts R, Tong W, Liu Z. Tox-GAN: An AI Approach Alternative to Animal Studies-a Case Study with Toxicogenomics. Toxicol Sci 2021; 186:242-259. [PMID: 34971401 DOI: 10.1093/toxsci/kfab157] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Animal studies are a critical component in biomedical research, pharmaceutical product development, and regulatory submissions. There is a worldwide effort in toxicology towards "reducing, refining and replacing" (3Rs) animal use. Here, we proposed a deep generative adversarial network (GAN)-based framework capable of deriving new animal results from existing animal studies without additional experiments. To prove the concept, we employed this Tox-GAN framework to generate both gene activities and expression profiles for multiple doses and treatment durations in toxicogenomics (TGx). Using the pre-existing rat liver TGx data from the Open TG-GATEs, we generated Tox-GAN transcriptomic profiles with high similarity (0.997 ± 0.002 in intensity and 0.740 ± 0.082 in fold change) to the corresponding real gene expression profiles. Consequently, Tox-GAN showed an outstanding performance in two critical TGx applications, gaining a molecular understanding of underlying toxicological mechanisms and gene expression-based biomarker development. For the former, over 87% agreement in Gene Ontology was found between Tox-GAN results and real gene expression data. For the latter, the concordance of biomarkers between real and generated data was high in both predictive performance and biomarker genes. We also demonstrated that the Tox-GAN models constructed with TG-GATEs data were capable of generating transcriptomic profiles reported in DrugMatrix. Finally, we demonstrated potential utility for Tox-GAN in aiding chemical-based read-across. To the best of our knowledge, the proposed Tox-GAN model is novel in its ability to generate in vivo transcriptomic profiles at different treatment conditions from chemical structures. Overall, Tox-GAN holds great promise for generating high-quality toxicogenomic profiles without animal experimentation.
Collapse
Affiliation(s)
- Xi Chen
- National Center for Toxicological Research, Food and Drug Administration, Jefferson, Arkansas 72079, USA
| | - Ruth Roberts
- ApconiX Ltd, Alderley Edge SK10 4TG, UK
- Department of Biosciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Weida Tong
- National Center for Toxicological Research, Food and Drug Administration, Jefferson, Arkansas 72079, USA
| | - Zhichao Liu
- National Center for Toxicological Research, Food and Drug Administration, Jefferson, Arkansas 72079, USA
| |
Collapse
|
10
|
Sakhteman A, Failli M, Kublbeck J, Levonen AL, Fortino V. A toxicogenomic data space for system-level understanding and prediction of EDC-induced toxicity. ENVIRONMENT INTERNATIONAL 2021; 156:106751. [PMID: 34271427 DOI: 10.1016/j.envint.2021.106751] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 06/13/2023]
Abstract
Endocrine disrupting compounds (EDCs) are a persistent threat to humans and wildlife due to their ability to interfere with endocrine signaling pathways. Inspired by previous work to improve chemical hazard identification through the use of toxicogenomics data, we developed a genomic-oriented data space for profiling the molecular activity of EDCs in an in silico manner, and for creating predictive models that identify and prioritize EDCs. Predictive models of EDCs, derived from gene expression data from rats (in vivo and in vitro primary hepatocytes) and humans (in vitro primary hepatocytes and HepG2), achieve testing accuracy greater than 90%. Negative test sets indicate that known safer chemicals are not predicted as EDCs. The rat in vivo-based classifiers achieve accuracy greater than 75% when tested for invitro to in vivoextrapolation. This study reveals key metabolic pathways and genes affected by EDCs together with a set of predictive models that utilize these pathways to prioritize EDCs in dose/time dependent manner and to predict EDCevokedmetabolic diseases.
Collapse
Affiliation(s)
- A Sakhteman
- Institute of Biomedicine, University of Eastern Finland, Kuopio 70210, Finland
| | - M Failli
- Department of Chemical, Materials and Industrial Engineering, University of Naples, 'Federico II', Naples 80125, Italy
| | - J Kublbeck
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio 70210, Finland; School of Pharmacy, University of Eastern Finland, Kuopio 70210, Finland
| | - A L Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio 70210, Finland
| | - V Fortino
- Institute of Biomedicine, University of Eastern Finland, Kuopio 70210, Finland.
| |
Collapse
|
11
|
Vall A, Sabnis Y, Shi J, Class R, Hochreiter S, Klambauer G. The Promise of AI for DILI Prediction. Front Artif Intell 2021; 4:638410. [PMID: 33937745 PMCID: PMC8080874 DOI: 10.3389/frai.2021.638410] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 02/02/2021] [Indexed: 12/11/2022] Open
Abstract
Drug-induced liver injury (DILI) is a common reason for the withdrawal of a drug from the market. Early assessment of DILI risk is an essential part of drug development, but it is rendered challenging prior to clinical trials by the complex factors that give rise to liver damage. Artificial intelligence (AI) approaches, particularly those building on machine learning, range from random forests to more recent techniques such as deep learning, and provide tools that can analyze chemical compounds and accurately predict some of their properties based purely on their structure. This article reviews existing AI approaches to predicting DILI and elaborates on the challenges that arise from the as yet limited availability of data. Future directions are discussed focusing on rich data modalities, such as 3D spheroids, and the slow but steady increase in drugs annotated with DILI risk labels.
Collapse
Affiliation(s)
- Andreu Vall
- LIT AI Lab, Johannes Kepler University Linz, Linz, Austria.,Institute for Machine Learning, Johannes Kepler University Linz, Linz, Austria
| | | | - Jiye Shi
- UCB Biopharma SRL, Braine-l'Alleud, Belgium
| | | | - Sepp Hochreiter
- LIT AI Lab, Johannes Kepler University Linz, Linz, Austria.,Institute for Machine Learning, Johannes Kepler University Linz, Linz, Austria.,Institute of Advanced Research in Artificial Intelligence (IARAI), Vienna, Austria
| | - Günter Klambauer
- LIT AI Lab, Johannes Kepler University Linz, Linz, Austria.,Institute for Machine Learning, Johannes Kepler University Linz, Linz, Austria
| |
Collapse
|
12
|
Li D, Knox B, Gong B, Chen S, Guo L, Liu Z, Tong W, Ning B. Identification of Translational microRNA Biomarker Candidates for Ketoconazole-Induced Liver Injury Using Next-Generation Sequencing. Toxicol Sci 2021; 179:31-43. [PMID: 33078836 PMCID: PMC7855383 DOI: 10.1093/toxsci/kfaa162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Drug-induced liver injury (DILI) is a leading cause of acute liver failure. Reliable and translational biomarkers are needed for early detection of DILI. microRNAs (miRNAs) have received wide attention as a novel class of potential DILI biomarkers. However, it is unclear how DILI drugs other than acetaminophen may influence miRNA expression or which miRNAs could serve as useful biomarkers in humans. We selected ketoconazole (KCZ), a classic hepatotoxin, to study miRNA biomarkers for DILI as a proof of concept for a workflow that integrated in vivo, in vitro, and bioinformatics analyses. We examined hepatic miRNA expression in KCZ-treated rats at multiple doses and durations using miRNA-sequencing and correlated our results with conventional DILI biomarkers such as liver histology. Significant dysregulation of rno-miR-34a-5p, rno-miR-331-3p, rno-miR-15b-3p, and rno-miR-676 was associated with cytoplasmic vacuolization, a phenotype in rat livers with KCZ-induced injury, which preceded the elevation of serum liver transaminases (ALT and AST). Between rats and humans, miR-34a-5p, miR-331-3p, and miR-15b-3p were evolutionarily conserved with identical sequences, whereas miR-676 showed 73% sequence similarity. Using quantitative PCR, we found that the levels of hsa-miR-34a-5p, hsa-miR-331-3p, and hsa-miR-15b-3p were significantly elevated in the culture media of HepaRG cells treated with 100 µM KCZ (a concentration that induced cytotoxicity). Additionally, we computationally characterized the miRNA candidates for their gene targeting, target functions, and miRNA/target evolutionary conservation. In conclusion, we identified miR-34a-5p, miR-331-3p, and miR-15b-3p as translational biomarker candidates for early detection of KCZ-induced liver injury with a workflow applicable to computational toxicology studies.
Collapse
Affiliation(s)
- Dongying Li
- National Center for Toxicological Research, U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079
| | - Bridgett Knox
- National Center for Toxicological Research, U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079
| | - Binsheng Gong
- National Center for Toxicological Research, U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079
| | - Si Chen
- National Center for Toxicological Research, U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079
| | - Lei Guo
- National Center for Toxicological Research, U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079
| | - Zhichao Liu
- National Center for Toxicological Research, U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079
| | - Weida Tong
- National Center for Toxicological Research, U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079
| | - Baitang Ning
- National Center for Toxicological Research, U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079
| |
Collapse
|
13
|
Luijten M, Wackers PFK, Rorije E, Pennings JLA, Heusinkveld HJ. Relevance of In Vitro Transcriptomics for In Vivo Mode of Action Assessment. Chem Res Toxicol 2020; 34:452-459. [PMID: 33378166 DOI: 10.1021/acs.chemrestox.0c00313] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Recently, we reported an in vitro toxicogenomics comparison approach to categorize chemical substances according to similarities in their proposed toxicological modes of action. Use of such an approach for regulatory purposes requires, among others, insight into the extent of biological concordance between in vitro and in vivo findings. To that end, we applied the comparison approach to transcriptomics data from the Open TG-GATEs database for 137 substances with diverging modes of action and evaluated the outcomes obtained for rat primary hepatocytes and for rat liver. The results showed that a relatively small number of matches observed in vitro were also observed in vivo, whereas quite a large number of matches between substances were found to be relevant solely in vivo or in vitro. The latter could not be explained by physicochemical properties, leading to insufficient bioavailability or poor water solubility. Nevertheless, pathway analyses indicated that for relevant matches the mechanisms perturbed in vitro are consistent with those perturbed in vivo. These findings support the utility of the comparison approach as tool in mechanism-based risk assessment.
Collapse
Affiliation(s)
- Mirjam Luijten
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), 3720 BA Bilthoven, The Netherlands
| | - Paul F K Wackers
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), 3720 BA Bilthoven, The Netherlands
| | - Emiel Rorije
- Centre for Safety of Substances and Products, National Institute for Public Health and the Environment (RIVM), 3720 BA Bilthoven, The Netherlands
| | - Jeroen L A Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), 3720 BA Bilthoven, The Netherlands
| | - Harm J Heusinkveld
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), 3720 BA Bilthoven, The Netherlands
| |
Collapse
|
14
|
Hattori N, Takumi A, Saito K, Saito Y. Effects of serial cervical or tail blood sampling on toxicity and toxicokinetic evaluation in rats. J Toxicol Sci 2020; 45:599-609. [PMID: 33012728 DOI: 10.2131/jts.45.599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
To assess the influences of blood sampling volumes or sites on toxicological and toxicokinetic (TK) evaluations, 4-week duration animal studies and a single-dose TK study of imipramine were conducted. In the toxicological evaluation, six-week-old Sprague-Dawley rats were divided into no blood and blood sampling groups. Fifty microliters (microsampling) or 100 μL (larger sampling) of blood/time point was collected from the jugular vein (50 μL of data was reported previously as Yokoyama et al., 2020) or the tail vein 6 to 7 times on days 1/2 and in week 4. Although no parameters were affected by the 100 μL sample from the tail vein, the 100 μL jugular vein sampling decreased the red blood cell parameters in females, possibly due to hemorrhage at the sampling site. Regarding the TK assessment, 50 μL of blood/site/time point was collected at 6 time points from the tail and jugular vein of the same male rats after single oral administration of 10 or 100 mg/kg imipramine, which was selected as a representative drug with high distribution volume. Although there were no differences in the AUC0-24hr and Cmax values between the sites, the plasma concentrations at the early time points were significantly lower from the tail vein than the jugular vein. From our studies, 50 μL of jugular and tail vein microsampling did not affect the toxicity parameters or AUC/Cmax. However, appropriate toxicity considerations and/or selection of the blood sampling site may be important in the case of larger sampling volumes or blood concentration assessment.
Collapse
|
15
|
Dougherty BV, Papin JA. Systems biology approaches help to facilitate interpretation of cross-species comparisons. CURRENT OPINION IN TOXICOLOGY 2020. [DOI: 10.1016/j.cotox.2020.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
16
|
Saheb Sharif-Askari N, Saheb Sharif-Askari F, Mdkhana B, Al Heialy S, Ratemi E, Alghamdi M, Abusnana S, Kashour T, Hamid Q, Halwani R. Effect of common medications on the expression of SARS-CoV-2 entry receptors in liver tissue. Arch Toxicol 2020; 94:4037-4041. [PMID: 32808185 PMCID: PMC7430937 DOI: 10.1007/s00204-020-02869-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 08/12/2020] [Indexed: 01/08/2023]
Abstract
Besides lung drastic involvement, SARS-CoV-2 severely affected other systems including liver. Emerging epidemiological studies brought the attentions towards liver injury and impairment as a potential outcome of COVID19. Angiotensin-converting enzyme 2 (ACE2) and Transmembrane serine protease (TMPRSS2) are the main cell entry receptors of SARS-CoV-2. We have tested the ability of medications to regulate expression of SARS-CoV-2 receptors. Understanding that may reflect how such medications may affect the level of infectivity and permissibility of the liver following COVID-19. Using transcriptomic datasets, Toxicogenomic Project-Genomics Assisted Toxicity Evaluation System (Open TG-GATEs) and GSE30351, we have tested the ability of ninety common medications to regulate COVID-19 receptors expression in human primary hepatocytes. Most medications displayed a dose-dependent change in expression of receptors which could hint at a potentially more pronounced change with chronic use. The expression level of TMPRSS2 was increased noticeably with a number of medications such as metformin. Within the analgesics, acetaminophen revealed a dose-dependent reduction in expression of ACE2, while non-steroidal anti-inflammatory drugs had mixed effect on receptors expression. To confirm the observed effects on primary human hepatocytes, rat hepatocyte treatments data was obtained from DrugMatrix toxicogenomic database (GSE57805), which showed a similar ACE2 and TMPRSS2 expression pattern. Treatment of common co-morbidities often require chronic use of multiple medications, which may result in an additive increase in the expression of ACE2 and TMPRSS2. More research is needed to determine the effect of different medications on COVID-19 receptors.
Collapse
Affiliation(s)
- Narjes Saheb Sharif-Askari
- College of Medicine, Sharjah Institute of Medical Research, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Fatemeh Saheb Sharif-Askari
- College of Medicine, Sharjah Institute of Medical Research, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Bushra Mdkhana
- College of Medicine, Sharjah Institute of Medical Research, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Saba Al Heialy
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates.,Meakins-Christie Laboratories, Research Institute of the McGill University Healthy Center, McGill University, Montreal, QC, Canada
| | - Elaref Ratemi
- Department of Chemical and Process Engineering Technology, Jubail Industrial College, Jubail Industrial City, Al Jubail, Saudi Arabia
| | - Malak Alghamdi
- Department of Pediatrics, Medical Genetic Division, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Salah Abusnana
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Diabetes and Endocrinology Department, University Hospital Sharjah, Sharjah, United Arab Emirates
| | - Tarek Kashour
- Department of Cardiology, King Fahad Cardiac Center, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Qutayba Hamid
- College of Medicine, Sharjah Institute of Medical Research, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Meakins-Christie Laboratories, Research Institute of the McGill University Healthy Center, McGill University, Montreal, QC, Canada
| | - Rabih Halwani
- College of Medicine, Sharjah Institute of Medical Research, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates. .,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates. .,Prince Abdullah Ben Khaled Celiac Disease Research Chair, Department of pediatrics, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
17
|
Huang SH, Lin YC, Tung CW. Identification of Time-Invariant Biomarkers for Non-Genotoxic Hepatocarcinogen Assessment. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17124298. [PMID: 32560183 PMCID: PMC7345770 DOI: 10.3390/ijerph17124298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/12/2020] [Accepted: 06/14/2020] [Indexed: 12/12/2022]
Abstract
Non-genotoxic hepatocarcinogens (NGHCs) can only be confirmed by 2-year rodent studies. Toxicogenomics (TGx) approaches using gene expression profiles from short-term animal studies could enable early assessment of NGHCs. However, high variance in the modulation of the genes had been noted among exposure styles and datasets. Expanding from our previous strategy in identifying consensus biomarkers in multiple experiments, we aimed to identify time-invariant biomarkers for NGHCs in short-term exposure styles and validate their applicability to long-term exposure styles. In this study, nine time-invariant biomarkers, namely A2m, Akr7a3, Aqp7, Ca3, Cdc2a, Cdkn3, Cyp2c11, Ntf3, and Sds, were identified from four large-scale microarray datasets. Machine learning techniques were subsequently employed to assess the prediction performance of the biomarkers. The biomarker set along with the Random Forest models gave the highest median area under the receiver operating characteristic curve (AUC) of 0.824 and a low interquartile range (IQR) variance of 0.036 based on a leave-one-out cross-validation. The application of the models to the external validation datasets achieved high AUC values of greater than or equal to 0.857. Enrichment analysis of the biomarkers inferred the involvement of chronic inflammatory diseases such as liver cirrhosis, fibrosis, and hepatocellular carcinoma in NGHCs. The time-invariant biomarkers provided a robust alternative for NGHC prediction.
Collapse
Affiliation(s)
- Shan-Han Huang
- Ph. D. Program in Toxicology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (S.-H.H.); (Y.-C.L.)
| | - Ying-Chi Lin
- Ph. D. Program in Toxicology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (S.-H.H.); (Y.-C.L.)
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chun-Wei Tung
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taipei 11031, Taiwan
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli County 35053, Taiwan
- Correspondence:
| |
Collapse
|
18
|
Nicolaidou V, Koufaris C. Application of transcriptomic and microRNA profiling in the evaluation of potential liver carcinogens. Toxicol Ind Health 2020; 36:386-397. [PMID: 32419640 DOI: 10.1177/0748233720922710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hepatocarcinogens are agents that increase the incidence of liver cancer in exposed animals or humans. It is now established that carcinogenic exposures have a widespread impact on the transcriptome, inducing both adaptive and adverse changes in the activities of genes and pathways. Chemical hepatocarcinogens have also been shown to affect expression of microRNA (miRNA), the evolutionarily conserved noncoding RNA that regulates gene expression posttranscriptionally. Considerable effort has been invested into examining the involvement of mRNA in chemical hepatocarcinogenesis and their potential usage for the classification and prediction of new chemical entities. For miRNA, there has been an increasing number of studies reported over the past decade, although not to the same degree as for transcriptomic studies. Current data suggest that it is unlikely that any gene or miRNA signature associated with short-term carcinogen exposure can replace the rodent bioassay. In this review, we discuss the application of transcriptomic and miRNA profiles to increase mechanistic understanding of chemical carcinogens and to aid in their classification.
Collapse
Affiliation(s)
- Vicky Nicolaidou
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
| | - Costas Koufaris
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
19
|
Akahori Y, Yamashita K, Ishida K, Saito F, Nakai M. [Transcriptomics-driven Evaluation on Liver Toxicity Using Adverse Outcome Pathways (AOP)]. YAKUGAKU ZASSHI 2020; 140:491-498. [PMID: 32238630 DOI: 10.1248/yakushi.19-00190-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Because the liver is the primary target organ for chemicals and pharmaceuticals, evaluation of these substances' liver toxicity is of critical importance. New evaluation methods without animal testing (i.e., in vitro and/or in silico) are eagerly anticipated, both for animal welfare and for decreasing cost. Also, the importance of mechanistic interpretation of the output derived from non-animal testing has been increasing. Accordingly, we investigated the potential for evaluating liver toxicity by applying the adverse outcome pathway (AOP) concept using gene set enrichment analysis (GSEA) from gene expression (GEx) data. A case study targeting hepatocellular fatty degeneration (HFD) is reported and discussed. We first identified the events detectable in an in vitro system by comparing the GEx data from the rat primary hepatocyte (in vitro) and rat liver (in vivo) treated with a chemical with the ability to induce HFD as one of the phenotypes in a 28-day repeated-dose toxicity test. Then, the scores based on GSEA were calculated after establishing the gene sets for each event leading to HFD. As a result, the mechanistic information leading to HFD was obtained from the score calculated based on the GSEA and the usefulness of the transcriptome-driven evaluation using AOP was demonstrated.
Collapse
Affiliation(s)
- Yumi Akahori
- Chemicals Assessment and Research Center, Chemicals Evaluation and Research Institute (CERI)
| | - Kyousuke Yamashita
- Chemicals Assessment and Research Center, Chemicals Evaluation and Research Institute (CERI)
| | - Kazuya Ishida
- Chemicals Assessment and Research Center, Chemicals Evaluation and Research Institute (CERI)
| | - Fumiyo Saito
- Chemicals Assessment and Research Center, Chemicals Evaluation and Research Institute (CERI)
| | - Makoto Nakai
- Chemicals Assessment and Research Center, Chemicals Evaluation and Research Institute (CERI)
| |
Collapse
|
20
|
Kampa JM, Sahin M, Slopianka M, Giampà M, Bednarz H, Ernst R, Riefke B, Niehaus K, Fatangare A. Mass spectrometry imaging reveals lipid upregulation and bile acid changes indicating amitriptyline induced steatosis in a rat model. Toxicol Lett 2020; 325:43-50. [PMID: 32092452 DOI: 10.1016/j.toxlet.2020.02.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/03/2020] [Accepted: 02/14/2020] [Indexed: 12/16/2022]
Abstract
As a consequence of the detoxification process, drugs and drug related metabolites can accumulate in the liver, resulting in drug induced liver injury (DILI), which is the major cause for dose limitation. Amitriptyline, a commonly used tricyclic anti-depressant, is known to cause DILI. The mechanism of Amitriptyline induced liver injury is not yet completely understood. However, as it undergoes extensive hepatic metabolism, unraveling the molecular changes in the liver upon Amitriptyline treatment can help understand Amitriptyline's mode of toxicity. In this study, Amitriptyline treated male rat liver tissue was analyzed using Matrix Assisted Laser Desorption/Ionization-Mass Spectrometry Imaging (MALDI-MSI) to investigate the spatial abundances of Amitriptyline, lipids, and bile acids. The metabolism of Amitriptyline in liver tissue was successfully demonstrated, as the spatial distribution of Amitriptyline and its metabolites localize throughout treatment group liver samples. Several lipids appear upregulated, from which nine were identified as distinct phosphatidylcholine (PC) species. The detected bile acids were found to be lower in Amitriptyline treatment group. The combined results from histological findings, Oil Red O staining, and lipid zonation by MSI revealed lipid upregulation in the periportal area indicating drug induced macrovesicular steatosis (DIS).
Collapse
Affiliation(s)
- Judith M Kampa
- Proteome and Metabolome Research, Center for Biotechnology (CeBiTec), Faculty of Biology, Bielefeld University, Bielefeld, Germany
| | - Mikail Sahin
- Proteome and Metabolome Research, Center for Biotechnology (CeBiTec), Faculty of Biology, Bielefeld University, Bielefeld, Germany
| | - Markus Slopianka
- Metabolic Profiling and Clinical Pathology, Investigational Toxicology, Pharmaceuticals Division, Bayer AG, Berlin, Germany
| | - Marco Giampà
- Proteome and Metabolome Research, Center for Biotechnology (CeBiTec), Faculty of Biology, Bielefeld University, Bielefeld, Germany
| | - Hanna Bednarz
- Proteome and Metabolome Research, Center for Biotechnology (CeBiTec), Faculty of Biology, Bielefeld University, Bielefeld, Germany
| | - Rainer Ernst
- Metabolic Profiling and Clinical Pathology, Investigational Toxicology, Pharmaceuticals Division, Bayer AG, Berlin, Germany
| | - Bjoern Riefke
- Metabolic Profiling and Clinical Pathology, Investigational Toxicology, Pharmaceuticals Division, Bayer AG, Berlin, Germany
| | - Karsten Niehaus
- Proteome and Metabolome Research, Center for Biotechnology (CeBiTec), Faculty of Biology, Bielefeld University, Bielefeld, Germany
| | - Amol Fatangare
- Metabolic Profiling and Clinical Pathology, Investigational Toxicology, Pharmaceuticals Division, Bayer AG, Berlin, Germany.
| |
Collapse
|
21
|
Liu Y, Jing R, Wen Z, Li M. Narrowing the Gap Between In Vitro and In Vivo Genetic Profiles by Deconvoluting Toxicogenomic Data In Silico. Front Pharmacol 2020; 10:1489. [PMID: 31992983 PMCID: PMC6964707 DOI: 10.3389/fphar.2019.01489] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 11/18/2019] [Indexed: 01/09/2023] Open
Abstract
Toxicogenomics (TGx) is a powerful method to evaluate toxicity and is widely used in both in vivo and in vitro assays. For in vivo TGx, reduction, refinement, and replacement represent the unremitting pursuit of live-animal tests, but in vitro assays, as alternatives, usually demonstrate poor correlation with real in vivo assays. In living subjects, in addition to drug effects, inner-environmental reactions also affect genetic variation, and these two factors are further jointly reflected in gene abundance. Thus, finding a strategy to factorize inner-environmental factor from in vivo assays based on gene expression levels and to further utilize in vitro data to better simulate in vivo data is needed. We proposed a strategy based on post-modified non-negative matrix factorization, which can estimate the gene expression profiles and contents of major factors in samples. The applicability of the strategy was first verified, and the strategy was then utilized to simulate in vivo data by correcting in vitro data. The similarities between real in vivo data and simulated data (single-dose 0.72, repeat-doses 0.75) were higher than those observed when directly comparing real in vivo data with in vitro data (single-dose 0.56, repeat-doses 0.70). Moreover, by keeping environment-related factor, a simulation can always be generated by using in vitro data to provide potential substitutions for in vivo TGx and to reduce the launch of live-animal tests.
Collapse
Affiliation(s)
- Yuan Liu
- College of Chemistry, Sichuan University, Chengdu, China
| | - Runyu Jing
- College of Cybersecurity, Sichuan University, Chengdu, China
| | - Zhining Wen
- College of Chemistry, Sichuan University, Chengdu, China
| | - Menglong Li
- College of Chemistry, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Krewski D, Andersen ME, Tyshenko MG, Krishnan K, Hartung T, Boekelheide K, Wambaugh JF, Jones D, Whelan M, Thomas R, Yauk C, Barton-Maclaren T, Cote I. Toxicity testing in the 21st century: progress in the past decade and future perspectives. Arch Toxicol 2019; 94:1-58. [DOI: 10.1007/s00204-019-02613-4] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 11/05/2019] [Indexed: 12/19/2022]
|
23
|
Su R, Wu H, Liu X, Wei L. Predicting drug-induced hepatotoxicity based on biological feature maps and diverse classification strategies. Brief Bioinform 2019; 22:428-437. [PMID: 31838506 DOI: 10.1093/bib/bbz165] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/20/2019] [Indexed: 12/15/2022] Open
Abstract
Identifying hepatotoxicity as early as possible is significant in drug development. In this study, we developed a drug-induced hepatotoxicity prediction model taking account of both the biological context and the computational efficacy based on toxicogenomics data. Specifically, we proposed a novel gene selection algorithm considering gene's participation, named BioCB, to choose the discriminative genes and make more efficient prediction. Then instead of using the raw gene expression levels to characterize each drug, we developed a two-dimensional biological process feature pattern map to represent each drug. Then we employed two strategies to handle the maps and identify the hepatotoxicity, the direct use of maps, named Two-dim branch, and vectorization of maps, named One-dim branch. The two strategies subsequently used the deep convolutional neural networks and LightGBM as predictors, respectively. Additionally, we here for the first time proposed a stacked vectorized gene matrix, which was more predictive than the raw gene matrix. Results validated on both in vivo and in vitro data from two public data sets, the TG-GATES and DrugMatrix, show that the proposed One-dim branch outperforms the deep framework, the Two-dim branch, and has achieved high accuracy and efficiency. The implementation of the proposed method is available at https://github.com/RanSuLab/Hepatotoxicity.
Collapse
Affiliation(s)
- Ran Su
- School of Computer Software, College of Intelligence and Computing, Tianjin University, China
| | - Huichen Wu
- School of Computer Software, College of Intelligence and Computing, Tianjin University, China
| | - Xinyi Liu
- School of Computer Software, College of Intelligence and Computing, Tianjin University, China
| | - Leyi Wei
- School of Software, Shandong University, China
| |
Collapse
|
24
|
Rawls KD, Blais EM, Dougherty BV, Vinnakota KC, Pannala VR, Wallqvist A, Kolling GL, Papin JA. Genome-Scale Characterization of Toxicity-Induced Metabolic Alterations in Primary Hepatocytes. Toxicol Sci 2019; 172:279-291. [PMID: 31501904 PMCID: PMC6876259 DOI: 10.1093/toxsci/kfz197] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Context-specific GEnome-scale metabolic Network REconstructions (GENREs) provide a means to understand cellular metabolism at a deeper level of physiological detail. Here, we use transcriptomics data from chemically-exposed rat hepatocytes to constrain a GENRE of rat hepatocyte metabolism and predict biomarkers of liver toxicity using the Transcriptionally Inferred Metabolic Biomarker Response algorithm. We profiled alterations in cellular hepatocyte metabolism following in vitro exposure to four toxicants (acetaminophen, carbon tetrachloride, 2,3,7,8-tetrachlorodibenzodioxin, and trichloroethylene) for six hour. TIMBR predictions were compared with paired fresh and spent media metabolomics data from the same exposure conditions. Agreement between computational model predictions and experimental data led to the identification of specific metabolites and thus metabolic pathways associated with toxicant exposure. Here, we identified changes in the TCA metabolites citrate and alpha-ketoglutarate along with changes in carbohydrate metabolism and interruptions in ATP production and the TCA Cycle. Where predictions and experimental data disagreed, we identified testable hypotheses to reconcile differences between the model predictions and experimental data. The presented pipeline for using paired transcriptomics and metabolomics data provides a framework for interrogating multiple omics datasets to generate mechanistic insight of metabolic changes associated with toxicological responses.
Collapse
Affiliation(s)
- Kristopher D Rawls
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908
| | - Edik M Blais
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908
| | - Bonnie V Dougherty
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908
| | - Kalyan C Vinnakota
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. (HJF), Bethesda, Maryland 20817
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Maryland 21702
| | - Venkat R Pannala
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. (HJF), Bethesda, Maryland 20817
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Maryland 21702
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Maryland 21702
| | - Glynis L Kolling
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908
- Department of Medicine, Division of Infectious Diseases and International Health
| | - Jason A Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908
- Department of Medicine, Division of Infectious Diseases and International Health
- Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, Virginia 22908
| |
Collapse
|
25
|
Cárdenas-Ovando RA, Fernández-Figueroa EA, Rueda-Zárate HA, Noguez J, Rangel-Escareño C. A feature selection strategy for gene expression time series experiments with hidden Markov models. PLoS One 2019; 14:e0223183. [PMID: 31600242 PMCID: PMC6786538 DOI: 10.1371/journal.pone.0223183] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 09/16/2019] [Indexed: 01/11/2023] Open
Abstract
Studies conducted in time series could be far more informative than those that only capture a specific moment in time. However, when it comes to transcriptomic data, time points are sparse creating the need for a constant search for methods capable of extracting information out of experiments of this kind. We propose a feature selection algorithm embedded in a hidden Markov model applied to gene expression time course data on either single or even multiple biological conditions. For the latter, in a simple case-control study features or genes are selected under the assumption of no change over time for the control samples, while the case group must have at least one change. The proposed model reduces the feature space according to a two-state hidden Markov model. The two states define change/no-change in gene expression. Features are ranked in consonance with three scores: number of changes across time, magnitude of such changes and quality of replicates as a measure of how much they deviate from the mean. An important highlight is that this strategy overcomes the few samples limitation, common in transcriptome experiments through a process of data transformation and rearrangement. To prove this method, our strategy was applied to three publicly available data sets. Results show that feature domain is reduced by up to 90% leaving only few but relevant features yet with findings consistent to those previously reported. Moreover, our strategy proved to be robust, stable and working on studies where sample size is an issue otherwise. Hence, even with two biological replicates and/or three time points our method proves to work well.
Collapse
Affiliation(s)
- Roberto A. Cárdenas-Ovando
- School of Engineering and Sciences, Tecnológico de Monterrey, Mexico City, Mexico
- Computational Genomics Lab, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | | | - Héctor A. Rueda-Zárate
- School of Engineering and Sciences, Tecnológico de Monterrey, Mexico City, Mexico
- Computational Genomics Lab, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Julieta Noguez
- School of Engineering and Sciences, Tecnológico de Monterrey, Mexico City, Mexico
| | - Claudia Rangel-Escareño
- Computational Genomics Lab, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
- * E-mail:
| |
Collapse
|
26
|
Assessment of Drugs Toxicity and Associated Biomarker Genes Using Hierarchical Clustering. ACTA ACUST UNITED AC 2019; 55:medicina55080451. [PMID: 31398888 PMCID: PMC6723056 DOI: 10.3390/medicina55080451] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/04/2019] [Accepted: 08/06/2019] [Indexed: 12/13/2022]
Abstract
Background and objectives: Assessment of drugs toxicity and associated biomarker genes is one of the most important tasks in the pre-clinical phase of drug development pipeline as well as in toxicogenomic studies. There are few statistical methods for the assessment of doses of drugs (DDs) toxicity and their associated biomarker genes. However, these methods consume more time for computation of the model parameters using the EM (expectation-maximization) based iterative approaches. To overcome this problem, in this paper, an attempt is made to propose an alternative approach based on hierarchical clustering (HC) for the same purpose. Methods and materials: There are several types of HC approaches whose performance depends on different similarity/distance measures. Therefore, we explored suitable combinations of distance measures and HC methods based on Japanese Toxicogenomics Project (TGP) datasets for better clustering/co-clustering between DDs and genes as well as to detect toxic DDs and their associated biomarker genes. Results: We observed that Word’s HC method with each of Euclidean, Manhattan, and Minkowski distance measures produces better clustering/co-clustering results. For an example, in the case of the glutathione metabolism pathway (GMP) dataset LOC100359539/Rrm2, Gpx6, RGD1562107, Gstm4, Gstm3, G6pd, Gsta5, Gclc, Mgst2, Gsr, Gpx2, Gclm, Gstp1, LOC100912604/Srm, Gstm4, Odc1, Gsr, Gss are the biomarker genes and Acetaminophen_Middle, Acetaminophen_High, Methapyrilene_High, Nitrofurazone_High, Nitrofurazone_Middle, Isoniazid_Middle, Isoniazid_High are their regulatory (associated) DDs explored by our proposed co-clustering algorithm based on the distance and HC method combination Euclidean: Word. Similarly, for the peroxisome proliferator-activated receptor signaling pathway (PPAR-SP) dataset Cpt1a, Cyp8b1, Cyp4a3, Ehhadh, Plin5, Plin2, Fabp3, Me1, Fabp5, LOC100910385, Cpt2, Acaa1a, Cyp4a1, LOC100365047, Cpt1a, LOC100365047, Angptl4, Aqp7, Cpt1c, Cpt1b, Me1 are the biomarker genes and Aspirin_Low, Aspirin_Middle, Aspirin_High, Benzbromarone_Middle, Benzbromarone_High, Clofibrate_Middle, Clofibrate_High, WY14643_Low, WY14643_High, WY14643_Middle, Gemfibrozil_Middle, Gemfibrozil_High are their regulatory DDs. Conclusions: Overall, the methods proposed in this article, co-cluster the genes and DDs as well as detect biomarker genes and their regulatory DDs simultaneously consuming less time compared to other mentioned methods. The results produced by the proposed methods have been validated by the available literature and functional annotation.
Collapse
|
27
|
Cho E, Buick JK, Williams A, Chen R, Li H, Corton JC, Fornace AJ, Aubrecht J, Yauk CL. Assessment of the performance of the TGx-DDI biomarker to detect DNA damage-inducing agents using quantitative RT-PCR in TK6 cells. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2019; 60:122-133. [PMID: 30488505 PMCID: PMC6588084 DOI: 10.1002/em.22257] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 05/05/2023]
Abstract
Gene expression biomarkers are now available for application in the identification of genotoxic hazards. The TGx-DDI transcriptomic biomarker can accurately distinguish DNA damage-inducing (DDI) from non-DDI exposures based on changes in the expression of 64 biomarker genes. The 64 genes were previously derived from whole transcriptome DNA microarray profiles of 28 reference agents (14 DDI and 14 non-DDI) after 4 h treatments of TK6 human lymphoblastoid cells. To broaden the applicability of TGx-DDI, we tested the biomarker using quantitative RT-PCR (qPCR), which is accessible to most molecular biology laboratories. First, we selectively profiled the expression of the 64 biomarker genes using TaqMan qPCR assays in 96-well arrays after exposing TK6 cells to the 28 reference agents for 4 h. To evaluate the classification capability of the qPCR profiles, we used the reference qPCR signature to classify 24 external validation chemicals using two different methods-a combination of three statistical analyses and an alternative, the Running Fisher test. The qPCR results for the reference set were comparable to the original microarray biomarker; 27 of the 28 reference agents (96%) were accurately classified. Moreover, the two classification approaches supported the conservation of TGx-DDI classification capability using qPCR; the combination of the two approaches accurately classified 21 of the 24 external validation chemicals, demonstrating 100% sensitivity, 81% specificity, and 91% balanced accuracy. This study demonstrates that qPCR can be used when applying the TGx-DDI biomarker and will improve the accessibility of TGx-DDI for genotoxicity screening. Environ. Mol. Mutagen. 60: 122-133, 2019. © 2018 Her Majesty the Queen in Right of Canada Environmental and Molecular Mutagenesis.
Collapse
Affiliation(s)
- Eunnara Cho
- Environmental Health Science and Research BureauHealth CanadaOttawaOntarioCanada
- Department of BiologyCarleton UniversityOttawaOntarioCanada
| | - Julie K. Buick
- Environmental Health Science and Research BureauHealth CanadaOttawaOntarioCanada
| | - Andrew Williams
- Environmental Health Science and Research BureauHealth CanadaOttawaOntarioCanada
| | - Renxiang Chen
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Heng‐Hong Li
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDistrict of Columbia
- Department of Biochemistry and Molecular and Cellular BiologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | | | - Albert J. Fornace
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDistrict of Columbia
- Department of Biochemistry and Molecular and Cellular BiologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Jiri Aubrecht
- Takeda Pharmaceuticals USA Inc.CambridgeMassachusetts
| | - Carole L. Yauk
- Environmental Health Science and Research BureauHealth CanadaOttawaOntarioCanada
- Department of BiologyCarleton UniversityOttawaOntarioCanada
| |
Collapse
|
28
|
Taškova K, Fontaine JF, Mrowka R, Andrade-Navarro MA. Literature optimized integration of gene expression for organ-specific evaluation of toxicogenomics datasets. PLoS One 2019; 14:e0210467. [PMID: 30640953 PMCID: PMC6331104 DOI: 10.1371/journal.pone.0210467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 12/24/2018] [Indexed: 11/30/2022] Open
Abstract
The study of drug toxicity in human organs is complicated by their complex inter-relations and by the obvious difficulty to testing drug effects on biologically relevant material. Animal models and human cell cultures offer alternatives for systematic and large-scale profiling of drug effects on gene expression level, as typically found in the so-called toxicogenomics datasets. However, the complexity of these data, which includes variable drug doses, time points, and experimental setups, makes it difficult to choose and integrate the data, and to evaluate the appropriateness of one or another model system to study drug toxicity (of particular drugs) of particular human organs. Here, we define a protocol to integrate drug-wise rankings of gene expression changes in toxicogenomics data, which we apply to the TG-GATEs dataset, to prioritize genes for association to drug toxicity in liver or kidney. Contrast of the results with sets of known human genes associated to drug toxicity in the literature allows to compare different rank aggregation approaches for the task at hand. Collectively, ranks from multiple models point to genes not previously associated to toxicity, notably, the PCNA clamp associated factor (PCLAF), and genes regulated by the master regulator of the antioxidant response NFE2L2, such as NQO1 and SRXN1. In addition, comparing gene ranks from different models allowed us to evaluate striking differences in terms of toxicity-associated genes between human and rat hepatocytes or between rat liver and rat hepatocytes. We interpret these results to point to the different molecular functions associated to organ toxicity that are best described by each model. We conclude that the expected production of toxicogenomics panels with larger numbers of drugs and models, in combination with the ongoing increase of the experimental literature in organ toxicity, will lead to increasingly better associations of genes for organism toxicity.
Collapse
Affiliation(s)
| | | | - Ralf Mrowka
- Experimentelle Nephrologie, Universitätsklinikum Jena, KIM III, Jena, Germany
| | | |
Collapse
|
29
|
Biomarker assay validation for clinical trials: a questionnaire survey to pharmaceutical companies in Japan. Bioanalysis 2019; 11:55-60. [DOI: 10.4155/bio-2018-0257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
30
|
Hasan MN, Begum AA, Rahman M, Haque Mollah MN. Robust identification of significant interactions between toxicogenomic biomarkers and their regulatory chemical compounds using logistic moving range chart. Comput Biol Chem 2018; 78:375-381. [PMID: 30606695 DOI: 10.1016/j.compbiolchem.2018.12.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 12/25/2018] [Indexed: 10/27/2022]
Abstract
Identification of significant interactions between genes and chemical compounds/drugs is an important issue in toxicogenomic studies as well as in drug discovery and development. There are some online and offline computational tools for toxicogenomic data analysis to identify the biomarker genes and their regulatory chemical compounds/drugs. However, none of the researchers has considered yet the identification of significant interactions between genes and compounds. Therefore, in this paper, we have discussed two approaches namely moving range chart (MRC) and logistic moving range chart (LMRC) for the identification of significant up-regulatory (UpR) and down-regulatory (DnR) gene-compound interactions as well as toxicogenomic biomarkers and their regulatory chemical compounds/drugs. We have investigated the performance of both MRC and LMRC approaches using simulated datasets. Simulation results show that both approaches perform almost equally in absence of outliers. However, in presence of outliers, the LMRC shows much better performance than the MRC. In case of real life toxicogenomic data analysis, the proposed LMRC approach detected some important down-regulated biomarker genes those were not detected by other approaches. Therefore, in this paper, our proposal is to use LMRC for robust identification of significant interactions between genes and chemical compounds/drugs.
Collapse
Affiliation(s)
- Mohammad Nazmol Hasan
- Bioinformatics Lab., Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh; Department of Statistics, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh.
| | - Anjuman Ara Begum
- Bioinformatics Lab., Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Moizur Rahman
- Department of Veterinary and Animal Sciences, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md Nurul Haque Mollah
- Bioinformatics Lab., Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh.
| |
Collapse
|
31
|
Hasan MN, Rana MM, Begum AA, Rahman M, Mollah MNH. Robust Co-clustering to Discover Toxicogenomic Biomarkers and Their Regulatory Doses of Chemical Compounds Using Logistic Probabilistic Hidden Variable Model. Front Genet 2018; 9:516. [PMID: 30450112 PMCID: PMC6225736 DOI: 10.3389/fgene.2018.00516] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/12/2018] [Indexed: 11/13/2022] Open
Abstract
Detection of biomarker genes and their regulatory doses of chemical compounds (DCCs) is one of the most important tasks in toxicogenomic studies as well as in drug design and development. There is an online computational platform "Toxygates" to identify biomarker genes and their regulatory DCCs by co-clustering approach. Nevertheless, the algorithm of that platform based on hierarchical clustering (HC) does not share gene-DCC two-way information simultaneously during co-clustering between genes and DCCs. Also it is sensitive to outlying observations. Thus, this platform may produce misleading results in some cases. The probabilistic hidden variable model (PHVM) is a more effective co-clustering approach that share two-way information simultaneously, but it is also sensitive to outlying observations. Therefore, in this paper we have proposed logistic probabilistic hidden variable model (LPHVM) for robust co-clustering between genes and DCCs, since gene expression data are often contaminated by outlying observations. We have investigated the performance of the proposed LPHVM co-clustering approach in a comparison with the conventional PHVM and Toxygates co-clustering approaches using simulated and real life TGP gene expression datasets, respectively. Simulation results show that the proposed method improved the performance over the conventional PHVM in presence of outliers; otherwise, it keeps equal performance. In the case of real life TGP data analysis, three DCCs (glibenclamide-low, perhexilline-low, and hexachlorobenzene-medium) for glutathione metabolism pathway dataset as well as two DCCs (acetaminophen-medium and methapyrilene-low) for PPAR signaling pathway dataset were incorrectly co-clustered by the Toxygates online platform, while only one DCC (hexachlorobenzene-low) for glutathione metabolism pathway was incorrectly co-clustered by the proposed LPHVM approach. Our findings from the real data analysis are also supported by the other findings in the literature.
Collapse
Affiliation(s)
- Mohammad Nazmol Hasan
- Bioinformatics Laboratory, Department of Statistics, University of Rajshahi, Rajshahi, Bangladesh.,Department of Statistics, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
| | - Md Masud Rana
- Bioinformatics Laboratory, Department of Statistics, University of Rajshahi, Rajshahi, Bangladesh
| | - Anjuman Ara Begum
- Bioinformatics Laboratory, Department of Statistics, University of Rajshahi, Rajshahi, Bangladesh
| | - Moizur Rahman
- Department of Veterinary and Animal Sciences, University of Rajshahi, Rajshahi, Bangladesh
| | - Md Nurul Haque Mollah
- Bioinformatics Laboratory, Department of Statistics, University of Rajshahi, Rajshahi, Bangladesh
| |
Collapse
|
32
|
Barel G, Herwig R. Network and Pathway Analysis of Toxicogenomics Data. Front Genet 2018; 9:484. [PMID: 30405693 PMCID: PMC6204403 DOI: 10.3389/fgene.2018.00484] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/28/2018] [Indexed: 12/20/2022] Open
Abstract
Toxicogenomics is the study of the molecular effects of chemical, biological and physical agents in biological systems, with the aim of elucidating toxicological mechanisms, building predictive models and improving diagnostics. The vast majority of toxicogenomics data has been generated at the transcriptome level, including RNA-seq and microarrays, and large quantities of drug-treatment data have been made publicly available through databases and repositories. Besides the identification of differentially expressed genes (DEGs) from case-control studies or drug treatment time series studies, bioinformatics methods have emerged that infer gene expression data at the molecular network and pathway level in order to reveal mechanistic information. In this work we describe different resources and tools that have been developed by us and others that relate gene expression measurements with known pathway information such as over-representation and gene set enrichment analyses. Furthermore, we highlight approaches that integrate gene expression data with molecular interaction networks in order to derive network modules related to drug toxicity. We describe the two main parts of the approach, i.e., the construction of a suitable molecular interaction network as well as the conduction of network propagation of the experimental data through the interaction network. In all cases we apply methods and tools to publicly available rat in vivo data on anthracyclines, an important class of anti-cancer drugs that are known to induce severe cardiotoxicity in patients. We report the results and functional implications achieved for four anthracyclines (doxorubicin, epirubicin, idarubicin, and daunorubicin) and compare the information content inherent in the different computational approaches.
Collapse
Affiliation(s)
| | - Ralf Herwig
- Department Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| |
Collapse
|
33
|
Darde TA, Chalmel F, Svingen T. Exploiting advances in transcriptomics to improve on human-relevant toxicology. CURRENT OPINION IN TOXICOLOGY 2018. [DOI: 10.1016/j.cotox.2019.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
34
|
Auerbach SS, Paules RS. Genomic dose response: Successes, challenges, and next steps. CURRENT OPINION IN TOXICOLOGY 2018. [DOI: 10.1016/j.cotox.2019.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
35
|
Liu Z, Delavan B, Roberts R, Tong W. Transcriptional Responses Reveal Similarities Between Preclinical Rat Liver Testing Systems. Front Genet 2018; 9:74. [PMID: 29616076 PMCID: PMC5870427 DOI: 10.3389/fgene.2018.00074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 02/19/2018] [Indexed: 01/03/2023] Open
Abstract
Toxicogenomics (TGx) is an important tool to gain an enhanced understanding of toxicity at the molecular level. Previously, we developed a pair ranking (PRank) method to assess in vitro to in vivo extrapolation (IVIVE) using toxicogenomic datasets from the Open Toxicogenomics Project-Genomics Assisted Toxicity Evaluation System (TG-GATEs) database. With this method, we investiagted three important questions that were not addressed in our previous study: (1) is a 1-day in vivo short-term assay able to replace the 28-day standard and expensive toxicological assay? (2) are some biological processes more conservative across different preclinical testing systems than others? and (3) do these preclinical testing systems have the similar resolution in differentiating drugs by their therapeutic uses? For question 1, a high similarity was noted (PRank score = 0.90), indicating the potential utility of shorter term in vivo studies to predict outcome in longer term and more expensive in vivo model systems. There was a moderate similarity between rat primary hepatocytes and in vivo repeat-dose studies (PRank score = 0.71) but a low similarity (PRank score = 0.56) between rat primary hepatocytes and in vivo single dose studies. To address question 2, we limited the analysis to gene sets relevant to specific toxicogenomic pathways and we found that pathways such as lipid metabolism were consistently over-represented in all three assay systems. For question 3, all three preclinical assay systems could distinguish compounds from different therapeutic categories. This suggests that any noted differences in assay systems was biological process-dependent and furthermore that all three systems have utility in assessing drug responses within a certain drug class. In conclusion, this comparison of three commonly used rat TGx systems provides useful information in utility and application of TGx assays.
Collapse
Affiliation(s)
- Zhichao Liu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States
| | - Brian Delavan
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States.,Department of Biosciences, University of Arkansas at Little Rock, Little Rock, AR, United States
| | - Ruth Roberts
- ApconiX, Alderley Edge, United Kingdom.,Department of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Weida Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States
| |
Collapse
|
36
|
Venkatratnam A, House JS, Konganti K, McKenney C, Threadgill DW, Chiu WA, Aylor DL, Wright FA, Rusyn I. Population-based dose-response analysis of liver transcriptional response to trichloroethylene in mouse. Mamm Genome 2018; 29:168-181. [PMID: 29353386 DOI: 10.1007/s00335-018-9734-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 01/17/2018] [Indexed: 12/23/2022]
Abstract
Studies of gene expression are common in toxicology and provide important clues to mechanistic understanding of adverse effects of chemicals. Most prior studies have been performed in a single strain or cell line; however, gene expression is heavily influenced by the genetic background, and these genotype-expression differences may be key drivers of inter-individual variation in response to chemical toxicity. In this study, we hypothesized that the genetically diverse Collaborative Cross mouse population can be used to gain insight and suggest mechanistic hypotheses for the dose- and genetic background-dependent effects of chemical exposure. This hypothesis was tested using a model liver toxicant trichloroethylene (TCE). Liver transcriptional responses to TCE exposure were evaluated 24 h after dosing. Transcriptomic dose-responses were examined for both TCE and its major oxidative metabolite trichloroacetic acid (TCA). As expected, peroxisome- and fatty acid metabolism-related pathways were among the most dose-responsive enriched pathways in all strains. However, nearly half of the TCE-induced liver transcriptional perturbation was strain-dependent, with abundant evidence of strain/dose interaction, including in the peroxisomal signaling-associated pathways. These effects were highly concordant between the administered TCE dose and liver levels of TCA. Dose-response analysis of gene expression at the pathway level yielded points of departure similar to those derived from the traditional toxicology studies for both non-cancer and cancer effects. Mapping of expression-genotype-dose relationships revealed some significant associations; however, the effects of TCE on gene expression in liver appear to be highly polygenic traits that are challenging to positionally map. This study highlights the usefulness of mouse population-based studies in assessing inter-individual variation in toxicological responses, but cautions that genetic mapping may be challenging because of the complexity in gene exposure-dose relationships.
Collapse
Affiliation(s)
- Abhishek Venkatratnam
- Department of Veterinary Integrative Biosciences, Texas A&M University, 4458 TAMU, College Station, Texas, 77843, USA.,Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina, 27599, USA
| | - John S House
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, 27695, USA.,Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina, 27695, USA
| | - Kranti Konganti
- Department of Veterinary Integrative Biosciences, Texas A&M University, 4458 TAMU, College Station, Texas, 77843, USA
| | - Connor McKenney
- NCSU Undergraduate program in Genetics, North Carolina State University, Raleigh, North Carolina, 27695, USA
| | - David W Threadgill
- Department of Veterinary Integrative Biosciences, Texas A&M University, 4458 TAMU, College Station, Texas, 77843, USA
| | - Weihsueh A Chiu
- Department of Veterinary Integrative Biosciences, Texas A&M University, 4458 TAMU, College Station, Texas, 77843, USA
| | - David L Aylor
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, 27695, USA.,Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina, 27695, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, 27695, USA
| | - Fred A Wright
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, 27695, USA.,Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina, 27695, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, 27695, USA.,Department of Statistics, North Carolina State University, Raleigh, North Carolina, 27695, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, 4458 TAMU, College Station, Texas, 77843, USA.
| |
Collapse
|
37
|
The use of omics-based approaches in regulatory toxicology: an alternative approach to assess the no observed transcriptional effect level. Microchem J 2018. [DOI: 10.1016/j.microc.2017.01.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
38
|
Hardt C, Bauer C, Schuchhardt J, Herwig R. Computational Network Analysis for Drug Toxicity Prediction. Methods Mol Biol 2018; 1819:335-355. [PMID: 30421412 DOI: 10.1007/978-1-4939-8618-7_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The computational prediction of compound effects from molecular data is an important task in hazard and risk assessment and pivotal for judging the safety of any drug, chemical or cosmetic compound. In particular, the identification of such compound effects at the level of molecular interaction networks can be helpful for the construction of adverse outcome pathways (AOPs). AOPs emerged as a guiding concept for toxicity prediction, because of the inherent mechanistic information of such networks. In fact, integrating molecular interactions in transcriptome analysis and observing expression changes in closely interacting genes might allow identifying the key molecular initiating events of compound toxicity.In this work we describe a computational approach that is suitable for the identification of such network modules from transcriptomics data, which is the major molecular readout of toxicogenomics studies. The approach is composed of different tools (1) for primary data analysis, i.e., the biostatistical quantification of the gene expression changes, (2) for functional annotation and prioritization of genes using literature mining, as well as (3) for the construction of an interaction network that consists of interactions with high confidence and the identification of predictive modules from these networks. We describe the different steps of the approach and demonstrate its performance with public data on drugs that induce hepatic and cardiac toxicity.
Collapse
Affiliation(s)
- C Hardt
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Ihnestr. 73, D-14195, Berlin, Germany
| | - C Bauer
- MicroDiscovery GmbH, Marienburgerstr. 1, D-10405, Berlin, Germany
| | - J Schuchhardt
- MicroDiscovery GmbH, Marienburgerstr. 1, D-10405, Berlin, Germany
| | - R Herwig
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Ihnestr. 73, D-14195, Berlin, Germany.
| |
Collapse
|
39
|
Kuna L, Božić I, Kizivat T, Bojanić K, Mršo M, Kralj E, Smolić R, Wu GY, Smolić M. Models of Drug Induced Liver Injury (DILI) - Current Issues and Future Perspectives. Curr Drug Metab 2018; 19:830-838. [PMID: 29788883 PMCID: PMC6174638 DOI: 10.2174/1389200219666180523095355] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/20/2018] [Accepted: 03/28/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Drug-induced Liver Injury (DILI) is an important cause of acute liver failure cases in the United States, and remains a common cause of withdrawal of drugs in both preclinical and clinical phases. METHODS A structured search of bibliographic databases - Web of Science Core Collection, Scopus and Medline for peer-reviewed articles on models of DILI was performed. The reference lists of relevant studies was prepared and a citation search for the included studies was carried out. In addition, the characteristics of screened studies were described. RESULTS One hundred and six articles about the existing knowledge of appropriate models to study DILI in vitro and in vivo with special focus on hepatic cell models, variations of 3D co-cultures, animal models, databases and predictive modeling and translational biomarkers developed to understand the mechanisms and pathophysiology of DILI are described. CONCLUSION Besides descriptions of current applications of existing modeling systems, associated advantages and limitations of each modeling system and future directions for research development are discussed as well.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Martina Smolić
- Address correspondence to this author at the J. J. Strossmayer University of Osijek, Faculty of Medicine Osijek, Department of Pharmacology, J. Huttlera 4, 31 000 Osijek, Croatia; Tel: + 0385-31-512-800; Fax: +385-31-512-833; E-mail:
| |
Collapse
|
40
|
Thakkar S, Chen M, Fang H, Liu Z, Roberts R, Tong W. The Liver Toxicity Knowledge Base (LKTB) and drug-induced liver injury (DILI) classification for assessment of human liver injury. Expert Rev Gastroenterol Hepatol 2018; 12:31-38. [PMID: 28931315 DOI: 10.1080/17474124.2018.1383154] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Drug-induced liver injury (DILI) is challenging for drug development, clinical practice and regulation. The Liver Toxicity Knowledge Base (LTKB) provides essential data for DILI study. Areas covered: The LTKB provided various types of data that can be used to assess and predict DILI. Among much information available, several reference drug lists with annotated human DILI risk are of important. The LTKB DILI classification data include DILI severity concern determined by the FDA drug labeling, DILI severity score from the NIH LiverTox database, and other DILI classification schemes from the literature. Overall, ~1000 drugs were annotated with at least one classification scheme, of which around 750 drugs were flagged for some degree of DILI risk. Expert commentary: The LTKB provides a centralized repository of information for DILI study and predictive model development. The DILI classification data in LTKB could be a useful resource for developing biomarkers, predictive models and assessing data from emerging technologies such as in silico, high-throughput and high-content screening methodologies. In coming years, streamlining the prediction process by including DILI predictive models for both DILI severity and types in LTKB would enhance the identification of compounds with the DILI potential earlier in drug development and risk assessment.
Collapse
Affiliation(s)
- Shraddha Thakkar
- a Division of Bioinformatics and Biostatistics , National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson , AR , USA
| | - Minjun Chen
- a Division of Bioinformatics and Biostatistics , National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson , AR , USA
| | - Hong Fang
- b Office of Scientific Coordination , National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson , AR , USA
| | - Zhichao Liu
- a Division of Bioinformatics and Biostatistics , National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson , AR , USA
| | - Ruth Roberts
- c ApconiX Ltd , Alderley Edge , UK.,d School of Biosciences, University of Birmingham , Birmingham , UK
| | - Weida Tong
- a Division of Bioinformatics and Biostatistics , National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson , AR , USA
| |
Collapse
|
41
|
Sier JH, Thumser AE, Plant NJ. Linking physiologically-based pharmacokinetic and genome-scale metabolic networks to understand estradiol biology. BMC SYSTEMS BIOLOGY 2017; 11:141. [PMID: 29246152 PMCID: PMC5732473 DOI: 10.1186/s12918-017-0520-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 11/28/2017] [Indexed: 11/16/2022]
Abstract
Background Estrogen is a vital hormone that regulates many biological functions within the body. These include roles in the development of the secondary sexual organs in both sexes, plus uterine angiogenesis and proliferation during the menstrual cycle and pregnancy in women. The varied biological roles of estrogens in human health also make them a therapeutic target for contraception, mitigation of the adverse effects of the menopause, and treatment of estrogen-responsive tumours. In addition, endogenous (e.g. genetic variation) and external (e.g. exposure to estrogen-like chemicals) factors are known to impact estrogen biology. To understand how these multiple factors interact to determine an individual’s response to therapy is complex, and may be best approached through a systems approach. Methods We present a physiologically-based pharmacokinetic model (PBPK) of estradiol, and validate it against plasma kinetics in humans following intravenous and oral exposure. We extend this model by replacing the intrinsic clearance term with: a detailed kinetic model of estrogen metabolism in the liver; or, a genome-scale model of liver metabolism. Both models were validated by their ability to reproduce clinical data on estradiol exposure. We hypothesise that the enhanced mechanistic information contained within these models will lead to more robust predictions of the biological phenotype that emerges from the complex interactions between estrogens and the body. Results To demonstrate the utility of these models we examine the known drug-drug interactions between phenytoin and oral estradiol. We are able to reproduce the approximate 50% reduction in area under the concentration-time curve for estradiol associated with this interaction. Importantly, the inclusion of a genome-scale metabolic model allows the prediction of this interaction without directly specifying it within the model. In addition, we predict that PXR activation by drugs results in an enhanced ability of the liver to excrete glucose. This has important implications for the relationship between drug treatment and metabolic syndrome. Conclusions We demonstrate how the novel coupling of PBPK models with genome-scale metabolic networks has the potential to aid prediction of drug action, including both drug-drug interactions and changes to the metabolic landscape that may predispose an individual to disease development. Electronic supplementary material The online version of this article (10.1186/s12918-017-0520-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joanna H Sier
- School of Food Science and Nutrition, Faculty of Mathematics and Physical Sciences, University of Leeds, Leeds, LS2 9JT, UK.,School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Alfred E Thumser
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Nick J Plant
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK. .,School of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
42
|
House JS, Grimm FA, Jima DD, Zhou YH, Rusyn I, Wright FA. A Pipeline for High-Throughput Concentration Response Modeling of Gene Expression for Toxicogenomics. Front Genet 2017; 8:168. [PMID: 29163636 PMCID: PMC5672545 DOI: 10.3389/fgene.2017.00168] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 10/18/2017] [Indexed: 12/21/2022] Open
Abstract
Cell-based assays are an attractive option to measure gene expression response to exposure, but the cost of whole-transcriptome RNA sequencing has been a barrier to the use of gene expression profiling for in vitro toxicity screening. In addition, standard RNA sequencing adds variability due to variable transcript length and amplification. Targeted probe-sequencing technologies such as TempO-Seq, with transcriptomic representation that can vary from hundreds of genes to the entire transcriptome, may reduce some components of variation. Analyses of high-throughput toxicogenomics data require renewed attention to read-calling algorithms and simplified dose–response modeling for datasets with relatively few samples. Using data from induced pluripotent stem cell-derived cardiomyocytes treated with chemicals at varying concentrations, we describe here and make available a pipeline for handling expression data generated by TempO-Seq to align reads, clean and normalize raw count data, identify differentially expressed genes, and calculate transcriptomic concentration–response points of departure. The methods are extensible to other forms of concentration–response gene-expression data, and we discuss the utility of the methods for assessing variation in susceptibility and the diseased cellular state.
Collapse
Affiliation(s)
- John S House
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, United States.,Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States
| | - Fabian A Grimm
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, United States
| | - Dereje D Jima
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, United States.,Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States
| | - Yi-Hui Zhou
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, United States.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, United States
| | - Fred A Wright
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, United States.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States.,Department of Statistics, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
43
|
Barh D, García-Solano ME, Tiwari S, Bhattacharya A, Jain N, Torres-Moreno D, Ferri B, Silva A, Azevedo V, Ghosh P, Blum K, Conesa-Zamora P, Perry G. BARHL1 Is Downregulated in Alzheimer's Disease and May Regulate Cognitive Functions through ESR1 and Multiple Pathways. Genes (Basel) 2017; 8:genes8100245. [PMID: 28956815 PMCID: PMC5664095 DOI: 10.3390/genes8100245] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/13/2017] [Accepted: 09/20/2017] [Indexed: 12/22/2022] Open
Abstract
The Transcription factor BarH like homeobox 1 (BARHL1) is overexpressed in medulloblastoma and plays a role in neurogenesis. However, much about the BARHL1 regulatory networks and their functions in neurodegenerative and neoplastic disorders is not yet known. In this study, using a tissue microarray (TMA), we report for the first time that BARHL1 is downregulated in hormone-negative breast cancers and Alzheimer’s disease (AD). Furthermore, using an integrative bioinformatics approach and mining knockout mouse data, we show that: (i) BARHL1 and Estrogen Receptor 1 (ESR1) may constitute a network that regulates Neurotrophin 3 (NTF3)- and Brain Derived Neurotrophic Factor (BDNF)-mediated neurogenesis and neural survival; (ii) this is probably linked to AD pathways affecting aberrant post-translational modifications including SUMOylation and ubiquitination; (iii) the BARHL1-ESR1 network possibly regulates β-amyloid metabolism and memory; and (iv) hsa-mir-18a, having common key targets in the BARHL1-ESR1 network and AD pathway, may modulate neuron death, reduce β-amyloid processing and might also be involved in hearing and cognitive decline associated with AD. We have also hypothesized why estrogen replacement therapy improves AD condition. In addition, we have provided a feasible new mechanism to explain the abnormal function of mossy fibers and cerebellar granule cells related to memory and cognitive decline in AD apart from the Tau and amyloid pathogenesis through our BARHL1-ESR1 axis.
Collapse
Affiliation(s)
- Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur, West Bengal 721172, India.
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil.
| | - María E García-Solano
- Department of Pathology, Santa Lucía General University Hospital (HGUSL), C/Mezquita s/n, 30202 Cartagena, Spain.
- Catholic University of Murcia (UCAM), 30107 Murcia, Spain.
| | - Sandeep Tiwari
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur, West Bengal 721172, India.
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil.
| | - Antaripa Bhattacharya
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur, West Bengal 721172, India.
| | - Neha Jain
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur, West Bengal 721172, India.
| | - Daniel Torres-Moreno
- Department of Pathology, Santa Lucía General University Hospital (HGUSL), C/Mezquita s/n, 30202 Cartagena, Spain.
- Catholic University of Murcia (UCAM), 30107 Murcia, Spain.
| | - Belén Ferri
- Department of Pathology, Virgen Arrixaca University Hospital (HUVA), Ctra. Madrid Cartagena sn, 30120 El Palmar, Spain.
| | - Artur Silva
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Rua Augusto Corrêa, 01-Guamá, Belém, PA 66075-110, Brazil.
| | - Vasco Azevedo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil.
| | - Preetam Ghosh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur, West Bengal 721172, India.
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA.
| | - Kenneth Blum
- Department of Psychiatry & McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL 32610, USA.
| | - Pablo Conesa-Zamora
- Department of Pathology, Santa Lucía General University Hospital (HGUSL), C/Mezquita s/n, 30202 Cartagena, Spain.
- Catholic University of Murcia (UCAM), 30107 Murcia, Spain.
| | - George Perry
- UTSA Neurosciences Institute and Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA.
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
44
|
Kiyosawa N, Manabe S. Data-intensive drug development in the information age: applications of Systems Biology/Pharmacology/Toxicology. J Toxicol Sci 2017; 41:SP15-SP25. [PMID: 28003636 DOI: 10.2131/jts.41.sp15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Pharmaceutical companies continuously face challenges to deliver new drugs with true medical value. R&D productivity of drug development projects depends on 1) the value of the drug concept and 2) data and in-depth knowledge that are used rationally to evaluate the drug concept's validity. A model-based data-intensive drug development approach is a key competitive factor used by innovative pharmaceutical companies to reduce information bias and rationally demonstrate the value of drug concepts. Owing to the accumulation of publicly available biomedical information, our understanding of the pathophysiological mechanisms of diseases has developed considerably; it is the basis for identifying the right drug target and creating a drug concept with true medical value. Our understanding of the pathophysiological mechanisms of disease animal models can also be improved; it can thus support rational extrapolation of animal experiment results to clinical settings. The Systems Biology approach, which leverages publicly available transcriptome data, is useful for these purposes. Furthermore, applying Systems Pharmacology enables dynamic simulation of drug responses, from which key research questions to be addressed in the subsequent studies can be adequately informed. Application of Systems Biology/Pharmacology to toxicology research, namely Systems Toxicology, should considerably improve the predictability of drug-induced toxicities in clinical situations that are difficult to predict from conventional preclinical toxicology studies. Systems Biology/Pharmacology/Toxicology models can be continuously improved using iterative learn-confirm processes throughout preclinical and clinical drug discovery and development processes. Successful implementation of data-intensive drug development approaches requires cultivation of an adequate R&D culture to appreciate this approach.
Collapse
Affiliation(s)
- Naoki Kiyosawa
- Translational Medicine & Clinical Pharmacology Department, Daiichi Sankyo Co. Ltd
| | | |
Collapse
|
45
|
Evaluation of in vivo and in vitro models of toxicity by comparison of toxicogenomics data with the literature. Methods 2017; 132:57-65. [PMID: 28716510 DOI: 10.1016/j.ymeth.2017.07.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 02/01/2023] Open
Abstract
Toxicity affecting humans is studied by observing the effects of chemical substances in animal organisms (in vivo) or in animal and human cultivated cell lines (in vitro). Toxicogenomics studies collect gene expression profiles and histopathology assessment data for hundreds of drugs and pollutants in standardized experimental designs using different model systems. These data are an invaluable source for analyzing genome-wide drug response in biological systems. However, a problem remains that is how to evaluate the suitability of heterogeneous in vitro and in vivo systems to model the many different aspects of human toxicity. We propose here that a given model system (cell type or animal organ) is supported to appropriately describe a particular aspect of human toxicity if the set of compounds associated in the literature with that aspect of toxicity causes a change in expression of genes with a particular function in the tested model system. This approach provides candidate genes to explain the toxicity effect (the differentially expressed genes) and the compounds whose effect could be modeled (the ones producing both the change of expression in the model system and that are associated with the human phenotype in the literature). Here we present an application of this approach using a computational pipeline that integrates compound-induced gene expression profiles (from the Open TG-GATEs database) and biomedical literature annotations (from the PubMed database) to evaluate the suitability of (human and rat) in vitro systems as well as rat in vivo systems to model human toxicity.
Collapse
|
46
|
Abstract
The generation of large-scale biomedical data is creating unprecedented opportunities for basic and translational science. Typically, the data producers perform initial analyses, but it is very likely that the most informative methods may reside with other groups. Crowdsourcing the analysis of complex and massive data has emerged as a framework to find robust methodologies. When the crowdsourcing is done in the form of collaborative scientific competitions, known as Challenges, the validation of the methods is inherently addressed. Challenges also encourage open innovation, create collaborative communities to solve diverse and important biomedical problems, and foster the creation and dissemination of well-curated data repositories.
Collapse
|
47
|
Demonstration Study: A Protocol to Combine Online Tools and Databases for Identifying Potentially Repurposable Drugs. DATA 2017. [DOI: 10.3390/data2020015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
48
|
Nyström-Persson J, Natsume-Kitatani Y, Igarashi Y, Satoh D, Mizuguchi K. Interactive Toxicogenomics: Gene set discovery, clustering and analysis in Toxygates. Sci Rep 2017; 7:1390. [PMID: 28469246 PMCID: PMC5431224 DOI: 10.1038/s41598-017-01500-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/29/2017] [Indexed: 01/08/2023] Open
Abstract
Toxygates was originally released as a user-friendly interface to enhance the accessibility of the large-scale toxicogenomics database, Open TG-GATEs, generated by the Japanese Toxicogenomics Project. Since the original release, significant new functionality has been added to enable users to perform sophisticated computational analysis with only modest bioinformatics skills. The new features include an orthologous mode for data comparison among different species, interactive clustering and heatmap visualisation, enrichment analysis of gene sets, and user data uploading. In a case study, we use these new functions to study the hepatotoxicity of peroxisome proliferator-activated receptor alpha (PPARα) agonist WY-14643. Our findings suggest that WY-14643 caused hypertrophy in the bile duct by intracellular Ca2+ dysregulation, which resulted in the induction of genes in a non-canonical WNT/Ca2+ signalling pathway. With this new release of Toxygates, we provide a suite of tools that allow anyone to carry out in-depth analysis of toxicogenomics in Open TG-GATEs, and of any other dataset that is uploaded.
Collapse
Affiliation(s)
- Johan Nyström-Persson
- Level Five Co., Ltd., GYB Akihabara 3F, 2-25, Kanda-Sudacho, Chiyoda-ku, Tokyo, 101-0041, Japan.
| | - Yayoi Natsume-Kitatani
- Bioinformatics Project, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8, Asagi, Saito, Ibaraki-shi, Osaka, 567-0085, Japan.
| | - Yoshinobu Igarashi
- Toxicogenomics-informatics Project, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8, Asagi, Saito, Ibaraki-shi, Osaka, 567-0085, Japan
| | - Daisuke Satoh
- Level Five Co., Ltd., GYB Akihabara 3F, 2-25, Kanda-Sudacho, Chiyoda-ku, Tokyo, 101-0041, Japan
| | - Kenji Mizuguchi
- Bioinformatics Project, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8, Asagi, Saito, Ibaraki-shi, Osaka, 567-0085, Japan.
| |
Collapse
|
49
|
Rueda-Zárate HA, Imaz-Rosshandler I, Cárdenas-Ovando RA, Castillo-Fernández JE, Noguez-Monroy J, Rangel-Escareño C. A computational toxicogenomics approach identifies a list of highly hepatotoxic compounds from a large microarray database. PLoS One 2017; 12:e0176284. [PMID: 28448553 PMCID: PMC5407788 DOI: 10.1371/journal.pone.0176284] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 04/07/2017] [Indexed: 11/18/2022] Open
Abstract
The liver and the kidney are the most common targets of chemical toxicity, due to their major metabolic and excretory functions. However, since the liver is directly involved in biotransformation, compounds in many currently and normally used drugs could affect it adversely. Most chemical compounds are already labeled according to FDA-approved labels using DILI-concern scale. Drug Induced Liver Injury (DILI) scale refers to an adverse drug reaction. Many compounds do not exhibit hepatotoxicity at early stages of development, so it is important to detect anomalies at gene expression level that could predict adverse reactions in later stages. In this study, a large collection of microarray data is used to investigate gene expression changes associated with hepatotoxicity. Using TG-GATEs a large-scale toxicogenomics database, we present a computational strategy to classify compounds by toxicity levels in human and animal models through patterns of gene expression. We combined machine learning algorithms with time series analysis to identify genes capable of classifying compounds by FDA-approved labeling as DILI-concern toxic. The goal is to define gene expression profiles capable of distinguishing the different subtypes of hepatotoxicity. The study illustrates that expression profiling can be used to classify compounds according to different hepatotoxic levels; to label those that are currently labeled as undertemined; and to determine if at the molecular level, animal models are a good proxy to predict hepatotoxicity in humans.
Collapse
Affiliation(s)
- Héctor A. Rueda-Zárate
- School of Engineering and Sciences, Tecnológico de Monterrey Mexico City, Mexico City, México
| | - Iván Imaz-Rosshandler
- Computational Genomics Lab., Instituto Nacional de Medicina Genómica, Mexico City, México
| | | | | | - Julieta Noguez-Monroy
- School of Engineering and Sciences, Tecnológico de Monterrey Mexico City, Mexico City, México
| | - Claudia Rangel-Escareño
- Computational Genomics Lab., Instituto Nacional de Medicina Genómica, Mexico City, México
- * E-mail:
| |
Collapse
|
50
|
Abstract
BACKGROUND Methods for inference and comparison of biological networks are emerging as powerful tools for the identification of groups of tightly connected genes whose activity may be altered during disease progression or due to chemical perturbations. Connectivity-based comparisons help identify aggregate changes that would be difficult to detect with differential analysis methods comparing individual genes. METHODS In this study, we describe a pipeline for network comparison and its application to the analysis of gene expression datasets from chemical perturbation experiments, with the goal of elucidating the modes of actions of the profiled perturbations. We apply our pipeline to the analysis of the DrugMatrix and the TG-GATEs, two of the largest toxicogenomics resources available, containing gene expression measurements for model organisms exposed to hundreds of chemical compounds with varying carcinogenicity and genotoxicity. RESULTS Starting from chemical-specific transcriptional networks inferred from these data, we show that the proposed comparative analysis of their associated networks identifies groups of chemicals with similar functions and similar carcinogenicity/genotoxicity profiles. We also show that the in-silico annotation by pathway enrichment analysis of the gene modules with a significant gain or loss of connectivity for specific groups of compounds can reveal molecular pathways significantly associated with the chemical perturbations and their likely modes of action. CONCLUSIONS The proposed pipeline for transcriptional network inference and comparison is highly reproducible and allows grouping chemicals with similar functions and carcinogenicity/genotoxicity profiles. In the context of drug discovery or drug repositioning, the methods presented here could help assign new functions to novel or existing drugs, based on the similarity of their associated network with those built for other known compounds. Additionally, the method has broad applicability beyond the uses here described and could be used as an alternative or as a complement to standard approaches of differential gene expression analysis.
Collapse
|