1
|
Sepehri S, De Win D, Heymans A, Van Goethem F, Rodrigues RM, Rogiers V, Vanhaecke T. Next generation risk assessment of hair dye HC yellow no. 13: Ensuring protection from liver steatogenic effects. Regul Toxicol Pharmacol 2025; 159:105794. [PMID: 40024558 DOI: 10.1016/j.yrtph.2025.105794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/17/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
This study employs animal-free Next Generation Risk Assessment (NGRA) principles to evaluate the safety of repeated dermal exposure to 2.5% (w/w) HC Yellow No. 13 (HCY13) hair dye. As multiple in silico tools consistently flagged hepatotoxic potential, likely due to HCY13's trifluoromethyl group, which is known to interfere with hepatic lipid metabolism, liver steatosis was chosen as the primary mode of action for evaluation. AOP-guided in vitro tests were conducted, exposing human stem cell-derived hepatic cells to varying HCY13 concentrations over 72 h. The expression of 11 lipid metabolism-related marker genes (AHR, PPARA, LXRA, APOB, ACOX1, CPT1A, FASN, SCD1, DGAT2, CD36, and PPARG) and triglyceride accumulation, a phenotypic hallmark of steatosis, were measured. PROAST software was used to calculate in vitro Points of Departure (PoDNAM) for each biomarker. Using GastroPlus 9.9, physiologically-based pharmacokinetic (PBPK) models estimated internal liver concentrations (Cmax liver) of HCY13, ranging from 4 to 20 pM. All PoDNAM values significantly exceeded the predicted Cmax liver, indicating that HCY13 at 2.5% (w/w) is unlikely to induce liver steatosis under the assumed conditions. This research demonstrates the utility of NGRA, integrating AOP-based in vitro assays and computational models to protect human health and support regulatory decision-making without animal testing.
Collapse
Affiliation(s)
- Sara Sepehri
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| | - Dinja De Win
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| | - Anja Heymans
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| | - Freddy Van Goethem
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| | - Robim M Rodrigues
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| | - Vera Rogiers
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| | - Tamara Vanhaecke
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
2
|
Wang D, Suzuki A, Tong W. The connection between Bayesian networks and adverse outcome pathway (AOP) networks and how to use it for predicting drug toxicity. Drug Discov Today 2025; 30:104350. [PMID: 40187482 DOI: 10.1016/j.drudis.2025.104350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
There is significant interest in combining adverse outcome pathways (AOPs) with Bayesian networks (BNs) because of their shared representation using directed acyclic graphs (DAGs). However, it has not been verified empirically whether AOP networks are mathematically congruent with BNs. Furthermore, important properties for BNs, such as Markov blankets, have not been emphasized, which is a missed opportunity for simplifying and optimizing the model. Here, we summarize the connection between AOP networks and BNs and explore the implications for toxicity modeling. We also present a case study in drug-related liver toxicity. Our results confirm that AOP networks are congruent mathematically with BNs, with incorporation of the mathematical properties of BN leading to significantly simplified and more efficient models.
Collapse
Affiliation(s)
- Dong Wang
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA.
| | - Ayako Suzuki
- Division of Gastroenterology, Duke University, Durham, NC, USA; Department of Medicine, Durham VA Medical Center, Durham, NC, USA
| | - Weida Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| |
Collapse
|
3
|
Jeong J, Gasparyan M, Choi J. Advancing the quantitative understanding of adverse outcome pathways: current status, methodologies, and future directions. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2025; 44:614-623. [PMID: 39864436 DOI: 10.1093/etojnl/vgae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 01/28/2025]
Abstract
An adverse outcome pathway (AOP) framework maps the sequence of events leading to adverse outcomes from chemical exposures, providing a mechanistic understanding often absent in traditional methods. The quantitative AOP (qAOP) advances AOP by integrating quantitative data and mathematical modeling, thereby providing a more precise comprehension of relationships between molecular initiating events, key events, and adverse outcomes. This review critically examines three primary methodologies: systems toxicology, regression modeling, and Bayesian network modeling, highlighting their strengths, limitations, and specific data requirements within toxicology. Through an analysis of current methodologies and challenges, this review emphasizes the integration of experimental and computational approaches to elucidate key event relationships and proposes strategies for overcoming limitations through standardized protocols and advanced computational tools. By outlining future research directions and the potential of qAOPs to transform chemical risk assessment, this review aims to contribute to the advancement of regulatory science and the protection of public health and the environment.
Collapse
Affiliation(s)
- Jaeseong Jeong
- School of Environmental Engineering, University of Seoul, Seoul, Korea
| | - Manvel Gasparyan
- School of Environmental Engineering, University of Seoul, Seoul, Korea
| | - Jinhee Choi
- School of Environmental Engineering, University of Seoul, Seoul, Korea
| |
Collapse
|
4
|
Jochum K, Miccoli A, Sommersdorf C, Poetz O, Braeuning A, Tralau T, Marx-Stoelting P. Comparative case study on NAMs: towards enhancing specific target organ toxicity analysis. Arch Toxicol 2024; 98:3641-3658. [PMID: 39207506 PMCID: PMC11489238 DOI: 10.1007/s00204-024-03839-7] [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: 07/03/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Traditional risk assessment methodologies in toxicology have relied upon animal testing, despite concerns regarding interspecies consistency, reproducibility, costs, and ethics. New Approach Methodologies (NAMs), including cell culture and multi-level omics analyses, hold promise by providing mechanistic information rather than assessing organ pathology. However, NAMs face limitations, like lacking a whole organism and restricted toxicokinetic interactions. This is an inherent challenge when it comes to the use of omics data from in vitro studies for the prediction of organ toxicity in vivo. One solution in this context are comparative in vitro-in vivo studies as they allow for a more detailed assessment of the transferability of the respective NAM data. Hence, hepatotoxic and nephrotoxic pesticide active substances were tested in human cell lines and the results subsequently related to the biology underlying established effects in vivo. To this end, substances were tested in HepaRG and RPTEC/tERT1 cells at non-cytotoxic concentrations and analyzed for effects on the transcriptome and parts of the proteome using quantitative real-time PCR arrays and multiplexed microsphere-based sandwich immunoassays, respectively. Transcriptomics data were analyzed using three bioinformatics tools. Where possible, in vitro endpoints were connected to in vivo observations. Targeted protein analysis revealed various affected pathways, with generally fewer effects present in RPTEC/tERT1. The strongest transcriptional impact was observed for Chlorotoluron in HepaRG cells (increased CYP1A1 and CYP1A2 expression). A comprehensive comparison of early cellular responses with data from in vivo studies revealed that transcriptomics outperformed targeted protein analysis, correctly predicting up to 50% of in vivo effects.
Collapse
Affiliation(s)
- Kristina Jochum
- Department of Pesticides Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Andrea Miccoli
- Department of Pesticides Safety, German Federal Institute for Risk Assessment, Berlin, Germany
- Institute for Marine Biological Resources and Biotechnology (IRBIM), National Research Council, Ancona, Italy
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | | | - Oliver Poetz
- Signatope GmbH, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Albert Braeuning
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Tewes Tralau
- Department of Pesticides Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Philip Marx-Stoelting
- Department of Pesticides Safety, German Federal Institute for Risk Assessment, Berlin, Germany.
| |
Collapse
|
5
|
Ortega-Vallbona R, Palomino-Schätzlein M, Tolosa L, Benfenati E, Ecker GF, Gozalbes R, Serrano-Candelas E. Computational Strategies for Assessing Adverse Outcome Pathways: Hepatic Steatosis as a Case Study. Int J Mol Sci 2024; 25:11154. [PMID: 39456937 PMCID: PMC11508863 DOI: 10.3390/ijms252011154] [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: 09/20/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
The evolving landscape of chemical risk assessment is increasingly focused on developing tiered, mechanistically driven approaches that avoid the use of animal experiments. In this context, adverse outcome pathways have gained importance for evaluating various types of chemical-induced toxicity. Using hepatic steatosis as a case study, this review explores the use of diverse computational techniques, such as structure-activity relationship models, quantitative structure-activity relationship models, read-across methods, omics data analysis, and structure-based approaches to fill data gaps within adverse outcome pathway networks. Emphasizing the regulatory acceptance of each technique, we examine how these methodologies can be integrated to provide a comprehensive understanding of chemical toxicity. This review highlights the transformative impact of in silico techniques in toxicology, proposing guidelines for their application in evidence gathering for developing and filling data gaps in adverse outcome pathway networks. These guidelines can be applied to other cases, advancing the field of toxicological risk assessment.
Collapse
Affiliation(s)
- Rita Ortega-Vallbona
- ProtoQSAR S.L., Calle Nicolás Copérnico 6, Parque Tecnológico de Valencia, 46980 Paterna, Spain; (R.O.-V.); (M.P.-S.); (R.G.)
| | - Martina Palomino-Schätzlein
- ProtoQSAR S.L., Calle Nicolás Copérnico 6, Parque Tecnológico de Valencia, 46980 Paterna, Spain; (R.O.-V.); (M.P.-S.); (R.G.)
| | - Laia Tolosa
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av. Fernando Abril Martorell 106, 46026 Valencia, Spain;
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, C/Monforte de Lemos, 28029 Madrid, Spain
| | - Emilio Benfenati
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy;
| | - Gerhard F. Ecker
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek Platz 2, 1090 Wien, Austria;
| | - Rafael Gozalbes
- ProtoQSAR S.L., Calle Nicolás Copérnico 6, Parque Tecnológico de Valencia, 46980 Paterna, Spain; (R.O.-V.); (M.P.-S.); (R.G.)
- MolDrug AI Systems S.L., Olimpia Arozena Torres 45, 46108 Valencia, Spain
| | - Eva Serrano-Candelas
- ProtoQSAR S.L., Calle Nicolás Copérnico 6, Parque Tecnológico de Valencia, 46980 Paterna, Spain; (R.O.-V.); (M.P.-S.); (R.G.)
| |
Collapse
|
6
|
Yan Z, Qin G, Shi X, Jiang X, Cheng Z, Zhang Y, Nan N, Cao F, Qiu X, Sang N. Multilevel Screening Strategy to Identify the Hydrophobic Organic Components of Ambient PM 2.5 Associated with Hepatocellular Steatosis. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:10458-10469. [PMID: 38836430 DOI: 10.1021/acs.est.3c10012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Hepatic steatosis is the first step in a series of events that drives hepatic disease and has been considerably associated with exposure to fine particulate matter (PM2.5). Although the chemical constituents of particles matter in the negative health effects, the specific components of PM2.5 that trigger hepatic steatosis remain unclear. New strategies prioritizing the identification of the key components with the highest potential to cause adverse effects among the numerous components of PM2.5 are needed. Herein, we established a high-resolution mass spectrometry (MS) data set comprising the hydrophobic organic components corresponding to 67 PM2.5 samples in total from Taiyuan and Guangzhou, two representative cities in North and South China, respectively. The lipid accumulation bioeffect profiles of the above samples were also obtained. Considerable hepatocyte lipid accumulation was observed in most PM2.5 extracts. Subsequently, 40 of 695 components were initially screened through machine learning-assisted data filtering based on an integrated bioassay with MS data. Next, nine compounds were further selected as candidates contributing to hepatocellular steatosis based on absorption, distribution, metabolism, and excretion evaluation and molecular dockingin silico. Finally, seven components were confirmed in vitro. This study provided a multilevel screening strategy for key active components in PM2.5 and provided insight into the hydrophobic PM2.5 components that induce hepatocellular steatosis.
Collapse
Affiliation(s)
- Zhipeng Yan
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Shanxi 030006, PR China
| | - Guohua Qin
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Shanxi 030006, PR China
| | - Xiaodi Shi
- State Key Joint Laboratory for Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing 100871, PR China
| | - Xing Jiang
- State Key Joint Laboratory for Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing 100871, PR China
| | - Zhen Cheng
- State Key Joint Laboratory for Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing 100871, PR China
| | - Yaru Zhang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Shanxi 030006, PR China
| | - Nan Nan
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Shanxi 030006, PR China
| | - Fuyuan Cao
- Key Laboratory of Computational Intelligence and Chinese Information Processing of Ministry of Education, School of Computer and Information Technology, Shanxi University, Shanxi 030006, PR China
| | - Xinghua Qiu
- State Key Joint Laboratory for Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, and Center for Environment and Health, Peking University, Beijing 100871, PR China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Shanxi 030006, PR China
| |
Collapse
|
7
|
Verhoeven A, van Ertvelde J, Boeckmans J, Gatzios A, Jover R, Lindeman B, Lopez-Soop G, Rodrigues RM, Rapisarda A, Sanz-Serrano J, Stinckens M, Sepehri S, Teunis M, Vinken M, Jiang J, Vanhaecke T. A quantitative weight-of-evidence method for confidence assessment of adverse outcome pathway networks: A case study on chemical-induced liver steatosis. Toxicology 2024; 505:153814. [PMID: 38677583 DOI: 10.1016/j.tox.2024.153814] [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: 01/31/2024] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 04/29/2024]
Abstract
The field of chemical toxicity testing is undergoing a transition to overcome the limitations of in vivo experiments. This evolution involves implementing innovative non-animal approaches to improve predictability and provide a more precise understanding of toxicity mechanisms. Adverse outcome pathway (AOP) networks are pivotal in organizing existing mechanistic knowledge related to toxicological processes. However, these AOP networks are dynamic and require regular updates to incorporate the latest data. Regulatory challenges also persist due to concerns about the reliability of the information they offer. This study introduces a generic Weight-of-Evidence (WoE) scoring method, aligned with the tailored Bradford-Hill criteria, to quantitatively assess the confidence levels in key event relationships (KERs) within AOP networks. We use the previously published AOP network on chemical-induced liver steatosis, a prevalent form of human liver injury, as a case study. Initially, the existing AOP network is optimized with the latest scientific information extracted from PubMed using the free SysRev platform for artificial intelligence (AI)-based abstract inclusion and standardized data collection. The resulting optimized AOP network, constructed using Cytoscape, visually represents confidence levels through node size (key event, KE) and edge thickness (KERs). Additionally, a Shiny application is developed to facilitate user interaction with the dataset, promoting future updates. Our analysis of 173 research papers yielded 100 unique KEs and 221 KERs among which 72 KEs and 170 KERs, respectively, have not been previously documented in the prior AOP network or AOP-wiki. Notably, modifications in de novo lipogenesis, fatty acid uptake and mitochondrial beta-oxidation, leading to lipid accumulation and liver steatosis, garnered the highest KER confidence scores. In conclusion, our study delivers a generic methodology for developing and assessing AOP networks. The quantitative WoE scoring method facilitates in determining the level of support for KERs within the optimized AOP network, offering valuable insights into its utility in both scientific research and regulatory contexts. KERs supported by robust evidence represent promising candidates for inclusion in an in vitro test battery for reliably predicting chemical-induced liver steatosis within regulatory frameworks.
Collapse
Affiliation(s)
- Anouk Verhoeven
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jonas van Ertvelde
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Joost Boeckmans
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Alexandra Gatzios
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ramiro Jover
- Joint Research Unit in Experimental Hepatology, University of Valencia, Health Research Institute Hospital La Fe & CIBER of Hepatic and Digestive Diseases, Valencia, Spain
| | - Birgitte Lindeman
- Department of Chemical Toxicology, Norwegian Institute of Public Health, Oslo, Norway
| | - Graciela Lopez-Soop
- Department of Chemical Toxicology, Norwegian Institute of Public Health, Oslo, Norway
| | - Robim M Rodrigues
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Anna Rapisarda
- Joint Research Unit in Experimental Hepatology, University of Valencia, Health Research Institute Hospital La Fe & CIBER of Hepatic and Digestive Diseases, Valencia, Spain
| | - Julen Sanz-Serrano
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marth Stinckens
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sara Sepehri
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marc Teunis
- Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences Utrecht, Utrecht, the Netherlands
| | - Mathieu Vinken
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jian Jiang
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Tamara Vanhaecke
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
8
|
López-Pascual E, Rienda I, Perez-Rojas J, Rapisarda A, Garcia-Llorens G, Jover R, Castell JV. Drug-Induced Fatty Liver Disease (DIFLD): A Comprehensive Analysis of Clinical, Biochemical, and Histopathological Data for Mechanisms Identification and Consistency with Current Adverse Outcome Pathways. Int J Mol Sci 2024; 25:5203. [PMID: 38791241 PMCID: PMC11121209 DOI: 10.3390/ijms25105203] [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: 03/25/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Drug induced fatty liver disease (DIFLD) is a form of drug-induced liver injury (DILI), which can also be included in the more general metabolic dysfunction-associated steatotic liver disease (MASLD), which specifically refers to the accumulation of fat in the liver unrelated to alcohol intake. A bi-directional relationship between DILI and MASLD is likely to exist: while certain drugs can cause MASLD by acting as pro-steatogenic factors, MASLD may make hepatocytes more vulnerable to drugs. Having a pre-existing MASLD significantly heightens the likelihood of experiencing DILI from certain medications. Thus, the prevalence of steatosis within DILI may be biased by pre-existing MASLD, and it can be concluded that the genuine true incidence of DIFLD in the general population remains unknown. In certain individuals, drug-induced steatosis is often accompanied by concomitant injury mechanisms such as oxidative stress, cell death, and inflammation, which leads to the development of drug-induced steatohepatitis (DISH). DISH is much more severe from the clinical point of view, has worse prognosis and outcome, and resembles MASH (metabolic-associated steatohepatitis), as it is associated with inflammation and sometimes with fibrosis. A literature review of clinical case reports allowed us to examine and evaluate the clinical features of DIFLD and their association with specific drugs, enabling us to propose a classification of DIFLD drugs based on clinical outcomes and pathological severity: Group 1, drugs with low intrinsic toxicity (e.g., ibuprofen, naproxen, acetaminophen, irinotecan, methotrexate, and tamoxifen), but expected to promote/aggravate steatosis in patients with pre-existing MASLD; Group 2, drugs associated with steatosis and only occasionally with steatohepatitis (e.g., amiodarone, valproic acid, and tetracycline); and Group 3, drugs with a great tendency to transit to steatohepatitis and further to fibrosis. Different mechanisms may be in play when identifying drug mode of action: (1) inhibition of mitochondrial fatty acid β-oxidation; (2) inhibition of fatty acid transport across mitochondrial membranes; (3) increased de novo lipid synthesis; (4) reduction in lipid export by the inhibition of microsomal triglyceride transfer protein; (5) induction of mitochondrial permeability transition pore opening; (6) dissipation of the mitochondrial transmembrane potential; (7) impairment of the mitochondrial respiratory chain/oxidative phosphorylation; (8) mitochondrial DNA damage, degradation and depletion; and (9) nuclear receptors (NRs)/transcriptomic alterations. Currently, the majority of, if not all, adverse outcome pathways (AOPs) for steatosis in AOP-Wiki highlight the interaction with NRs or transcription factors as the key molecular initiating event (MIE). This perspective suggests that chemical-induced steatosis typically results from the interplay between a chemical and a NR or transcription factors, implying that this interaction represents the primary and pivotal MIE. However, upon conducting this exhaustive literature review, it became evident that the current AOPs tend to overly emphasize this interaction as the sole MIE. Some studies indeed support the involvement of NRs in steatosis, but others demonstrate that such NR interactions alone do not necessarily lead to steatosis. This view, ignoring other mitochondrial-related injury mechanisms, falls short in encapsulating the intricate biological mechanisms involved in chemically induced liver steatosis, necessitating their consideration as part of the AOP's map road as well.
Collapse
Affiliation(s)
- Ernesto López-Pascual
- Department of Biochemistry and Molecular Biology, University of Valencia, 46010 Valencia, Spain
- Joint Research Unit in Experimental Hepatology, Health Research Institute La Fe, 46026 Valencia, Spain
| | - Ivan Rienda
- Pathology Department, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Judith Perez-Rojas
- Pathology Department, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Anna Rapisarda
- Department of Biochemistry and Molecular Biology, University of Valencia, 46010 Valencia, Spain
- Joint Research Unit in Experimental Hepatology, Health Research Institute La Fe, 46026 Valencia, Spain
| | - Guillem Garcia-Llorens
- Joint Research Unit in Experimental Hepatology, Health Research Institute La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ramiro Jover
- Department of Biochemistry and Molecular Biology, University of Valencia, 46010 Valencia, Spain
- Joint Research Unit in Experimental Hepatology, Health Research Institute La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José V. Castell
- Department of Biochemistry and Molecular Biology, University of Valencia, 46010 Valencia, Spain
- Joint Research Unit in Experimental Hepatology, Health Research Institute La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
9
|
Ma P, Gao H, Shen N, Zhang L, Zhang Y, Zheng K, Xu B, Qin J, He J, Xu T, Li Y, Wu J, Yuan Y, Xue B. Association of urinary chlorpyrifos, paraquat, and cyproconazole levels with the severity of fatty liver based on MRI. BMC Public Health 2024; 24:807. [PMID: 38486191 PMCID: PMC10941454 DOI: 10.1186/s12889-024-18129-1] [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: 08/29/2023] [Accepted: 02/16/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND The objective of this study was to detect the urinary levels of chlorpyrifos, paraquat, and cyproconazole in residents living in Fuyang City and to analyze the correlation between these urinary pesticides levels and the severity of fatty liver disease (FLD). METHODS All participants' fat fraction (FF) values were recorded by MRI (Magnetic resonance imaging). First-morning urine samples were collected from 53 participants from Fuyang Peoples'Hospital. The levels of three urinary pesticides were measured using β-glucuronidase hydrolysis followed by a. The results were analyzed by using Pearson correlation analysis and binary logistic regression analysis to reveal the correlation between three urinary pesticides and the severity of fatty liver. RESULTS 53 individuals were divided into 3 groups based on the results from MRI, with 20 cases in the normal control group, 16 cases in the mild fatty liver group, and 17 cases in the moderate and severe fatty liver group. Urinary chlorpyrifos level was increased along with the increase of the severity of fatty liver. Urinary paraquat level was significantly higher both in the low-grade fatty liver group and moderate & serve grade fatty liver group compared with the control group. No significant differences in urinary cyproconazole levels were observed among the three groups. Furthermore, urinary chlorpyrifos and paraquat levels were positively correlated with FF value. And chlorpyrifos was the risk factor that may be involved in the development of FLD and Receiver Operating Characteristic curve (ROC curve) analysis showed that chlorpyrifos and paraquat may serve as potential predictors of FLD. CONCLUSION The present findings indicate urinary chlorpyrifos and paraquat were positively correlated with the severity of fatty liver. Moreover, urinary chlorpyrifos and paraquat have the potential to be considered as the predictors for development of FLD. Thus, this study may provide a new perspective from the environmental factors for the diagnosis, prevention, and treatment of FLD.
Collapse
Affiliation(s)
- Peiqi Ma
- Medical imaging center, Fuyang People's Hospital, 236000, Fuyang, China
| | - Hongliang Gao
- Core Laboratory, Department of Clinical Laboratory Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 211166, Nanjing, China
- School of Clinical Medicine, Wannan Medical College, 241000, Wuhu, China
| | - Ning Shen
- China Exposomics Institute (CEI) Precision Medicine Co. Ltd, 200120, Shanghai, China
| | - Lei Zhang
- Medical imaging center, Fuyang People's Hospital, 236000, Fuyang, China
| | - Yang Zhang
- Medical imaging center, Fuyang People's Hospital, 236000, Fuyang, China
| | - Kai Zheng
- Jiangsu Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, 210029, Nanjing, China
| | - Boqun Xu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Nanjing Medical University, 210011, Nanjing, China
| | - Jian Qin
- Department of Orthopaedics, Sir Run Run Hospital, Nanjing Medical University, 211100, Nanjing, China
| | - Jian He
- Department of Nuclear Medicine, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, 210029, Nanjing, China
| | - Tao Xu
- Core Laboratory, Department of Clinical Laboratory Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 211166, Nanjing, China
| | - Yan Li
- Core Laboratory, Department of Clinical Laboratory Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 211166, Nanjing, China.
| | - Jing Wu
- Core Laboratory, Department of Clinical Laboratory Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 211166, Nanjing, China.
| | - Yushan Yuan
- Medical imaging center, Fuyang People's Hospital, 236000, Fuyang, China.
| | - Bin Xue
- Department of General Surgery, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, 213003, Changzhou, China.
- Core Laboratory, Department of Clinical Laboratory Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, 211166, Nanjing, China.
| |
Collapse
|
10
|
Sadrabadi F, Alarcan J, Sprenger H, Braeuning A, Buhrke T. Impact of perfluoroalkyl substances (PFAS) and PFAS mixtures on lipid metabolism in differentiated HepaRG cells as a model for human hepatocytes. Arch Toxicol 2024; 98:507-524. [PMID: 38117326 PMCID: PMC10794458 DOI: 10.1007/s00204-023-03649-3] [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: 08/29/2023] [Accepted: 11/22/2023] [Indexed: 12/21/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are environmental contaminants with various adverse health effects in humans including disruption of lipid metabolism. Aim of the present study was to elucidate the molecular mechanisms of PFAS-mediated effects on lipid metabolism in human cells. Here, we examined the impact of a number of PFAS (PFOS, PFOA, PFNA, PFDA, PFHxA, PFBA, PFHxS, PFBS, HFPO-DA, and PMPP) and of some exposure-relevant PFAS mixtures being composed of PFOS, PFOA, PFNA and PFHxS on lipid metabolism in human HepaRG cells, an in vitro model for human hepatocytes. At near cytotoxic concentrations, the selected PFAS and PFAS mixtures induced triglyceride accumulation in HepaRG cells and consistently affected the expression of marker genes for steatosis, as well as PPARα target genes and genes related to lipid and cholesterol metabolism, pointing to common molecular mechanisms of PFAS in disrupting cellular lipid and cholesterol homeostasis. PPARα activation was examined by a transactivation assay in HEK293T cells, and synergistic effects were observed for the selected PFAS mixtures at sum concentrations higher than 25 µM, whereas additivity was observed at sum concentrations lower than 25 µM. Of note, any effect observed in the in vitro assays occurred at PFAS concentrations that were at least four to five magnitudes above real-life internal exposure levels of the general population.
Collapse
Affiliation(s)
- Faezeh Sadrabadi
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Jimmy Alarcan
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Heike Sprenger
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Albert Braeuning
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Thorsten Buhrke
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany.
| |
Collapse
|
11
|
Peslalz P, Grieshober M, Kraus F, Bleisch A, Izzo F, Lichtenstein D, Hammer H, Vorbach A, Momoi K, Zanger UM, Brötz-Oesterhelt H, Braeuning A, Plietker B, Stenger S. Unnatural Endotype B PPAPs as Novel Compounds with Activity against Mycobacterium tuberculosis. J Med Chem 2023; 66:15073-15083. [PMID: 37822271 DOI: 10.1021/acs.jmedchem.3c01172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Pre-SARS-CoV-2, tuberculosis was the leading cause of death by a single pathogen. Repetitive exposure of Mycobacterium tuberculosis(Mtb) supported the development of multidrug- and extensively drug-resistant strains, demanding novel drugs. Hyperforin, a natural type A polyprenylated polycyclic acylphloroglucinol from St. John's wort, exhibits antidepressant and antibacterial effects also against Mtb. Yet, Hyperforin's instability limits the utility in clinical practice. Here, we present photo- and bench-stable type B PPAPs with enhanced antimycobacterial efficacy. PPAP22 emerged as a lead compound, further improved as the sodium salt PPAP53, drastically enhancing solubility. PPAP53 inhibits the growth of virulent extracellular and intracellular Mtb without harming primary human macrophages. Importantly, PPAP53 is active against drug-resistant strains of Mtb. Furthermore, we analyzed the in vitro properties of PPAP53 in terms of CYP induction and the PXR interaction. Taken together, we introduce type PPAPs as a new class of antimycobacterial compounds, with remarkable antibacterial activity and favorable biophysical properties.
Collapse
Affiliation(s)
- Philipp Peslalz
- Chair of Organic Chemistry, Faculty of Chemistry and Food Chemistry, Technical University Dresden, Bergstr. 66, Dresden01069 ,Germany
| | - Mark Grieshober
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Albert-Einstein-Allee 11, Ulm D-89081, Germany
| | - Frank Kraus
- Institut für Organische Chemie, Universität Stuttgart,Pfaffenwaldring 55, Stuttgart 70569, Germany
| | - Anton Bleisch
- Chair of Organic Chemistry, Faculty of Chemistry and Food Chemistry, Technical University Dresden, Bergstr. 66, Dresden01069 ,Germany
| | - Flavia Izzo
- Institut für Organische Chemie, Universität Stuttgart,Pfaffenwaldring 55, Stuttgart 70569, Germany
| | - Dajana Lichtenstein
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, Berlin 10589, Germany
| | - Helen Hammer
- SIGNATOPE GmbH, Markwiesenstr. 55, Reutlingen 72770, Germany
| | - Andreas Vorbach
- Interfaculty Institute of Microbiology and Infection Medicine, Tübingen 72076, Germany
| | - Kyoko Momoi
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology Auerbachstr. 112, University of Tübingen, 70376 Stuttgart, Tübingen 72076, Germany
| | - Ulrich M Zanger
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology Auerbachstr. 112, University of Tübingen, 70376 Stuttgart, Tübingen 72076, Germany
| | - Heike Brötz-Oesterhelt
- Interfaculty Institute of Microbiology and Infection Medicine, Tübingen 72076, Germany
- German Center for Infection Research, Partner Site Tübingen, Tübingen 72076, Germany
| | - Albert Braeuning
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, Berlin 10589, Germany
| | - Bernd Plietker
- Chair of Organic Chemistry, Faculty of Chemistry and Food Chemistry, Technical University Dresden, Bergstr. 66, Dresden01069 ,Germany
- Institut für Organische Chemie, Universität Stuttgart,Pfaffenwaldring 55, Stuttgart 70569, Germany
| | - Steffen Stenger
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Albert-Einstein-Allee 11, Ulm D-89081, Germany
| |
Collapse
|
12
|
Karaca M, Fritsche K, Lichtenstein D, Vural Ö, Kreuzer K, Alarcan J, Braeuning A, Marx-Stoelting P, Tralau T. Adverse outcome pathway-based analysis of liver steatosis in vitro using human liver cell lines. STAR Protoc 2023; 4:102500. [PMID: 37616165 PMCID: PMC10463250 DOI: 10.1016/j.xpro.2023.102500] [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: 04/28/2023] [Revised: 06/21/2023] [Accepted: 07/19/2023] [Indexed: 08/25/2023] Open
Abstract
Here, we present an in vitro test battery to analyze chemicals for their potential to induce liver triglyceride accumulation, a hallmark of liver steatosis. We describe steps for using HepG2 and HepaRG human hepatoma cells in conjunction with a combination of several in vitro assays covering the different molecular initiating events and key events of the respective adverse outcome pathway. This protocol is suitable for assessing single substance effects as well as mixtures allowing their classification as steatotic or non-steatotic. For complete details on the use and execution of this protocol, please refer to Luckert et al. (2018),1 Lichtenstein et al. (2020),2 and Knebel et al. (2019).3.
Collapse
Affiliation(s)
- Mawien Karaca
- German Federal Institute for Risk Assessment, Department of Pesticides Safety, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany
| | - Kristin Fritsche
- German Federal Institute for Risk Assessment, Department Food Safety, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany
| | - Dajana Lichtenstein
- German Federal Institute for Risk Assessment, Department Food Safety, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany
| | - Özlem Vural
- German Federal Institute for Risk Assessment, Department Food Safety, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany
| | - Katrin Kreuzer
- German Federal Institute for Risk Assessment, Department Food Safety, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany
| | - Jimmy Alarcan
- German Federal Institute for Risk Assessment, Department Food Safety, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany
| | - Albert Braeuning
- German Federal Institute for Risk Assessment, Department Food Safety, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany
| | - Philip Marx-Stoelting
- German Federal Institute for Risk Assessment, Department of Pesticides Safety, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; Technical University of Berlin, Institute for Chemistry, Straße des 17. Juni 115, 10623 Berlin, Germany.
| | - Tewes Tralau
- German Federal Institute for Risk Assessment, Department of Pesticides Safety, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany
| |
Collapse
|
13
|
Zhao HN, Thomas SP, Zylka MJ, Dorrestein PC, Hu W. Urine Excretion, Organ Distribution, and Placental Transfer of 6PPD and 6PPD-Quinone in Mice and Potential Developmental Toxicity through Nuclear Receptor Pathways. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:13429-13438. [PMID: 37642336 PMCID: PMC11648498 DOI: 10.1021/acs.est.3c05026] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
The rubber antioxidant 6PPD has gained significant attention due to its highly toxic transformation product, 6PPD-quinone (6PPDQ). Despite their detection in urines of pregnant women, the placental transfer and developmental toxicity of 6PPD and 6PPDQ are unknown. Here, we treated C57Bl/6 mice with 4 mg/kg 6PPD or 6PPDQ to investigate their urine excretion and placental transfer. Female and male mice exhibited sex difference in excretion profiles of 6PPD and 6PPDQ. Urine concentrations of 6PPDQ were one order of magnitude lower than those of 6PPD, suggesting lower excretion and higher bioaccumulation of 6PPDQ. In pregnant mice treated with 6PPD or 6PPDQ from embryonic day 11.5 to 15.5, 6PPDQ showed ∼1.5-8 times higher concentrations than 6PPD in placenta, embryo body, and embryo brain, suggesting higher placental transfer of 6PPDQ. Using in vitro dual-luciferase reporter assays, we revealed that 6PPDQ activated the human retinoic acid receptor α (RARα) and retinoid X receptor α (RXRα) at concentrations as low as 0.3 μM, which was ∼10-fold higher than the concentrations detected in human urines. 6PPD activated the RXRα at concentrations as low as 1.2 μM. These results demonstrate the exposure risks of 6PPD and 6PPDQ during pregnancy and emphasize the need for further toxicological and epidemiological investigations.
Collapse
Affiliation(s)
- Haoqi Nina Zhao
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
| | - Sydney P. Thomas
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
| | - Mark J. Zylka
- University of North Carolina Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Pieter C. Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
- Collaborative Mass Spectrometry Innovation Center, University of California San Diego, La Jolla, California 92093, United States
- Department of Pharmacology, University of California San Diego, La Jolla, California 92093, United States
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California 92093, United States
| | - Wenxin Hu
- University of North Carolina Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
14
|
Thrikawala S, Mesmar F, Bhattacharya B, Muhsen M, Mukhopadhyay S, Flores S, Upadhyay S, Vergara L, Gustafsson JÅ, Williams C, Bondesson M. Triazole fungicides induce adipogenesis and repress osteoblastogenesis in zebrafish. Toxicol Sci 2023; 193:119-130. [PMID: 36951524 PMCID: PMC10230286 DOI: 10.1093/toxsci/kfad031] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023] Open
Abstract
Triazoles are a major group of azole fungicides commonly used in agriculture, and veterinary and human medicine. Maternal exposure to certain triazole antifungal medication causes congenital malformations, including skeletal malformations. We hypothesized that triazoles used as pesticides in agriculture also pose a risk of causing skeletal malformations in developing embryos. In this study, teratogenic effects of three commonly used triazoles, cyproconazole, paclobutrazol, and triadimenol, were investigated in zebrafish, Danio rerio. Exposure to the triazole fungicides caused bone and cartilage malformations in developing zebrafish larvae. Data from whole-embryo transcriptomics with cyproconazole suggested that exposure to this compound induces adipogenesis while repressing skeletal development. Confirming this finding, the expression of selected bone and cartilage marker genes were significantly downregulated with triazoles exposure as determined by quantitative PCR. The expression of selected adipogenic genes was upregulated by the triazoles. Furthermore, exposure to each of the three triazoles induced adipogenesis and lipid droplet formation in vitro in 3T3-L1 pre-adipocyte cells. In vivo in zebrafish larvae, cyproconazole exposure caused lipid accumulation. These results suggest that exposure to triazoles promotes adipogenesis at the expense of skeletal development, and thus they expand the chemical group of bona fide bone to fat switchers.
Collapse
Affiliation(s)
- Savini Thrikawala
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
| | - Fahmi Mesmar
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana, USA
| | - Beas Bhattacharya
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana, USA
| | - Maram Muhsen
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana, USA
| | - Srijita Mukhopadhyay
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, USA
| | - Sara Flores
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, USA
| | | | - Leoncio Vergara
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, USA
| | - Jan-Åke Gustafsson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, USA
| | - Cecilia Williams
- Science for Life Laboratory, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), KTH Royal Institute of Technology, Solna, Sweden
| | - Maria Bondesson
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana, USA
| |
Collapse
|
15
|
Karaca M, Willenbockel CT, Tralau T, Bloch D, Marx-Stoelting P. Toxicokinetic and toxicodynamic mixture effects of plant protection products: A case study. Regul Toxicol Pharmacol 2023; 141:105400. [PMID: 37116736 DOI: 10.1016/j.yrtph.2023.105400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 04/30/2023]
Abstract
Authorisation of ready to use plant protection products (PPPs) usually relies on the testing of acute and local toxicity only. This is in stark contrast to the situation for active substances where the mandatory data set comprises a most comprehensive set of studies. While the combination of certain active ingredients and co-formulants may nevertheless result in increased toxicity of the final product such combinations have never been evaluated systematically for complex and long-term toxicological endpoints. We therefore investigated the effect of three frequently used co-formulants on the toxicokinetic and toxicodynamic of the representative active substance combination of tebuconazol (Teb) and prothioconazol (Pro) or of cypermethrin (Cpm) and piperonyl butoxide (Pip), respectively. With all four active substances being potential liver steatogens, cytotoxicity and triglyceride accumulation in HepaRG were used as primary endpoints. Concomitantly transcriptomics and biochemical studies were applied to interrogate for effects on gene expression or inhibition of CYP3A4 as key enzyme for functionalization. Some of the tested combinations clearly showed more than additive effects, partly due to CYP3A4 enzyme inhibition. Other effects comprised the modulation of the expression and activity of steatosis-related nuclear key receptors. Altogether, the findings highlight the need for a more systematic consideration of toxicodynamic and toxicokinetic mixture effects during assessment of PPPs.
Collapse
Affiliation(s)
- Mawien Karaca
- German Federal Institute for Risk Assessment, Department of Pesticides Safety, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany; Technical University of Berlin, Institute for Chemistry, Straße des 17. Juni 115, 10623, Berlin, Germany
| | - Christian Tobias Willenbockel
- German Federal Institute for Risk Assessment, Department of Pesticides Safety, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Tewes Tralau
- German Federal Institute for Risk Assessment, Department of Pesticides Safety, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Denise Bloch
- German Federal Institute for Risk Assessment, Department of Pesticides Safety, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Philip Marx-Stoelting
- German Federal Institute for Risk Assessment, Department of Pesticides Safety, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany; Technical University of Berlin, Institute for Chemistry, Straße des 17. Juni 115, 10623, Berlin, Germany.
| |
Collapse
|
16
|
Müller FA, Stamou M, Englert FH, Frenzel O, Diedrich S, Suter-Dick L, Wambaugh JF, Sturla SJ. In vitro to in vivo extrapolation and high-content imaging for simultaneous characterization of chemically induced liver steatosis and markers of hepatotoxicity. Arch Toxicol 2023; 97:1701-1721. [PMID: 37046073 PMCID: PMC10182956 DOI: 10.1007/s00204-023-03490-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023]
Abstract
Chemically induced steatosis is characterized by lipid accumulation associated with mitochondrial dysfunction, oxidative stress and nucleus distortion. New approach methods integrating in vitro and in silico models are needed to identify chemicals that may induce these cellular events as potential risk factors for steatosis and associated hepatotoxicity. In this study we used high-content imaging for the simultaneous quantification of four cellular markers as sentinels for hepatotoxicity and steatosis in chemically exposed human liver cells in vitro. Furthermore, we evaluated the results with a computational model for the extrapolation of human oral equivalent doses (OED). First, we tested 16 reference chemicals with known capacities to induce cellular alterations in nuclear morphology, lipid accumulation, mitochondrial membrane potential and oxidative stress. Then, using physiologically based pharmacokinetic modeling and reverse dosimetry, OEDs were extrapolated from data of any stimulated individual sentinel response. The extrapolated OEDs were confirmed to be within biologically relevant exposure ranges for the reference chemicals. Next, we tested 14 chemicals found in food, selected from thousands of putative chemicals on the basis of structure-based prediction for nuclear receptor activation. Amongst these, orotic acid had an extrapolated OED overlapping with realistic exposure ranges. Thus, we were able to characterize known steatosis-inducing chemicals as well as data-scarce food-related chemicals, amongst which we confirmed orotic acid to induce hepatotoxicity. This strategy addresses needs of next generation risk assessment and can be used as a first chemical prioritization hazard screening step in a tiered approach to identify chemical risk factors for steatosis and hepatotoxicity-associated events.
Collapse
Affiliation(s)
- Fabrice A Müller
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, 8092, Zurich, Switzerland
| | - Marianna Stamou
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, 8092, Zurich, Switzerland
| | - Felix H Englert
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, 8092, Zurich, Switzerland
| | - Ole Frenzel
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, 8092, Zurich, Switzerland
| | - Sabine Diedrich
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, 8092, Zurich, Switzerland
| | - Laura Suter-Dick
- School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland, 4132, Muttenz, Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT), 4001, Basel, Switzerland
| | - John F Wambaugh
- Center for Computational Toxicology and Exposure, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, Durham, NC, 27711, USA
| | - Shana J Sturla
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, 8092, Zurich, Switzerland.
| |
Collapse
|
17
|
Louisse J, Fragki S, Rijkers D, Janssen A, van Dijk B, Leenders L, Staats M, Bokkers B, Zeilmaker M, Piersma A, Luijten M, Hoogenboom R, Peijnenburg A. Determination of in vitro hepatotoxic potencies of a series of perfluoroalkyl substances (PFASs) based on gene expression changes in HepaRG liver cells. Arch Toxicol 2023; 97:1113-1131. [PMID: 36864359 PMCID: PMC10025204 DOI: 10.1007/s00204-023-03450-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 01/24/2023] [Indexed: 03/04/2023]
Abstract
Per- and polyfluoroalkyl substances (PFASs) are omnipresent and have been shown to induce a wide range of adverse health effects, including hepatotoxicity, developmental toxicity, and immunotoxicity. The aim of the present work was to assess whether human HepaRG liver cells can be used to obtain insight into differences in hepatotoxic potencies of a series of PFASs. Therefore, the effects of 18 PFASs on cellular triglyceride accumulation (AdipoRed assay) and gene expression (DNA microarray for PFOS and RT-qPCR for all 18 PFASs) were studied in HepaRG cells. BMDExpress analysis of the PFOS microarray data indicated that various cellular processes were affected at the gene expression level. From these data, ten genes were selected to assess the concentration-effect relationship of all 18 PFASs using RT-qPCR analysis. The AdipoRed data and the RT-qPCR data were used for the derivation of in vitro relative potencies using PROAST analysis. In vitro relative potency factors (RPFs) could be obtained for 8 PFASs (including index chemical PFOA) based on the AdipoRed data, whereas for the selected genes, in vitro RPFs could be obtained for 11-18 PFASs (including index chemical PFOA). For the readout OAT5 expression, in vitro RPFs were obtained for all PFASs. In vitro RPFs were found to correlate in general well with each other (Spearman correlation) except for the PPAR target genes ANGPTL4 and PDK4. Comparison of in vitro RPFs with RPFs obtained from in vivo studies in rats indicate that best correlations (Spearman correlation) were obtained for in vitro RPFs based on OAT5 and CXCL10 expression changes and external in vivo RPFs. HFPO-TA was found to be the most potent PFAS tested, being around tenfold more potent than PFOA. Altogether, it may be concluded that the HepaRG model may provide relevant data to provide insight into which PFASs are relevant regarding their hepatotoxic effects and that it can be applied as a screening tool to prioritize other PFASs for further hazard and risk assessment.
Collapse
Affiliation(s)
- Jochem Louisse
- Wageningen Food Safety Research (WFSR), Wageningen, The Netherlands.
| | - Styliani Fragki
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Deborah Rijkers
- Wageningen Food Safety Research (WFSR), Wageningen, The Netherlands
| | - Aafke Janssen
- Wageningen Food Safety Research (WFSR), Wageningen, The Netherlands
| | - Bas van Dijk
- Wageningen Food Safety Research (WFSR), Wageningen, The Netherlands
| | - Liz Leenders
- Wageningen Food Safety Research (WFSR), Wageningen, The Netherlands
| | - Martijn Staats
- Wageningen Food Safety Research (WFSR), Wageningen, The Netherlands
| | - Bas Bokkers
- Centre for Safety of Substances and Products, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Marco Zeilmaker
- Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Aldert Piersma
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Mirjam Luijten
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Ron Hoogenboom
- Wageningen Food Safety Research (WFSR), Wageningen, The Netherlands
| | - Ad Peijnenburg
- Wageningen Food Safety Research (WFSR), Wageningen, The Netherlands
| |
Collapse
|
18
|
Fritsche K, Ziková-Kloas A, Marx-Stoelting P, Braeuning A. Metabolism-Disrupting Chemicals Affecting the Liver: Screening, Testing, and Molecular Pathway Identification. Int J Mol Sci 2023; 24:ijms24032686. [PMID: 36769005 PMCID: PMC9916672 DOI: 10.3390/ijms24032686] [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: 12/14/2022] [Revised: 01/26/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
The liver is the central metabolic organ of the body. The plethora of anabolic and catabolic pathways in the liver is tightly regulated by physiological signaling but may become imbalanced as a consequence of malnutrition or exposure to certain chemicals, so-called metabolic endocrine disrupters, or metabolism-disrupting chemicals (MDCs). Among different metabolism-related diseases, obesity and non-alcoholic fatty liver disease (NAFLD) constitute a growing health problem, which has been associated with a western lifestyle combining excessive caloric intake and reduced physical activity. In the past years, awareness of chemical exposure as an underlying cause of metabolic endocrine effects has continuously increased. Within this review, we have collected and summarized evidence that certain environmental MDCs are capable of contributing to metabolic diseases such as liver steatosis and cholestasis by different molecular mechanisms, thereby contributing to the metabolic syndrome. Despite the high relevance of metabolism-related diseases, standardized mechanistic assays for the identification and characterization of MDCs are missing. Therefore, the current state of candidate test systems to identify MDCs is presented, and their possible implementation into a testing strategy for MDCs is discussed.
Collapse
Affiliation(s)
- Kristin Fritsche
- German Federal Institute for Risk Assessment, Department Food Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| | - Andrea Ziková-Kloas
- German Federal Institute for Risk Assessment, Department Pesticides Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| | - Philip Marx-Stoelting
- German Federal Institute for Risk Assessment, Department Pesticides Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| | - Albert Braeuning
- German Federal Institute for Risk Assessment, Department Food Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
- Correspondence: ; Tel.: +49-(0)30-18412-25100
| |
Collapse
|
19
|
Knebel C, Süssmuth RD, Hammer HS, Braeuning A, Marx-Stoelting P. New Approach Methods for Hazard Identification: A Case Study with Azole Fungicides Affecting Molecular Targets Associated with the Adverse Outcome Pathway for Cholestasis. Cells 2022; 11:cells11203293. [PMID: 36291160 PMCID: PMC9600068 DOI: 10.3390/cells11203293] [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: 08/18/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 12/03/2022] Open
Abstract
Triazole fungicides such as propiconazole (Pi) or tebuconazole (Te) show hepatotoxicity in vivo, e.g., hypertrophy and vacuolization of liver cells following interaction with nuclear receptors such as PXR (pregnane-X-receptor) and CAR (constitutive androstane receptor). Accordingly, azoles affect gene expression associated with these adverse outcomes in vivo but also in human liver cells in vitro. Additionally, genes indicative of liver cholestasis are affected in vivo and in vitro. We therefore analyzed the capability of Pi and Te to cause cholestasis in an adverse outcome pathway (AOP)-driven approach in hepatic cells of human origin in vitro, considering also previous in vivo studies. Bile salt export pump (BSEP) activity assays confirmed that both azoles are weak inhibitors of BSEP. They alternate the expression of various cholestasis-associated target genes and proteins as well as the mitochondrial membrane function. Published in vivo data, however, demonstrate that neither Pi nor Te cause cholestasis in rodent bioassays. This discrepancy can be explained by the in vivo concentrations of both azoles being well below their EC50 for BSEP inhibition. From a regulatory perspective, this illustrates that toxicogenomics and human in vitro models are valuable tools to detect the potential of a substance to cause a specific type of toxicity. To come to a sound regulatory conclusion on the in vivo relevance of such a finding, results will have to be considered in a broader context also including toxicokinetics in a weight-of-evidence approach.
Collapse
Affiliation(s)
- Constanze Knebel
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Street 8-10, 10589 Berlin, Germany
| | - Roderich D. Süssmuth
- Institute of Chemistry, Technical University Berlin, Straße des 17. Juni 124, 10623 Berlin, Germany
| | - Helen S. Hammer
- Signatope GmbH, Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Albert Braeuning
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Street 8-10, 10589 Berlin, Germany
- Correspondence: (A.B.); (P.M.-S.); Tel.: +49-(0)30-18412-25100 (A.B.); Fax: +49-(0)30-18412-63758 (A.B.)
| | - Philip Marx-Stoelting
- Department Pesticides Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Street 8-10, 10589 Berlin, Germany
- Correspondence: (A.B.); (P.M.-S.); Tel.: +49-(0)30-18412-25100 (A.B.); Fax: +49-(0)30-18412-63758 (A.B.)
| |
Collapse
|
20
|
Driessen M, van der Plas-Duivesteijn S, Kienhuis AS, van den Brandhof EJ, Roodbergen M, van de Water B, Spaink HP, Palmblad M, van der Ven LTM, Pennings JLA. Identification of proteome markers for drug-induced liver injury in zebrafish embryos. Toxicology 2022; 477:153262. [PMID: 35868597 DOI: 10.1016/j.tox.2022.153262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/26/2022] [Accepted: 07/18/2022] [Indexed: 10/17/2022]
Abstract
The zebrafish embryo (ZFE) is a promising alternative non-rodent model in toxicology, and initial studies suggested its applicability in detecting hepatic responses related to drug-induced liver injury (DILI). Here, we hypothesize that detailed analysis of underlying mechanisms of hepatotoxicity in ZFE contributes to the improved identification of hepatotoxic properties of compounds and to the reduction of rodents used for hepatotoxicity assessment. ZFEs were exposed to nine reference hepatotoxicants, targeted at induction of steatosis, cholestasis, and necrosis, and effects compared with negative controls. Protein profiles of the individual compounds were generated using LC-MS/MS. We identified differentially expressed proteins and pathways, but as these showed considerable overlap, phenotype-specific responses could not be distinguished. This led us to identify a set of common hepatotoxicity marker proteins. At the pathway level, these were mainly associated with cellular adaptive stress-responses, whereas single proteins could be linked to common hepatotoxicity-associated processes. Applying several stringency criteria to our proteomics data as well as information from other data sources resulted in a set of potential robust protein markers, notably Igf2bp1, Cox5ba, Ahnak, Itih3b.2, Psma6b, Srsf3a, Ces2b, Ces2a, Tdo2b, and Anxa1c, for the detection of adverse responses.
Collapse
Affiliation(s)
- Marja Driessen
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), P.O.Box 1, 3720 BA Bilthoven, the Netherlands; Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, the Netherlands
| | | | - Anne S Kienhuis
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), P.O.Box 1, 3720 BA Bilthoven, the Netherlands
| | - Evert-Jan van den Brandhof
- Centre for Environmental Quality, National Institute for Public Health and the Environment (RIVM), P.O.Box 1, 3720 BA Bilthoven, the Netherlands
| | - Marianne Roodbergen
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), P.O.Box 1, 3720 BA Bilthoven, the Netherlands; Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, the Netherlands
| | - Bob van de Water
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, the Netherlands
| | - Herman P Spaink
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC Leiden, the Netherlands
| | - Magnus Palmblad
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Leo T M van der Ven
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), P.O.Box 1, 3720 BA Bilthoven, the Netherlands
| | - Jeroen L A Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), P.O.Box 1, 3720 BA Bilthoven, the Netherlands.
| |
Collapse
|
21
|
Cui S, Yu Y, Zhan T, Gao Y, Zhang J, Zhang L, Ge Z, Liu W, Zhang C, Zhuang S. Carcinogenic Risk of 2,6-Di- tert-Butylphenol and Its Quinone Metabolite 2,6-DTBQ Through Their Interruption of RARβ: In Vivo, In Vitro, and In Silico Investigations. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:480-490. [PMID: 34927421 DOI: 10.1021/acs.est.1c06866] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Thousands of contaminants are used worldwide and eventually released into the environment, presenting a challenge of health risk assessment. The identification of key toxic pathways and characterization of interactions with target biomacromolecules are essential for health risk assessments. The adverse outcome pathway (AOP) incorporates toxic mechanisms into health risk assessment by emphasizing the relationship among molecular initiating events (MIEs), key events (KEs), and adverse outcome (AO). Herein, we attempted the use of AOP to decipher the toxic effects of 2,6-di-tert-butylphenol (2,6-DTBP) and its para-quinone metabolite 2,6-di-tert-butyl-1,4-benzoquinone (2,6-DTBQ) based on integrated transcriptomics, molecular modeling, and cell-based assays. Through transcriptomics and quantitative real-time PCR validation, we identified retinoic acid receptor β (RARβ) as the key target biomacromolecule. The epigenetic analysis and molecular modeling revealed RARβ interference as one MIE, including DNA methylation and conformational changes. In vitro assays extended subsequent KEs, including altered protein expression of p-Erk1/2 and COX-2, and promoted cancer cell H4IIE proliferation and metastasis. These toxic effects altogether led to carcinogenic risk as the AO of 2,6-DTBP and 2,6-DTBQ, in line with chemical carcinogenesis identified from transcriptome profiling. Overall, our simplified AOP network of 2,6-DTBP and 2,6-DTBQ facilitates relevant health risk assessment.
Collapse
Affiliation(s)
- Shixuan Cui
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yang Yu
- Solid Waste and Chemicals Management Center, Ministry of Ecology and Environment (MEE), Beijing 100029, China
| | - Tingjie Zhan
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
- Women's Reproductive Health Key Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Yuchen Gao
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiachen Zhang
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Liang Zhang
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhiwei Ge
- Analysis Center of Agrobiology and Environmental Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weiping Liu
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chunlong Zhang
- Department of Environmental Sciences, University of Houston-Clear Lake, Houston, Texas 77058, United States
| | - Shulin Zhuang
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
- Women's Reproductive Health Key Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| |
Collapse
|
22
|
El Hamdaoui Y, Zheng F, Fritz N, Ye L, Tran MA, Schwickert K, Schirmeister T, Braeuning A, Lichtenstein D, Hellmich UA, Weikert D, Heinrich M, Treccani G, Schäfer MKE, Nowak G, Nürnberg B, Alzheimer C, Müller CP, Friedland K. Analysis of hyperforin (St. John's wort) action at TRPC6 channel leads to the development of a new class of antidepressant drugs. Mol Psychiatry 2022; 27:5070-5085. [PMID: 36224261 PMCID: PMC9763113 DOI: 10.1038/s41380-022-01804-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 09/02/2022] [Accepted: 09/14/2022] [Indexed: 01/14/2023]
Abstract
St. John's wort is an herb, long used in folk medicine for the treatment of mild depression. Its antidepressant constituent, hyperforin, has properties such as chemical instability and induction of drug-drug interactions that preclude its use for individual pharmacotherapies. Here we identify the transient receptor potential canonical 6 channel (TRPC6) as a druggable target to control anxious and depressive behavior and as a requirement for hyperforin antidepressant action. We demonstrate that TRPC6 deficiency in mice not only results in anxious and depressive behavior, but also reduces excitability of hippocampal CA1 pyramidal neurons and dentate gyrus granule cells. Using electrophysiology and targeted mutagenesis, we show that hyperforin activates the channel via a specific binding motif at TRPC6. We performed an analysis of hyperforin action to develop a new antidepressant drug that uses the same TRPC6 target mechanism for its antidepressant action. We synthesized the hyperforin analog Hyp13, which shows similar binding to TRPC6 and recapitulates TRPC6-dependent anxiolytic and antidepressant effects in mice. Hyp13 does not activate pregnan-X-receptor (PXR) and thereby loses the potential to induce drug-drug interactions. This may provide a new approach to develop better treatments for depression, since depression remains one of the most treatment-resistant mental disorders, warranting the development of effective drugs based on naturally occurring compounds.
Collapse
Affiliation(s)
- Yamina El Hamdaoui
- grid.5802.f0000 0001 1941 7111Pharmacology & Toxicology, Institute for Pharmaceutical and Biomedical Sciences, Johannes-Gutenberg Universität Mainz (JGU), Mainz, Germany
| | - Fang Zheng
- grid.5330.50000 0001 2107 3311Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Nikolas Fritz
- grid.5802.f0000 0001 1941 7111Pharmacology & Toxicology, Institute for Pharmaceutical and Biomedical Sciences, Johannes-Gutenberg Universität Mainz (JGU), Mainz, Germany
| | - Lian Ye
- grid.5802.f0000 0001 1941 7111Pharmacology & Toxicology, Institute for Pharmaceutical and Biomedical Sciences, Johannes-Gutenberg Universität Mainz (JGU), Mainz, Germany
| | - Mai Anh Tran
- grid.9613.d0000 0001 1939 2794Institute of Organic Chemistry and Macromolecular Chemistry, Faculty of Chemistry and Earth Science, Friedrich Schiller University Jena, Jena, Germany ,grid.5802.f0000 0001 1941 7111Biochemistry, Department of Chemistry, Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | - Kevin Schwickert
- grid.5802.f0000 0001 1941 7111Pharmacology & Toxicology, Institute for Pharmaceutical and Biomedical Sciences, Johannes-Gutenberg Universität Mainz (JGU), Mainz, Germany
| | - Tanja Schirmeister
- grid.5802.f0000 0001 1941 7111Pharmacology & Toxicology, Institute for Pharmaceutical and Biomedical Sciences, Johannes-Gutenberg Universität Mainz (JGU), Mainz, Germany
| | - Albert Braeuning
- grid.417830.90000 0000 8852 3623Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| | - Dajana Lichtenstein
- grid.417830.90000 0000 8852 3623Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| | - Ute A. Hellmich
- grid.9613.d0000 0001 1939 2794Institute of Organic Chemistry and Macromolecular Chemistry, Faculty of Chemistry and Earth Science, Friedrich Schiller University Jena, Jena, Germany ,grid.5802.f0000 0001 1941 7111Biochemistry, Department of Chemistry, Johannes-Gutenberg Universität Mainz, Mainz, Germany ,grid.517250.4Cluster of Excellence “Balance of the Microverse”, Friedrich-Schiller-Uniersität Jena, Jena, Germany ,grid.7839.50000 0004 1936 9721Center for Biomolecular Magnetic Resonance, Goethe-University, Frankfurt, Germany
| | - Dorothee Weikert
- grid.5330.50000 0001 2107 3311Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Markus Heinrich
- grid.5330.50000 0001 2107 3311Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Giulia Treccani
- grid.410607.4Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany ,grid.410607.4Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Michael K. E. Schäfer
- grid.410607.4Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1 (Bld. 505), 55131 Mainz, Germany
| | - Gabriel Nowak
- grid.5522.00000 0001 2162 9631Department of Pharmacobiology, Jagiellonian University Medical College, Krakow, Poland
| | - Bernd Nürnberg
- grid.10392.390000 0001 2190 1447Department of Pharmacology, Experimental Therapy & Toxicology, Eberhard-Karls-University of Tübingen, Tübingen, Germany
| | - Christian Alzheimer
- grid.5330.50000 0001 2107 3311Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Christian P. Müller
- grid.5330.50000 0001 2107 3311Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany ,grid.11875.3a0000 0001 2294 3534Centre for Drug Research, Universiti Sains Malaysia, 11800 Minden, Penang Malaysia
| | - Kristina Friedland
- Pharmacology & Toxicology, Institute for Pharmaceutical and Biomedical Sciences, Johannes-Gutenberg Universität Mainz (JGU), Mainz, Germany.
| |
Collapse
|
23
|
Alarcan J, de Sousa G, Katsanou ES, Spyropoulou A, Batakis P, Machera K, Rahmani R, Lampen A, Braeuning A, Lichtenstein D. Investigating the in vitro steatotic mixture effects of similarly and dissimilarly acting test compounds using an adverse outcome pathway-based approach. Arch Toxicol 2021; 96:211-229. [PMID: 34778935 PMCID: PMC8748329 DOI: 10.1007/s00204-021-03182-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/21/2021] [Indexed: 11/29/2022]
Abstract
Within the EuroMix project, we have previously developed an adverse outcome pathway (AOP)-based in vitro assay toolbox to investigate the combined effects of liver steatosis-inducing compounds in human HepaRG hepatocarcinoma cells. In this study, we applied the toolbox to further investigate mixture effects of combinations, featuring either similarly acting or dissimilarly acting substances. The valproic acid structural analogs 2-propylheptanoic acid (PHP) and 2-propylhexanoic acid (PHX) were chosen for establishing mixtures of similarly acting substances, while a combination with the pesticidal active substance clothianidin (CTD) was chosen for establishing mixtures of dissimilarly acting compounds. We first determined relative potency factors (RPFs) for each compound based on triglyceride accumulation results. Thereafter, equipotent mixtures were tested for nuclear receptor activation in transfected HepG2 cells, while gene expression and triglyceride accumulation were investigated in HepaRG cells, following the proposed AOP for liver steatosis. Dose addition was observed for all combinations and endpoints tested, indicating the validity of the additivity assumption also in the case of the tested mixtures of dissimilarly acting substances. Gene expression results indicate that the existing steatosis AOP can still be refined with respect to the early key event (KE) of gene expression, in order to reflect the diversity of molecular mechanisms underlying the adverse outcome.
Collapse
Affiliation(s)
- Jimmy Alarcan
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Georges de Sousa
- Institut Sophia Agrobiotech, Université Côte d'Azur-INRAE-CNRS, 06903, Sophia Antipolis, France
| | | | | | | | | | - Roger Rahmani
- Institut Sophia Agrobiotech, Université Côte d'Azur-INRAE-CNRS, 06903, Sophia Antipolis, France
| | - Alfonso Lampen
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Albert Braeuning
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany.
| | - Dajana Lichtenstein
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| |
Collapse
|
24
|
Primary Human Hepatocyte Spheroids as Tools to Study the Hepatotoxic Potential of Non-Pharmaceutical Chemicals. Int J Mol Sci 2021; 22:ijms222011005. [PMID: 34681664 PMCID: PMC8537720 DOI: 10.3390/ijms222011005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/07/2021] [Accepted: 10/07/2021] [Indexed: 12/18/2022] Open
Abstract
Drug-induced liver injury, including cholestasis, is an important clinical issue and economic burden for pharmaceutical industry and healthcare systems. However, human-relevant in vitro information on the ability of other types of chemicals to induce cholestatic hepatotoxicity is lacking. This work aimed at investigating the cholestatic potential of non-pharmaceutical chemicals using primary human hepatocytes cultured in 3D spheroids. Spheroid cultures were repeatedly (co-) exposed to drugs (cyclosporine-A, bosentan, macitentan) or non-pharmaceutical chemicals (paraquat, tartrazine, triclosan) and a concentrated mixture of bile acids for 4 weeks. Cell viability (adenosine triphosphate content) was checked every week and used to calculate the cholestatic index, an indicator of cholestatic liability. Microarray analysis was performed at specific time-points to verify the deregulation of genes related to cholestasis, steatosis and fibrosis. Despite the evident inter-donor variability, shorter exposures to cyclosporine-A consistently produced cholestatic index values below 0.80 with transcriptomic data partially supporting its cholestatic burden. Bosentan confirmed to be hepatotoxic, while macitentan was not toxic in the tested concentrations. Prolonged exposure to paraquat suggested fibrotic potential, while triclosan markedly deregulated genes involved in different types of hepatotoxicity. These results support the applicability of primary human hepatocyte spheroids to study hepatotoxicity of non-pharmaceutical chemicals in vitro.
Collapse
|
25
|
Lichtenstein D, Mentz A, Sprenger H, Schmidt FF, Albaum SP, Kalinowski J, Planatscher H, Joos TO, Poetz O, Braeuning A. A targeted transcriptomics approach for the determination of mixture effects of pesticides. Toxicology 2021; 460:152892. [PMID: 34371104 DOI: 10.1016/j.tox.2021.152892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/30/2021] [Accepted: 08/04/2021] [Indexed: 10/20/2022]
Abstract
While real-life exposure occurs to complex chemical mixtures, toxicological risk assessment mostly focuses on individual compounds. There is an increasing demand for in vitro tools and strategies for mixture toxicity analysis. Based on a previously established set of hepatotoxicity marker genes, we analyzed mixture effects of non-cytotoxic concentrations of different pesticides in exposure-relevant binary mixtures in human HepaRG hepatocarcinoma cells using targeted transcriptomics. An approach for mixture analysis at the level of a complex endpoint such as a transcript pattern is presented, including mixture design based on relative transcriptomic potencies and similarities. From a mechanistic point of view, goal of the study was to evaluate combinations of chemicals with varying degrees of similarity in order to determine whether differences in mechanisms of action lead to different mixtures effects. Using a model deviation ratio-based approach for assessing mixture effects, it was revealed that most data points are consistent with the assumption of dose addition. A tendency for synergistic effects was only observed at high concentrations of some combinations of the test compounds azoxystrobin, cyproconazole, difenoconazole, propiconazole and thiacloprid, which may not be representative of human real-life exposure. In summary, the findings of our study suggest that, for the pesticide mixtures investigated, risk assessment based on the general assumption of dose addition can be considered sufficiently protective for consumers. The way of data analysis presented in this paper can pave the way for a more comprehensive use of multi-gene expression data in experimental studies related to mixture toxicity.
Collapse
Affiliation(s)
- Dajana Lichtenstein
- German Federal Institute for Risk Assessment, Dept. Food Safety, Berlin, Germany
| | - Almut Mentz
- University of Bielefeld, CeBiTec, Bielefeld, Germany
| | - Heike Sprenger
- German Federal Institute for Risk Assessment, Dept. Food Safety, Berlin, Germany
| | - Felix F Schmidt
- NMI Natural and Medical Sciences Institute at the University of Tuebingen/Reutlingen, Germany; Signatope GmbH, 72770, Reutlingen, Germany
| | | | | | | | - Thomas O Joos
- NMI Natural and Medical Sciences Institute at the University of Tuebingen/Reutlingen, Germany; Signatope GmbH, 72770, Reutlingen, Germany
| | - Oliver Poetz
- NMI Natural and Medical Sciences Institute at the University of Tuebingen/Reutlingen, Germany; Signatope GmbH, 72770, Reutlingen, Germany
| | - Albert Braeuning
- German Federal Institute for Risk Assessment, Dept. Food Safety, Berlin, Germany.
| |
Collapse
|
26
|
Lichtenstein D, Lasch A, Alarcan J, Mentz A, Kalinowski J, Schmidt FF, Pötz O, Marx-Stoelting P, Braeuning A. An eight-compound mixture but not corresponding concentrations of individual chemicals induces triglyceride accumulation in human liver cells. Toxicology 2021; 459:152857. [PMID: 34273450 DOI: 10.1016/j.tox.2021.152857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/24/2021] [Accepted: 07/11/2021] [Indexed: 12/12/2022]
Abstract
In real life, organisms are exposed to complex mixtures of chemicals at low concentration levels, whereas research on toxicological effects is mostly focused on single compounds at comparably high doses. Mixture effects deviating from the assumption of additivity, especially synergistic effects, are of concern. In an adverse outcome pathway (AOP)-guided manner, we analyzed the accumulation of triglycerides in human HepaRG liver cells by a mixture of eight steatotic chemicals (amiodarone, benzoic acid, cyproconazole, flusilazole, imazalil, prochloraz, propiconazole and tebuconazole), each present below its individual effect concentration at 1-3 μM. Pronounced and significantly enhanced triglyceride accumulation was observed with the mixture, and similar effects were seen at the level of pregnane-X-receptor activation, a molecular initiating event leading to hepatic steatosis. Transcript pattern analysis indicated subtle pro-steatotic changes at low compound concentrations, which did not exert measurable effects on cellular triglycerides. Mathematical modeling of mixture effects indicated potentially more than additive behavior using a model for compounds with similar modes of action. The present data underline the usefulness of AOP-guided in vitro testing for the identification of mixture effects and highlight the need for further research on chemical mixtures and harmonization of data interpretation of mixture effects.
Collapse
Affiliation(s)
- Dajana Lichtenstein
- German Federal Institute for Risk Assessment, Dept. Food Safety, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Alexandra Lasch
- German Federal Institute for Risk Assessment, Dept. Pesticides Safety, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Jimmy Alarcan
- German Federal Institute for Risk Assessment, Dept. Food Safety, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Almut Mentz
- University of Bielefeld, CeBiTec, Universitätsstr. 27, 33615, Bielefeld, Germany
| | - Jörn Kalinowski
- University of Bielefeld, CeBiTec, Universitätsstr. 27, 33615, Bielefeld, Germany
| | - Felix F Schmidt
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstraße 55, 72770, Reutlingen, Germany; Signatope GmbH, Markwiesenstraße 55, 72770, Reutlingen, Germany
| | - Oliver Pötz
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstraße 55, 72770, Reutlingen, Germany; Signatope GmbH, Markwiesenstraße 55, 72770, Reutlingen, Germany
| | - Philip Marx-Stoelting
- German Federal Institute for Risk Assessment, Dept. Pesticides Safety, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Albert Braeuning
- German Federal Institute for Risk Assessment, Dept. Food Safety, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany.
| |
Collapse
|
27
|
Arnesdotter E, Spinu N, Firman J, Ebbrell D, Cronin MTD, Vanhaecke T, Vinken M. Derivation, characterisation and analysis of an adverse outcome pathway network for human hepatotoxicity. Toxicology 2021; 459:152856. [PMID: 34252478 DOI: 10.1016/j.tox.2021.152856] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/17/2021] [Accepted: 07/07/2021] [Indexed: 12/27/2022]
Abstract
Adverse outcome pathways (AOPs) and their networks are important tools for the development of mechanistically based non-animal testing approaches, such as in vitro and/or in silico assays, to assess toxicity induced by chemicals. In the present study, an AOP network connecting 14 linear AOPs related to human hepatotoxicity, currently available in the AOP-Wiki, was derived according to established criteria. The derived AOP network was characterised and analysed with regard to its structure and topological features. In-depth analysis of the AOP network showed that cell injury/death, oxidative stress, mitochondrial dysfunction and accumulation of fatty acids are the most highly connected and central key events. Consequently, these key events may be considered as the rational and mechanistically anchored basis for selecting, developing and/optimising in vitro and/or in silico assays to predict hepatotoxicity induced by chemicals in view of animal-free hazard identification.
Collapse
Affiliation(s)
- Emma Arnesdotter
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Brussels, Belgium.
| | - Nicoleta Spinu
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK.
| | - James Firman
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK.
| | - David Ebbrell
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK.
| | - Mark T D Cronin
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK.
| | - Tamara Vanhaecke
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Brussels, Belgium.
| | - Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Jette, Brussels, Belgium.
| |
Collapse
|
28
|
Schmidt FF, Lichtenstein D, Planatscher H, Mentz A, Kalinowski J, Steinhilber AE, Joos TO, Braeuning A, Pötz O. Pesticide mixture effects on liver protein abundance in HepaRG cells. Toxicology 2021; 458:152839. [PMID: 34153374 DOI: 10.1016/j.tox.2021.152839] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/27/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023]
Abstract
Toxicological effects of chemicals are mostly tested individually. However, consumers encounter exposure to complex mixtures, for example multiple pesticide residues, by consuming food such as crops, fruits or vegetables. Currently, more than 450 active substances are approved in the European Union, and there is little data on effects after combined exposure to several pesticides. Toxicological animal studies would increase enormously, if pesticide combinations had to be analyzed in vivo. Therefore, in vitro methods addressing this issue are needed. We have developed 32 immunoaffinity-based mass spectrometry assays to investigate the impact of hepatotoxic active substances on liver proteins in human HepaRG cells. Five compounds were selected based on their (dis)similar capability to modulate protein levels, and on their combined use in commercially available formulations. Four binary mixtures were prepared from these five substances and tested in different concentrations over three time points. We applied a novel statistical method to describe deviations from additivity and to detect antagonistic and synergistic effects. The results regarding the abundance of hepatotoxicity-related proteins showed additive behavior for 1323 out of 1427 endpoints tested, while 104 combinatorial effects deviating from additivity, such as antagonism or synergism were observed.
Collapse
Affiliation(s)
- Felix F Schmidt
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Dajana Lichtenstein
- German Federal Institute for Risk Assessment, Dept. Food Safety, Berlin, Germany
| | - Hannes Planatscher
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Almut Mentz
- University of Bielefeld, CeBiTec, Bielefeld, Germany
| | | | - Andreas E Steinhilber
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Thomas O Joos
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany; SIGNATOPE GmbH, 72770 Reutlingen, Germany
| | - Albert Braeuning
- German Federal Institute for Risk Assessment, Dept. Food Safety, Berlin, Germany
| | - Oliver Pötz
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany; SIGNATOPE GmbH, 72770 Reutlingen, Germany.
| |
Collapse
|
29
|
Lasch A, Marx-Stoelting P, Braeuning A, Lichtenstein D. More than additive effects on liver triglyceride accumulation by combinations of steatotic and non-steatotic pesticides in HepaRG cells. Arch Toxicol 2021; 95:1397-1411. [PMID: 33575850 PMCID: PMC8032629 DOI: 10.1007/s00204-021-02997-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/28/2021] [Indexed: 12/30/2022]
Abstract
The liver is constantly exposed to mixtures of hepatotoxic compounds, such as food contaminants and pesticides. Dose addition is regularly assumed for mixtures in risk assessment, which however might not be sufficiently protective in case of synergistic effects. Especially the prediction of combination effects of substances which do not share a common adverse outcome (AO) might be problematic. In this study, the focus was on the endpoint liver triglyceride accumulation in vitro, an indicator of hepatic fatty acid changes. The hepatotoxic compounds difenoconazole, propiconazole and tebuconazole were chosen which cause hepatic fatty acid changes in vivo, whereas fludioxonil was chosen as a hepatotoxic substance not causing fatty acid changes. Triglyceride accumulation was analyzed for combinations of steatotic and non-steatotic pesticides in human HepaRG hepatocarcinoma cells. Investigations revealed a potentiation of triglyceride accumulation by mixtures of the steatotic compounds with the non-steatotic fludioxonil, as compared to the single compounds. Mathematical modeling of combination effects indicated more than additive effects for the tested combinations if the method by Chou was applied, and a decrease in EC50 values of the steatotic compounds when applied in mixtures. Use of an adverse outcome pathway (AOP)-driven testing strategy for liver steatosis showed interactions of the test compounds with the nuclear receptors AHR, CAR and PXR, as well as a downregulation of ACOX2. An ACOX2-dependent mechanism underlying the observed mixture effect could not be verified using a siRNA approach. By contrast, a toxicokinetic interaction was identified including an inhibition of the metabolic enzyme CYP3A4 by fludioxonil and a decreased metabolic conversion of the CYP3A4 substrate difenoconazole when used in mixture experiments. In conclusion, an interaction by a steatotic and a non-steatotic compound at the toxicokinetic level on the endpoint triglyceride accumulation in vitro was described.
Collapse
Affiliation(s)
- Alexandra Lasch
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Philip Marx-Stoelting
- Department of Pesticides Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Albert Braeuning
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany.
| | - Dajana Lichtenstein
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| |
Collapse
|
30
|
Hammer H, Schmidt F, Marx-Stoelting P, Pötz O, Braeuning A. Cross-species analysis of hepatic cytochrome P450 and transport protein expression. Arch Toxicol 2021; 95:117-133. [PMID: 33150952 PMCID: PMC7811513 DOI: 10.1007/s00204-020-02939-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/20/2020] [Indexed: 12/19/2022]
Abstract
Most drugs and xenobiotics are metabolized in the liver. Amongst others, different cytochrome P450 (CYP) enzymes catalyze the metabolic conversion of foreign compounds, and various transport proteins are engaged in the excretion of metabolites from the hepatocytes. Inter-species and inter-individual differences in the hepatic levels and activities of drug-metabolizing enzymes and transporters result from genetic as well as from environmental factors, and play a decisive role in determining the pharmacokinetic properties of a compound in a given test system. To allow for a meaningful comparison of results from metabolism studies, it is, therefore, of utmost importance to know about the specific metabolic properties of the test systems, especially about the levels of metabolic enzymes such as the CYPs. Using a targeted proteomics approach, we, therefore, compared the hepatic levels of important CYP enzymes and transporters in different experimental systems in vivo and in vitro, namely Wistar rats, C57/Bl6 mice, mice humanized for the two xeno-sensing receptors PXR (pregnane-X-receptor) and CAR (constitutive androstane receptor), mice with human hepatocyte-repopulated livers, human HepaRG hepatocarcinoma cells, primary human hepatocytes, and human liver biopsies. In addition, the effects of xenobiotic inducers of drug metabolism on CYP enzymes and transporters were analyzed in selected systems. This study for the first time presents a comprehensive overview of similarities and differences in important drug metabolism-related proteins among the different experimental models.
Collapse
Affiliation(s)
- Helen Hammer
- Signatope, Markwiesenstraße 55, 72770, Reutlingen, Germany
| | - Felix Schmidt
- Signatope, Markwiesenstraße 55, 72770, Reutlingen, Germany
| | - Philip Marx-Stoelting
- Department of Pesticides Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Oliver Pötz
- Signatope, Markwiesenstraße 55, 72770, Reutlingen, Germany
| | - Albert Braeuning
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany.
| |
Collapse
|
31
|
Rathman J, Yang C, Ribeiro JV, Mostrag A, Thakkar S, Tong W, Hobocienski B, Sacher O, Magdziarz T, Bienfait B. Development of a Battery of In Silico Prediction Tools for Drug-Induced Liver Injury from the Vantage Point of Translational Safety Assessment. Chem Res Toxicol 2020; 34:601-615. [PMID: 33356149 DOI: 10.1021/acs.chemrestox.0c00423] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Drug-induced liver injury (DILI) remains a challenge when translating knowledge from the preclinical stage to human use cases. Attempts to model human DILI directly based on the information from drug labels have had some success; however, the approach falls short of providing insights or addressing uncertainty due to the difficulty of decoupling the idiosyncratic nature of human DILI outcomes. Our approach in this comparative analysis is to leverage existing preclinical and clinical data as well as information on metabolism to better translate mammalian to human DILI. The human DILI knowledge base from the United States Food and Drug Administration (U.S. FDA) National Center for Toxicology Research contains 1036 pharmaceuticals from diverse therapeutic categories. A human DILI training set of 305 oral marketed drugs was prepared and a binary classification scheme applied. The second knowledge base consists of mammalian repeated dose toxicity with liver toxicity data from various regulatory sources. Within this knowledge base, we identified 278 pharmaceuticals containing 198 marketed or withdrawn oral drugs with data from the U.S. FDA new drug application and 98 active pharmaceutical ingredients from ToxCast. From this collection, a set of 225 oral drugs was prepared as the mammalian hepatotoxicity training set with particular end points of pathology findings in the liver and bile duct. Both human and mammalian data sets were processed using various learning algorithms, including artificial intelligence approaches. The external validations for both models were comparable to the training statistics. These data sets were also used to extract species-differentiating chemotypes that differentiate DILI effects on humans from mammals. A systematic workflow was devised to predict human DILI and provide mechanistic insights. For a given query molecule, both human and mammalian models are run. If the predictions are discordant, both metabolites and parents are investigated for quantitative structure-activity relationship and species-differentiating chemotypes. Their results are combined using the Dempster-Shafer decision theory to yield a final outcome prediction for human DILI with estimated uncertainty. Finally, these tools are implementable within an in silico platform for systematic evaluation.
Collapse
Affiliation(s)
- James Rathman
- Molecular Networks GmbH - Computerchemie (MN-AM), 90411 Nurnberg, Germany.,Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Chihae Yang
- Molecular Networks GmbH - Computerchemie (MN-AM), 90411 Nurnberg, Germany
| | - J Vinicius Ribeiro
- Molecular Networks GmbH - Computerchemie (MN-AM), 90411 Nurnberg, Germany
| | - Aleksandra Mostrag
- Molecular Networks GmbH - Computerchemie (MN-AM), 90411 Nurnberg, Germany
| | - Shraddha Thakkar
- National Center for Toxicology Research, United States Food and Drug Administration, Jefferson, Arkansas 72079, United States
| | - Weida Tong
- National Center for Toxicology Research, United States Food and Drug Administration, Jefferson, Arkansas 72079, United States
| | - Bryan Hobocienski
- Molecular Networks GmbH - Computerchemie (MN-AM), 90411 Nurnberg, Germany
| | - Oliver Sacher
- Molecular Networks GmbH - Computerchemie (MN-AM), 90411 Nurnberg, Germany
| | - Tomasz Magdziarz
- Molecular Networks GmbH - Computerchemie (MN-AM), 90411 Nurnberg, Germany
| | - Bruno Bienfait
- Molecular Networks GmbH - Computerchemie (MN-AM), 90411 Nurnberg, Germany
| |
Collapse
|
32
|
Rives C, Fougerat A, Ellero-Simatos S, Loiseau N, Guillou H, Gamet-Payrastre L, Wahli W. Oxidative Stress in NAFLD: Role of Nutrients and Food Contaminants. Biomolecules 2020; 10:E1702. [PMID: 33371482 PMCID: PMC7767499 DOI: 10.3390/biom10121702] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/14/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is often the hepatic expression of metabolic syndrome and its comorbidities that comprise, among others, obesity and insulin-resistance. NAFLD involves a large spectrum of clinical conditions. These range from steatosis, a benign liver disorder characterized by the accumulation of fat in hepatocytes, to non-alcoholic steatohepatitis (NASH), which is characterized by inflammation, hepatocyte damage, and liver fibrosis. NASH can further progress to cirrhosis and hepatocellular carcinoma. The etiology of NAFLD involves both genetic and environmental factors, including an unhealthy lifestyle. Of note, unhealthy eating is clearly associated with NAFLD development and progression to NASH. Both macronutrients (sugars, lipids, proteins) and micronutrients (vitamins, phytoingredients, antioxidants) affect NAFLD pathogenesis. Furthermore, some evidence indicates disruption of metabolic homeostasis by food contaminants, some of which are risk factor candidates in NAFLD. At the molecular level, several models have been proposed for the pathogenesis of NAFLD. Most importantly, oxidative stress and mitochondrial damage have been reported to be causative in NAFLD initiation and progression. The aim of this review is to provide an overview of the contribution of nutrients and food contaminants, especially pesticides, to oxidative stress and how they may influence NAFLD pathogenesis.
Collapse
Affiliation(s)
- Clémence Rives
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Anne Fougerat
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Sandrine Ellero-Simatos
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Nicolas Loiseau
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Hervé Guillou
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Laurence Gamet-Payrastre
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Walter Wahli
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore
- Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
| |
Collapse
|
33
|
Yang C, Rathman JF, Magdziarz T, Mostrag A, Kulkarni S, Barton-Maclaren TS. Do Similar Structures Have Similar No Observed Adverse Effect Level (NOAEL) Values? Exploring Chemoinformatics Approaches for Estimating NOAEL Bounds and Uncertainties. Chem Res Toxicol 2020; 34:616-633. [PMID: 33296179 DOI: 10.1021/acs.chemrestox.0c00429] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Determination of the no observed adverse effect level (NOAEL) of a substance is an important step in safety and regulatory assessments. Application of conventional in silico strategies, for example, quantitative structure-activity relationship (QSAR) models, to predict NOAEL values is inherently problematic. Whereas QSAR models for well-defined toxicity endpoints such as Ames mutagenicity or skin sensitization can be developed from mechanistic knowledge of molecular initiating events and adverse outcome pathways, QSAR is not appropriate for predicting a NOAEL value, a concentration at which "no effect" is observed. This paper presents a chemoinformatics approach and explores how it can be further refined through the incorporation of toxicity endpoint-specific information to estimate confidence bounds for the NOAEL of a target substance, given experimentally determined NOAEL values for one or more suitable analogues. With a sufficiently large NOAEL database, we analyze how a difference in NOAEL values for pairs of structures depends on their pairwise similarity, where similarity takes both structural features and physicochemical properties into account. The width of the estimate NOAEL confidence interval is proportional to the uncertainty. Using the new threshold of toxicological concern (TTC) database enriched with antimicrobials, examples are presented to illustrate how uncertainty decreases with increasing analogue quality and also how NOAEL bounds estimation can be significantly improved by filtering the full database to include only substances that are in structure categories relevant to the target and analogue.
Collapse
Affiliation(s)
- Chihae Yang
- Molecular Networks GmbH Computerchemie, Neumeyerstraße 28, 90411 Nürnberg, Germany
| | - James F Rathman
- Molecular Networks GmbH Computerchemie, Neumeyerstraße 28, 90411 Nürnberg, Germany.,Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Tomasz Magdziarz
- Molecular Networks GmbH Computerchemie, Neumeyerstraße 28, 90411 Nürnberg, Germany
| | - Aleksandra Mostrag
- Molecular Networks GmbH Computerchemie, Neumeyerstraße 28, 90411 Nürnberg, Germany
| | - Sunil Kulkarni
- Existing Substances Risk Assessment Bureau, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Tara S Barton-Maclaren
- Existing Substances Risk Assessment Bureau, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| |
Collapse
|
34
|
Transcript and protein marker patterns for the identification of steatotic compounds in human HepaRG cells. Food Chem Toxicol 2020; 145:111690. [DOI: 10.1016/j.fct.2020.111690] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/20/2020] [Accepted: 08/11/2020] [Indexed: 12/18/2022]
|
35
|
Wang Y, Zhu Y, Niu J, Deng Q, Guo S, Jiang H, Peng Z, Xue Y, Peng H, Xuan L, Pan G. A novel bile acid analog, A17, ameliorated non-alcoholic steatohepatitis in high-fat diet-fed hamsters. Toxicol Appl Pharmacol 2020; 404:115169. [PMID: 32738331 DOI: 10.1016/j.taap.2020.115169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/12/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022]
Abstract
Being endocrine signaling molecules that regulate lipid metabolism and affect energy balance, bile acids are potential drug candidates for non-alcoholic steatohepatitis (NASH). Obeticholic acid (OCA) could improve NASH accompanied by significant side effects. Therefore, it is worthwhile to develop safer and more effective bile acid analogs. In this study, a new bile acid analog A17 was synthesized and its potential anti-NASH effects were assessed in vitro and in vivo. The impact of A17 on steatosis was investigated in the rat primary hepatocytes challenged with oleic acid. It was found that A17 alleviated lipid accumulation by reducing fatty acid (FA) uptake and promoting FA oxidation. The reduction of FA uptake came from inhibiting fatty acid translocase (Cd36) expression. The promotion of FA oxidation came from stimulating the phosphorylation of adenosine monophosphate (AMP)-activated protein kinase alpha (AMPKα). In addition, A17 reduced lipopolysaccharide-induced inflammation in Raw264.7 cells by activating Takeda G protein-coupled receptor 5 (TGR5). In in vivo study, male Golden Syrian hamsters were fed with high fat (HF) diet and then treated with 50 mg/kg/d A17 for 6 weeks. A17 lowered the lipid profiles and liver enzyme levels in serum and improved liver pathological conditions with less side effects compared with OCA. Further studies confirmed that the molecular mechanisms of A17 in vivo were similar to those in vitro. In conclusion, a novel bile acid analog A17 was identified to ameliorate NASH in HF-fed hamsters. The potential mechanisms could be contributed to reducing FA uptake, stimulating FA oxidation and relieving inflammation.
Collapse
Affiliation(s)
- Ying Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yao Zhu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junxing Niu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiangqiang Deng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shimeng Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; National Center for Drug Screening, Shanghai 201203, China
| | - Haowen Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; National Center for Drug Screening, Shanghai 201203, China
| | - Zhaoliang Peng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaru Xue
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huige Peng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lijiang Xuan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guoyu Pan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
36
|
Guan M, Xia P, Tian M, Chen D, Zhang X. Molecular fingerprints of conazoles via functional genomic profiling of Saccharomyces cerevisiae. Toxicol In Vitro 2020; 69:104998. [PMID: 32919014 DOI: 10.1016/j.tiv.2020.104998] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/02/2020] [Accepted: 09/06/2020] [Indexed: 10/23/2022]
Abstract
Conazoles were designed to inhibit ergosterol biosynthesis. Conazoles have been widely used as agricultural fungicides and are frequently detected in the environment. Although conazoles have been reported to have adverse effects, such as potential carcinogenic effects, the underlying molecular mechanisms of toxicity remain unclear. Here, the molecular fingerprints of five conazoles (propiconazole (Pro), penconazole (Pen), tebuconazole (Teb), flusilazole (Flu) and epoxiconazole (Epo)) were assessed in Saccharomyces cerevisiae (yeast) via functional genome-wide knockout mutant profiling. A total of 169 (4.49%), 176 (4.67%), 198 (5.26%), 218 (5.79%) and 173 (4.59%) responsive genes were identified at three concentrations (IC50, IC20 and IC10) of Pro, Pen, Teb, Flu and Epo, respectively. The five conazoles tended to have similar gene mutant fingerprints and toxicity mechanisms. "Ribosome" (sce03010) and "cytoplasmic translation" (GO: 0002181) were the common KEGG pathway and GO biological process term by gene set enrichment analysis of the responsive genes, which suggested that conazoles influenced protein synthesis. Conazoles also affected fatty acids synthesis because "biosynthesis of unsaturated fatty acids" pathway was among the top-ranked KEGG pathways. Moreover, two genes, YGR037C (acyl-CoA-binding protein) and YCR034W (fatty acid elongase), were key fingerprints of conazoles because they played vital roles in conazole-induced toxicity. Overall, the fingerprints derived from the yeast functional genomic screening provide an alternative approach to elucidate the molecular mechanisms of environmental pollutant conazoles.
Collapse
Affiliation(s)
- Miao Guan
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University, 163 Xianlin Ave., Nanjing, Jiangsu 210023, China; Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Rd., Nanjing, Jiangsu 210023, China.
| | - Pu Xia
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University, 163 Xianlin Ave., Nanjing, Jiangsu 210023, China
| | - Mingming Tian
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University, 163 Xianlin Ave., Nanjing, Jiangsu 210023, China
| | - Dong Chen
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University, 163 Xianlin Ave., Nanjing, Jiangsu 210023, China; Jiangsu Provincial Academy of Environmental Science, 176 North Jiangdong Rd., Nanjing, Jiangsu 210036, China
| | - Xiaowei Zhang
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University, 163 Xianlin Ave., Nanjing, Jiangsu 210023, China.
| |
Collapse
|
37
|
Inoue T, Iwazaki N, Araki T, Hitotsumachi H. Human-Induced Pluripotent Stem Cell-Derived Hepatocytes and their Culturing Methods to Maintain Liver Functions for Pharmacokinetics and Safety Evaluation of Pharmaceuticals. Curr Pharm Biotechnol 2020; 21:773-779. [PMID: 32003687 DOI: 10.2174/1389201021666200131123524] [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] [Received: 05/05/2019] [Revised: 10/23/2019] [Accepted: 12/31/2019] [Indexed: 12/12/2022]
Abstract
Human hepatocytes are essential cell types for pharmacokinetics and the safety evaluation of pharmaceuticals. However, widely used primary hepatocytes with individual variations in liver function lose those functions rapidly in culture. Hepatic cell lines are convenient to use but have low liver functions. Human-Induced Pluripotent Stem (hiPS) cells can be expanded and potentially differentiated into any cell or tissue, including the liver. HiPS cell-derived Hepatocyte-Like Cells (hiPSHeps) are expected to be extensively used as consistent functional human hepatocytes. Many laboratories are investigating methods of using hiPS cells to differentiate hepatocytes, but the derived cells still have immature liver functions. In this paper, we describe the current uses and limitations of conventional hepatic cells, evaluating the suitability of hiPS-Heps to pharmacokinetics and the safety evaluation of pharmaceuticals, and discuss the potential future use of non-conventional non-monolayer culture methods to derive fully functional hiPS-Heps.
Collapse
Affiliation(s)
- Tomoaki Inoue
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | - Norihiko Iwazaki
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | - Tetsuro Araki
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Japan
| | | |
Collapse
|
38
|
Voss L, Yilmaz K, Burkard L, Vidmar J, Stock V, Hoffmann U, Pötz O, Hammer HS, Peiser M, Braeuning A, Löschner K, Böhmert L, Sieg H. Impact of iron oxide nanoparticles on xenobiotic metabolism in HepaRG cells. Arch Toxicol 2020; 94:4023-4035. [PMID: 32914219 DOI: 10.1007/s00204-020-02904-1] [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] [Received: 07/22/2020] [Accepted: 09/02/2020] [Indexed: 12/17/2022]
Abstract
Iron oxide nanoparticles are used in various industrial fields, as a tool in biomedicine as well as in food colorants, and can therefore reach human metabolism via oral uptake or injection. However, their effects on the human body, especially the liver as one of the first target organs is still under elucidation. Here, we studied the influence of different representative iron oxide materials on xenobiotic metabolism of HepaRG cells. These included four iron oxide nanoparticles, one commercially available yellow food pigment (E172), and non-particulate ionic control FeSO4. The nanoparticles had different chemical and crystalline structures and differed in size and shape and were used at a concentration of 50 µg Fe/mL. We found that various CYP enzymes were downregulated by some but not all iron oxide nanoparticles, with the Fe3O4-particle, both γ-Fe2O3-particles, and FeSO4 exhibiting the strongest effects, the yellow food pigment E172 showing a minor effect and an α-Fe2O3 nanoparticle leading to almost no inhibition of phase I machinery. The downregulation was seen at the mRNA, protein expression, and activity levels. Thereby, no dependency on the size or chemical structure was found. This underlines the difficulty of the grouping of nanomaterials regarding their physiological impact, suggesting that every iron oxide nanoparticle species needs to be evaluated in a case-by-case approach.
Collapse
Affiliation(s)
- Linn Voss
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Kiymet Yilmaz
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Lea Burkard
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Janja Vidmar
- National Food Institute, Technical University of Denmark, Kemitorvet, Building 201, 2800 Kgs, Lyngby, Denmark
| | - Valerie Stock
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Ute Hoffmann
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Auerbachstr. 112, 70376, Stuttgart, Germany
| | - Oliver Pötz
- SIGNATOPE GmbH, Markwiesenstraße 55, 72770, Reutlingen, Germany
| | | | - Matthias Peiser
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Albert Braeuning
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Katrin Löschner
- National Food Institute, Technical University of Denmark, Kemitorvet, Building 201, 2800 Kgs, Lyngby, Denmark
| | - Linda Böhmert
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany.
| | - Holger Sieg
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Straße 8-10, 10589, Berlin, Germany.
| |
Collapse
|
39
|
Methodology for health risk assessment of combined exposures to multiple chemicals. Food Chem Toxicol 2020; 143:111520. [DOI: 10.1016/j.fct.2020.111520] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/13/2022]
|
40
|
Development of a QSAR model to predict hepatic steatosis using freely available machine learning tools. Food Chem Toxicol 2020; 142:111494. [DOI: 10.1016/j.fct.2020.111494] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/07/2020] [Accepted: 06/02/2020] [Indexed: 12/17/2022]
|
41
|
Sprong C, Crépet A, Metruccio F, Blaznik U, Anagnostopoulos C, Christodoulou DL, Jensen BH, Kennedy M, González N, Rehurkova I, Ruprich J, Te Biesebeek JD, Vanacker M, Moretto A, van Klaveren J. Cumulative dietary risk assessment overarching different regulatory silos using a margin of exposure approach: A case study with three chemical silos. Food Chem Toxicol 2020; 142:111416. [PMID: 32439593 DOI: 10.1016/j.fct.2020.111416] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/24/2020] [Accepted: 05/04/2020] [Indexed: 11/19/2022]
Abstract
Risk assessment of chemicals occurring in our diet is commonly performed for single chemicals without considering exposure to other chemicals. We performed a case study on risk assessment of combined dietary exposure to chemicals from different regulatory silos, i.e. pesticides (PPRs), persistent organic pollutants (POPs) and food additives (FAs). Chemicals were grouped into the cumulative assessment group (CAG) liver steatosis using a component-based approach. Based on literature, the CAG included 144 PPRs, 49 POPS and 7 FAs for which concentration data were available. For each silo, chronic combined dietary exposure was assessed for adults and children of nine European countries following the most commonly used exposure methodologies in Europe and by using a relative potency factor approach. For risk characterization, a Margin of Exposure (MOE) was calculated. To overarch the risk across silos, a normalised combined margin of exposure (nMOET) approach was proposed. This case study demonstrated that risk assessment of combined exposure to chemicals can be performed within regulatory silos. It also highlighted important differences in the conservatism of exposure scenarios, the derivation of point of departures and the subsequent acceptable MOEs between the silos. To overarch the risk despite these differences, a nMOET approach can be used.
Collapse
Affiliation(s)
- Corinne Sprong
- RIVM, National Institute for Public Health and the Environment, PO Box 1, 3720 BA, Bilthoven, the Netherlands.
| | - Amélie Crépet
- ANSES, French Agency for Food, Environmental and Occupational Health and Safety, Risk Assessment Department, Methodology and Studies Unit, 947001, Maisons-Alfort, France
| | - Francesca Metruccio
- ICPS, International Centre for PPRs and Health Risk Prevention, ASST Fatebenefratelli Sacco, Ospedale L. Sacco Via GB Grassi 74, 20157, Milano, Italy
| | - Urska Blaznik
- National Institute of Public Health, Environmental Health Centre, Trubarjeva 2, Ljubljana, Slovenia
| | - Chris Anagnostopoulos
- Benaki Phytopathological Institute, Department of Pesticide Control and Phytopharmacy, Laboratory of Pesticide Residues, 8 Stefanou Delta Street, Kifissia, Athens, 14561, Greece
| | | | - Bodil Hamborg Jensen
- Technical University of Denmark, National Food Institute, Division of Risk Assessment and Nutrition, Kemitorvet, Building 201, DK 2800, Lyngby, Denmark
| | - Marc Kennedy
- Fera Science Ltd, Sand Hutton, York, YO41 1LZ, United Kingdom
| | - Neus González
- Laboratory of Toxicology and Environmental Health, School of Medecine, IISPV, Universitat Rovira I Virgili, Reus, Catalonia, Spain
| | - Irena Rehurkova
- National Institute of Public Health in Prague, Centre for Health, Nutrition and Food, Brno, Czech Republic
| | - Jiří Ruprich
- National Institute of Public Health in Prague, Centre for Health, Nutrition and Food, Brno, Czech Republic
| | - Jan Dirk Te Biesebeek
- RIVM, National Institute for Public Health and the Environment, PO Box 1, 3720 BA, Bilthoven, the Netherlands
| | - Marie Vanacker
- ANSES, French Agency for Food, Environmental and Occupational Health and Safety, Risk Assessment Department, Methodology and Studies Unit, 947001, Maisons-Alfort, France
| | - Angelo Moretto
- ICPS, International Centre for PPRs and Health Risk Prevention, ASST Fatebenefratelli Sacco, Ospedale L. Sacco Via GB Grassi 74, 20157, Milano, Italy; Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Jacob van Klaveren
- RIVM, National Institute for Public Health and the Environment, PO Box 1, 3720 BA, Bilthoven, the Netherlands
| |
Collapse
|
42
|
The Connection of Azole Fungicides with Xeno-Sensing Nuclear Receptors, Drug Metabolism and Hepatotoxicity. Cells 2020; 9:cells9051192. [PMID: 32403288 PMCID: PMC7290820 DOI: 10.3390/cells9051192] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 12/21/2022] Open
Abstract
Azole fungicides, especially triazole compounds, are widely used in agriculture and as pharmaceuticals. For a considerable number of agricultural azole fungicides, the liver has been identified as the main target organ of toxicity. A number of previous studies points towards an important role of nuclear receptors such as the constitutive androstane receptor (CAR), the pregnane-X-receptor (PXR), or the aryl hydrocarbon receptor (AHR), within the molecular pathways leading to hepatotoxicity of these compounds. Nuclear receptor-mediated hepatic effects may comprise rather adaptive changes such as the induction of drug-metabolizing enzymes, to hepatocellular hypertrophy, histopathologically detectable fatty acid changes, proliferation of hepatocytes, and the promotion of liver tumors. Here, we present a comprehensive review of the current knowledge of the interaction of major agricultural azole-class fungicides with the three nuclear receptors CAR, PXR, and AHR in vivo and in vitro. Nuclear receptor activation profiles of the azoles are presented and related to histopathological findings from classic toxicity studies. Important issues such as species differences and multi-receptor agonism and the consequences for data interpretation and risk assessment are discussed.
Collapse
|
43
|
Lichtenstein D, Luckert C, Alarcan J, de Sousa G, Gioutlakis M, Katsanou ES, Konstantinidou P, Machera K, Milani ES, Peijnenburg A, Rahmani R, Rijkers D, Spyropoulou A, Stamou M, Stoopen G, Sturla SJ, Wollscheid B, Zucchini-Pascal N, Braeuning A, Lampen A. An adverse outcome pathway-based approach to assess steatotic mixture effects of hepatotoxic pesticides in vitro. Food Chem Toxicol 2020; 139:111283. [DOI: 10.1016/j.fct.2020.111283] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/06/2020] [Accepted: 03/16/2020] [Indexed: 12/29/2022]
|
44
|
Fischer BC, Rotter S, Schubert J, Marx-Stoelting P, Solecki R. Recommendations for international harmonisation, implementation and further development of suitable scientific approaches regarding the assessment of mixture effects. Food Chem Toxicol 2020; 141:111388. [PMID: 32348816 DOI: 10.1016/j.fct.2020.111388] [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] [Received: 12/12/2019] [Revised: 04/02/2020] [Accepted: 04/20/2020] [Indexed: 10/24/2022]
Abstract
Legal frameworks lay down requirements for risk assessment of combined exposure to multiple chemicals and their implementation where scientific methods are accepted by responsible authorities. In order to protect human health, an assessment of potential risks that might result from co-exposure to multiple chemical substances is requested by European legislation. Several approaches for risk assessment of mixtures of chemicals have been proposed, but none has been widely implemented in regulatory risk assessments, so far. EuroMix, an EU Horizon 2020 funded project, contributed to the improvement of internationally harmonised approaches for risk assessment of chemical mixtures. Based on in vitro and in silico tests, an integrated test strategy involving hazard and exposure assessment was developed and a web tool to conduct such assessments was provided. One further task within EuroMix was to make recommendations for international harmonisation, implementation and further development of suitable scientific approaches regarding the assessment of mixture effects. This paper briefly describes objectives and outcome of the EuroMix project as well as recent findings from OECD, WHO and EFSA addressing combined exposure to multiple chemicals. Building on this, five steps addressing further development needs and implementation of existing tools especially for risk managers and policy makers are proposed.
Collapse
Affiliation(s)
- Benjamin C Fischer
- Department Pesticides Safety, German Federal Institute for Risk Assessment - BfR, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Stefanie Rotter
- Department Pesticides Safety, German Federal Institute for Risk Assessment - BfR, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany.
| | - Jens Schubert
- Department Pesticides Safety, German Federal Institute for Risk Assessment - BfR, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Philip Marx-Stoelting
- Department Pesticides Safety, German Federal Institute for Risk Assessment - BfR, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| | - Roland Solecki
- Department Pesticides Safety, German Federal Institute for Risk Assessment - BfR, Max-Dohrn-Straße 8-10, 10589, Berlin, Germany
| |
Collapse
|
45
|
van der Voet H, Kruisselbrink JW, de Boer WJ, van Lenthe MS, van den Heuvel J(H, Crépet A, Kennedy MC, Zilliacus J, Beronius A, Tebby C, Brochot C, Luckert C, Lampen A, Rorije E, Sprong C, van Klaveren JD. The MCRA toolbox of models and data to support chemical mixture risk assessment. Food Chem Toxicol 2020; 138:111185. [DOI: 10.1016/j.fct.2020.111185] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/22/2020] [Accepted: 02/06/2020] [Indexed: 12/21/2022]
|
46
|
Lasch A, Alarcan J, Lampen A, Braeuning A, Lichtenstein D. Combinations of LXR and RXR agonists induce triglyceride accumulation in human HepaRG cells in a synergistic manner. Arch Toxicol 2020; 94:1303-1320. [DOI: 10.1007/s00204-020-02685-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/21/2020] [Indexed: 12/15/2022]
|
47
|
Braeuning A, Mentz A, Schmidt FF, Albaum SP, Planatscher H, Kalinowski J, Joos TO, Poetz O, Lichtenstein D. RNA-protein correlation of liver toxicity markers in HepaRG cells. EXCLI JOURNAL 2020; 19:135-153. [PMID: 32194361 PMCID: PMC7068204 DOI: 10.17179/excli2019-2005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/15/2020] [Indexed: 12/23/2022]
Abstract
The liver is a main target organ for the toxicity of many different compounds. While in general, in vivo testing is still routinely used for assessing the hepatotoxic potential of test chemicals, the use of in vitro models offers advantages with regard to throughput, consumption of resources, and animal welfare aspects. Using the human hepatoma cell line HepaRG, we performed a comparative evaluation of a panel of hepatotoxicity marker mRNAs and proteins after exposure of the cells to 30 different pesticidal active compounds comprising herbizides, fungicides, insecticides, and others. The panel of hepatotoxicity markers included nuclear receptor target genes, key players of fatty acid and bile acid metabolism-related pathways, as well as recently identified biomarkers of drug-induced liver injury. Moreover, marker genes and proteins were identified, for example, S100P, ANXA10, CYP1A1, and CYP7A1. These markers respond with high sensitivity to stimulation with chemically diverse test compounds already at non-cytotoxic concentrations. The potency of the test compounds, determined as an overall parameter of their ability to deregulate marker expression in vitro, was very similar between the mRNA and protein levels. Thus, this study does not only characterize the response of human liver cells to 30 different pesticides but also demonstrates that hepatotoxicity testing in human HepaRG cells yields well comparable results at the mRNA and protein levels. Furthermore, robust hepatotoxicity marker genes and proteins were identified in HepaRG cells.
Collapse
Affiliation(s)
- Albert Braeuning
- German Federal Institute for Risk Assessment, Dept. Food Safety, Berlin, Germany
| | - Almut Mentz
- Center for Biotechnology (CeBiTec), Universität Bielefeld, Bielefeld, Germany
| | | | - Stefan P. Albaum
- Center for Biotechnology (CeBiTec), Universität Bielefeld, Bielefeld, Germany
| | | | - Jörn Kalinowski
- Center for Biotechnology (CeBiTec), Universität Bielefeld, Bielefeld, Germany
| | - Thomas O. Joos
- Signatope GmbH, Reutlingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Tübingen,Germany
| | - Oliver Poetz
- Signatope GmbH, Reutlingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Tübingen,Germany
| | - Dajana Lichtenstein
- German Federal Institute for Risk Assessment, Dept. Food Safety, Berlin, Germany
| |
Collapse
|
48
|
Behr AC, Plinsch C, Braeuning A, Buhrke T. Activation of human nuclear receptors by perfluoroalkylated substances (PFAS). Toxicol In Vitro 2019; 62:104700. [PMID: 31676336 DOI: 10.1016/j.tiv.2019.104700] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 10/25/2022]
Abstract
Perfluoralkylated substances (PFAS) such as perfluorooctanoic acid (PFOA) or perfluorooctanesulfonic acid (PFOS) are used to produce, e.g., surface coatings with water- and dirt-repellent properties. These substances have been shown to be hepatotoxic in rodents, and the mechanism of action is mostly attributed to the PFAS-mediated activation of the peroxisome proliferator-activated receptor alpha (PPARα). In the present study, we investigated by using luciferase-based reporter gene assays whether PFOA, PFOS and six alternative PFAS can activate, in addition to PPARα, eight other human nuclear receptors. All tested PFAS except for perfluorobutanesulfonic acid (PFBS) were able to activate human PPARα. Perfluoro-2-methyl-3-oxahexanoic acid (PMOH) and 3H-perfluoro-3-[(3-methoxypropoxy) propanoic acid] (PMPP) were weak agonists of human PPARγ. The other human nuclear receptors (PPARδ, CAR, PXR, FXR, LXRα, RXRα and RARα) were not affected by any PFAS tested in this study. Although PMOH was more effective than PFOA in stimulating PPARα in the transactivation assay, it was less effective in stimulating PPARα-dependent target gene expression in human HepG2 hepatocarcinoma cells. Notably, any effect observed in this in vitro study only occurred at concentrations higher than 10 μM of the respective PFAS which is in all cases several magnitudes above the average blood concentration in the Western population. Thus, the results suggest that nuclear receptor activation may only play a minor role in potential PFAS-mediated adverse effects in humans.
Collapse
Affiliation(s)
- Anne-Cathrin Behr
- German Federal Institute for Risk Assessment, Department of Food Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| | - Christin Plinsch
- German Federal Institute for Risk Assessment, Department of Food Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| | - Albert Braeuning
- German Federal Institute for Risk Assessment, Department of Food Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| | - Thorsten Buhrke
- German Federal Institute for Risk Assessment, Department of Food Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany.
| |
Collapse
|
49
|
Müller FA, Sturla SJ. Human in vitro models of nonalcoholic fatty liver disease. CURRENT OPINION IN TOXICOLOGY 2019. [DOI: 10.1016/j.cotox.2019.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
50
|
Knebel C, Buhrke T, Süssmuth R, Lampen A, Marx-Stoelting P, Braeuning A. Pregnane X receptor mediates steatotic effects of propiconazole and tebuconazole in human liver cell lines. Arch Toxicol 2019; 93:1311-1322. [PMID: 30989312 DOI: 10.1007/s00204-019-02445-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/09/2019] [Indexed: 12/26/2022]
Abstract
Triazoles are commonly used fungicides which show liver toxicity in rodent studies. While hepatocellular hypertrophy is the most prominent finding, some triazoles have also been reported to cause hepatocellular steatosis. The aim of our study was to elucidate molecular mechanisms of triazole-mediated steatosis. Therefore, we used the two triazoles propiconazole (Pi) and tebuconazole (Te) as test compounds in in vitro assays using the human hepatocarcinoma cell lines HepG2 and HepaRG. Triglyceride accumulation was measured using the Adipored assay and by a gas-chromatographic method. Reporter gene analyses were used to assess the ability of Pi and Te to activate nuclear receptors, which are described as the molecular initiators in the adverse outcome pathway (AOP) for liver steatosis. The expression of steatosis-associated genes was investigated by RT-PCR. Mechanistic analyses of triazole-mediated steatosis were performed using HepaRG subclones that are deficient in different nuclear receptors. Pi and Te both interacted with the constitutive androstane receptor (CAR), the peroxisome proliferator-activated receptor alpha (PPARα), and the pregnane X receptor (PXR). Both compounds induced expression of steatosis-related genes and cellular triglyceride accumulation. The knockout of PXR in HepaRG cells, but not the CAR knockout, abolished triazole-induced triglyceride accumulation, thus underlining the crucial role of PXR in hepatic steatosis resulting from exposure to these fungicides. In conclusion, our findings provide new insight into the molecular mechanisms of steatosis induction by triazole fungicides and identify PXR as a critical mediator of this process.
Collapse
Affiliation(s)
- Constanze Knebel
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Thorsten Buhrke
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Roderich Süssmuth
- Institute of Chemistry, Technical University Berlin, Straße des 17.Juni 124, 10623, Berlin, Germany
| | - Alfonso Lampen
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Philip Marx-Stoelting
- Department Pesticides Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany.
| | - Albert Braeuning
- Department Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| |
Collapse
|