1
|
Wuerger LTD, Sprenger H, Krasikova K, Templin M, Stahl A, Herfurth UM, Sieg H, Braeuning A. A multi-omics approach to elucidate okadaic acid-induced changes in human HepaRG hepatocarcinoma cells. Arch Toxicol 2024; 98:2919-2935. [PMID: 38832940 PMCID: PMC11324782 DOI: 10.1007/s00204-024-03796-1] [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: 03/12/2024] [Accepted: 05/23/2024] [Indexed: 06/06/2024]
Abstract
Okadaic acid (OA), a prevalent marine biotoxin found in shellfish, is known for causing acute gastrointestinal symptoms. Despite its potential to reach the bloodstream and the liver, the hepatic effects of OA are not well understood, highlighting a significant research gap. This study aims to comprehensively elucidate the impact of OA on the liver by examining the transcriptome, proteome, and phosphoproteome alterations in human HepaRG liver cells exposed to non-cytotoxic OA concentrations. We employed an integrative multi-omics approach, encompassing RNA sequencing, shotgun proteomics, phosphoproteomics, and targeted DigiWest analysis. This enabled a detailed exploration of gene and protein expression changes, alongside phosphorylation patterns under OA treatment. The study reveals concentration- and time-dependent deregulation in gene and protein expression, with a significant down-regulation of xenobiotic and lipid metabolism pathways. Up-regulated pathways include actin crosslink formation and a deregulation of apoptotic pathways. Notably, our results revealed that OA, as a potent phosphatase inhibitor, induces alterations in actin filament organization. Phosphoproteomics data highlighted the importance of phosphorylation in enzyme activity regulation, particularly affecting proteins involved in the regulation of the cytoskeleton. OA's inhibition of PP2A further leads to various downstream effects, including alterations in protein translation and energy metabolism. This research expands the understanding of OA's systemic impact, emphasizing its role in modulating the phosphorylation landscape, which influences crucial cellular processes. The results underscore OA's multifaceted effects on the liver, particularly through PP2A inhibition, impacting xenobiotic metabolism, cytoskeletal dynamics, and energy homeostasis. These insights enhance our comprehension of OA's biological significance and potential health risks.
Collapse
Affiliation(s)
- Leonie T D Wuerger
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Heike Sprenger
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Ksenia Krasikova
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Markus Templin
- NMI Natural and Medical Sciences Institute at the University Tübingen, Reutlingen, Germany
| | - Aaron Stahl
- NMI Natural and Medical Sciences Institute at the University Tübingen, Reutlingen, Germany
| | - Uta M Herfurth
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Holger Sieg
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany.
| | - Albert Braeuning
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| |
Collapse
|
2
|
Abrehame S, Hung MY, Chen YY, Liu YT, Chen YT, Liu FC, Lin YC, Chen YP. Selection of Fermentation Supernatant from Probiotic Strains Exhibiting Intestinal Epithelial Barrier Protective Ability and Evaluation of Their Effects on Colitis Mouse and Weaned Piglet Models. Nutrients 2024; 16:1138. [PMID: 38674829 PMCID: PMC11053620 DOI: 10.3390/nu16081138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
The intestinal epithelial barrier can prevent the invasion of pathogenic microorganisms and food antigens to maintain a consistent intestinal homeostasis. However, an imbalance in this barrier can result in various diseases, such as inflammatory bowel disease, malnutrition, and metabolic disease. Thus, the aim of this study was to select probiotic strains with epithelial barrier-enhancing ability in cell-based model and further investigate them for their improving effects on colitis mouse and weaned piglet models. The results showed that selected specific cell-free fermentation supernatants (CFSs) from Ligilactobacillus salivarius P1, Lactobacillus gasseri P12, and Limosilactobacillus reuteri G7 promoted intestinal epithelial cell growth and proliferation, strengthening the intestinal barrier in an intestinal epithelial cell line Caco-2 model. Further, the administration of CFSs of L. salivarius P1, L. gasseri P12, and L. reuteri G7 were found to ameliorate DSS-induced colitis in mice. Additionally, spray-dried powders of CFS from the three strains were examined in a weaned piglet model, only CFS powder of L. reuteri G7 could ameliorate the feed/gain ratio and serum levels of D-lactate and endotoxin. In conclusion, a new potential probiotic strain, L. reuteri G7, was selected and showed ameliorating effects in both colitis mouse and weaned piglet models.
Collapse
Affiliation(s)
- Solomon Abrehame
- Department of Animal Science, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
- Ethiopian Agricultural Authority, Ministry of Agriculture of Ethiopia (MoA), P.O. Box 62347, Addis Ababa 1000, Ethiopia
| | - Man-Yun Hung
- Department of Animal Science, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
| | - Yu-Yi Chen
- Department of Animal Science, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
| | - Yu-Tse Liu
- Department of Animal Science, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
| | - Yung-Tsung Chen
- Department of Food Science, National Taiwan Ocean University, 2 Beining Road, Zhongzheng District, Keelung City 202, Taiwan
| | - Fang-Chueh Liu
- Animal Nutrition Division, Taiwan Livestock Research Institute, Ministry of Agriculture, 112 Farm Road, HsinHua District, Tainan City 712, Taiwan
| | - Yu-Chun Lin
- Animal Nutrition Division, Taiwan Livestock Research Institute, Ministry of Agriculture, 112 Farm Road, HsinHua District, Tainan City 712, Taiwan
- Fisheries Research Institute, Ministry of Agriculture, 199 Hou-Ih Road, Keelung City 202, Taiwan
| | - Yen-Po Chen
- Department of Animal Science, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, 145 Xingda Road, South District, Taichung City 402, Taiwan
| |
Collapse
|
3
|
Wuerger LT, Birkholz G, Oberemm A, Sieg H, Braeuning A. Proteomic analysis of hepatic effects of okadaic acid in HepaRG human liver cells. EXCLI JOURNAL 2023; 22:1135-1145. [PMID: 38054204 PMCID: PMC10694344 DOI: 10.17179/excli2023-6458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/18/2023] [Indexed: 12/07/2023]
Abstract
The marine biotoxin okadaic acid (OA) is produced by dinoflagellates and enters the human food chain by accumulating in the fatty tissue of filter-feeding shellfish. Consumption of highly contaminated shellfish can lead to diarrheic shellfish poisoning. However, apart from the acute effects in the intestine, OA can also provoke toxic effects in the liver, as it is able to pass the intestinal barrier into the blood stream. However, molecular details of OA-induced hepatotoxicity are still insufficiently characterized, and especially at the proteomic level data are scarce. In this study, we used human HepaRG liver cells and exposed them to non-cytotoxic OA concentrations for 24 hours. Global changes in protein expression were analyzed using 2-dimensional gel electrophoresis in combination with mass-spectrometric protein identification. The results constitute the first proteomic analysis of OA effects in human liver cells and indicate, amongst others, that OA affects the energy homeostasis, induces oxidative stress, and induces cytoskeletal changes.
Collapse
Affiliation(s)
- Leonie T.D. Wuerger
- German Federal Institute for Risk Assessment, Department of Food Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| | - Greta Birkholz
- German Federal Institute for Risk Assessment, Department of Food Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| | - Axel Oberemm
- German Federal Institute for Risk Assessment, Department of Food Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| | - Holger Sieg
- 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
| |
Collapse
|
4
|
Wuerger LTD, Kudiabor F, Alarcan J, Templin M, Poetz O, Sieg H, Braeuning A. Okadaic Acid Activates JAK/STAT Signaling to Affect Xenobiotic Metabolism in HepaRG Cells. Cells 2023; 12:770. [PMID: 36899906 PMCID: PMC10000888 DOI: 10.3390/cells12050770] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Okadaic acid (OA) is a marine biotoxin that is produced by algae and accumulates in filter-feeding shellfish, through which it enters the human food chain, leading to diarrheic shellfish poisoning (DSP) after ingestion. Furthermore, additional effects of OA have been observed, such as cytotoxicity. Additionally, a strong downregulation of the expression of xenobiotic-metabolizing enzymes in the liver can be observed. The underlying mechanisms of this, however, remain to be examined. In this study, we investigated a possible underlying mechanism of the downregulation of cytochrome P450 (CYP) enzymes and the nuclear receptors pregnane X receptor (PXR) and retinoid-X-receptor alpha (RXRα) by OA through NF-κB and subsequent JAK/STAT activation in human HepaRG hepatocarcinoma cells. Our data suggest an activation of NF-κB signaling and subsequent expression and release of interleukins, which then activate JAK-dependent signaling and thus STAT3. Moreover, using the NF-κB inhibitors JSH-23 and Methysticin and the JAK inhibitors Decernotinib and Tofacitinib, we were also able to demonstrate a connection between OA-induced NF-κB and JAK signaling and the downregulation of CYP enzymes. Overall, we provide clear evidence that the effect of OA on the expression of CYP enzymes in HepaRG cells is regulated through NF-κB and subsequent JAK signaling.
Collapse
Affiliation(s)
- Leonie T. D. Wuerger
- Department of Food Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| | - Felicia Kudiabor
- 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
| | - Markus Templin
- NMI Natural and Medical Sciences Institute, Markwiesenstraße 55, 72770 Reutlingen, Germany
| | - Oliver Poetz
- NMI Natural and Medical Sciences Institute, Markwiesenstraße 55, 72770 Reutlingen, Germany
- SIGNATOPE GmbH, Markwiesenstraße 55, 72770 Reutlingen, Germany
| | - Holger Sieg
- 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
| |
Collapse
|
5
|
Lee HJ, Mun SJ, Jung CR, Kang HM, Kwon JE, Ryu JS, Ahn HS, Kwon OS, Ahn J, Moon KS, Son MJ, Chung KS. In vitro modeling of liver fibrosis with 3D co-culture system using a novel human hepatic stellate cell line. Biotechnol Bioeng 2023; 120:1241-1253. [PMID: 36639871 DOI: 10.1002/bit.28333] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 12/28/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
Hepatic stellate cells (HSCs) play an important role in liver fibrosis; however, owing to the heterogeneity and limited supply of primary HSCs, the development of in vitro liver fibrosis models has been impeded. In this study, we established and characterized a novel human HSC line (LSC-1), and applied it to various types of three-dimensional (3D) co-culture systems with differentiated HepaRG cells. Furthermore, we compared LSC-1 with a commercially available HSC line on conventional monolayer culture. LSC-1 exhibited an overall upregulation of the expression of fibrogenic genes along with increased levels of matrix and adhesion proteins, suggesting a myofibroblast-like or transdifferentiated state. However, activated states reverted to a quiescent-like phenotype when cultured in different 3D culture formats with a relatively soft microenvironment. Additionally, LSC-1 exerted an overall positive effect on co-cultured differentiated HepaRG, which significantly increased hepatic functionality upon long-term cultivation compared with that achieved with other HSC line. In 3D spheroid culture, LSC-1 exhibited enhanced responsiveness to transforming growth factor beta 1 exposure that is caused by a different matrix-related protein expression mechanism. Therefore, the LSC-1 line developed in this study provides a reliable candidate model that can be used to address unmet needs, such as development of antifibrotic therapies.
Collapse
Affiliation(s)
- Ho-Joon Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Seon Ju Mun
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Department of Functional Genomics, Korea University of Science & Technology (UST), Daejeon, Republic of Korea
| | - Cho-Rok Jung
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Department of Functional Genomics, Korea University of Science & Technology (UST), Daejeon, Republic of Korea
| | - Hyun-Mi Kang
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Jae-Eun Kwon
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Department of Functional Genomics, Korea University of Science & Technology (UST), Daejeon, Republic of Korea
| | - Jae-Sung Ryu
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Hyo-Suk Ahn
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Ok-Seon Kwon
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Jiwon Ahn
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Kyung-Sik Moon
- General and Applied Toxicology Research Center, Korea Institute of Toxicology (KIT), Daejeon, Republic of Korea
| | - Myung Jin Son
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Department of Functional Genomics, Korea University of Science & Technology (UST), Daejeon, Republic of Korea
| | - Kyung-Sook Chung
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Department of Functional Genomics, Korea University of Science & Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
6
|
Mazzari ALDA, Lacerda MG, Milton FA, Mulin Montechiari Machado JA, Sinoti SBP, Toullec AS, Rodrigues PM, Neves FDAR, Simeoni LA, Silveira D, Prieto JM. In vitro effects of European and Latin-American medicinal plants in CYP3A4 gene expression, glutathione levels, and P-glycoprotein activity. Front Pharmacol 2022; 13:826395. [PMID: 36278236 PMCID: PMC9579425 DOI: 10.3389/fphar.2022.826395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 08/31/2022] [Indexed: 11/18/2022] Open
Abstract
Many medicinal plants species from European -such as Artemisia absinthium, Equisetum arvense, Lamium album, Malva sylvestris, Morus nigra, Passiflora incarnata, Frangula purshiana, and Salix alba- as well as Latin American traditions -such as Libidibia ferrea, Bidens pilosa, Casearia sylvestris, Costus spicatus, Monteverdia ilicifolia, Persea americana, Schinus terebinthifolia, Solidago chilensis, Syzygium cumini, Handroanthus impetiginosus, and Vernonanthura phosphorica- are shortlisted by the Brazilian National Health System for future clinical use. However, they lack many data on their action upon some key ADME targets. In this study, we assess non-toxic concentrations (up to100 μg/ml) of their infusions for in vitro ability to modulate CYP3A4 mRNA gene expression and intracellular glutathione levels in HepG2 cells, as well as P-glycoprotein (P-gp) activity in vincristine-resistant Caco-2 cells (Caco-2 VCR). We further investigated the activation of human pregnane X receptor (hPXR) in transiently co-transfected HeLa cells and the inhibition of Gamma-glutamyl transferase (GGT) in HepG2 cells. Our results demonstrate L. ferrea, C. sylvestris, M. ilicifolia, P. americana, S. terebinthifolia, S. cumini, V. phosphorica, E. arvense, P. incarnata, F. purshiana, and S. alba can significantly increase CYP3A4 mRNA gene expression in HepG2 cells. Only F. purshiana shown to do so likely via hPXR activation. P-gp activity was affected by L. ferrea, F. purshiana, S. terebinthifolia, and S. cumini. Total intracellular glutathione levels were significantly depleted by exposure to all extracts except S. alba and S. cumini This was accompanied by a lower GGT activity in the case of C. spicatus, P. americana, S. alba, and S. terebinthifolia, whilst L. ferrea, P. incarnata and F. purshiana increased it. Surprisingly, S. cumini aqueous extract drastically decreased GGT activity (−48%, p < 0.01). In conclusion, this preclinical study shows that the administration of some of these herbal medicines causes in vitro disturbances to key drug metabolism mechanisms. We recommend active pharmacovigilance for Libidibia ferrea (Mart.) L. P. Queiroz, Frangula purshiana Cooper, Schinus terebinthifolia Raddi, and Salix alba L. which were able to alter all targets in our preclinical study.
Collapse
Affiliation(s)
| | | | - Flora Aparecida Milton
- Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
- Instituto de Saúde de Nova Friburgo, Universidade Federal Fluminense, Niterói, Brazil
| | | | | | | | | | - Francisco de Assis Rocha Neves
- Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
- *Correspondence: Francisco de Assis Rocha Neves, ; Dâmaris Silveira, ; Jose Maria Prieto,
| | | | - Dâmaris Silveira
- Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
- *Correspondence: Francisco de Assis Rocha Neves, ; Dâmaris Silveira, ; Jose Maria Prieto,
| | - Jose Maria Prieto
- School of Pharmacy, University College London, London, United Kingdom
- Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
- *Correspondence: Francisco de Assis Rocha Neves, ; Dâmaris Silveira, ; Jose Maria Prieto,
| |
Collapse
|
7
|
In Vitro Models for Studying Chronic Drug-Induced Liver Injury. Int J Mol Sci 2022; 23:ijms231911428. [PMID: 36232728 PMCID: PMC9569683 DOI: 10.3390/ijms231911428] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/08/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
Drug-induced liver injury (DILI) is a major clinical problem in terms of patient morbidity and mortality, cost to healthcare systems and failure of the development of new drugs. The need for consistent safety strategies capable of identifying a potential toxicity risk early in the drug discovery pipeline is key. Human DILI is poorly predicted in animals, probably due to the well-known interspecies differences in drug metabolism, pharmacokinetics, and toxicity targets. For this reason, distinct cellular models from primary human hepatocytes or hepatoma cell lines cultured as 2D monolayers to emerging 3D culture systems or the use of multi-cellular systems have been proposed for hepatotoxicity studies. In order to mimic long-term hepatotoxicity in vitro, cell models, which maintain hepatic phenotype for a suitably long period, should be used. On the other hand, repeated-dose administration is a more relevant scenario for therapeutics, providing information not only about toxicity, but also about cumulative effects and/or delayed responses. In this review, we evaluate the existing cell models for DILI prediction focusing on chronic hepatotoxicity, highlighting how better characterization and mechanistic studies could lead to advance DILI prediction.
Collapse
|
8
|
Dehelean CA, Coricovac D, Pinzaru I, Marcovici I, Macasoi IG, Semenescu A, Lazar G, Cinta Pinzaru S, Radulov I, Alexa E, Cretu O. Rutin bioconjugates as potential nutraceutical prodrugs: An in vitro and in ovo toxicological screening. Front Pharmacol 2022; 13:1000608. [PMID: 36210849 PMCID: PMC9538480 DOI: 10.3389/fphar.2022.1000608] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022] Open
Abstract
Rutin (RUT) is considered one the most attractive flavonoids from a therapeutic perspective due to its multispectral pharmacological activities including antiradical, anti-inflammatory, antiproliferative, and antimetastatic among others. Still, this compound presents a low bioavailability what narrows its clinical applications. To overcome this inconvenience, the current paper was focused on the synthesis, characterization, and toxicological assessment of two RUT bioconjugates obtained by enzymatic esterification with oleic acid (OA) and linoleic acid (LA)—rutin oleate (RUT-O) and rutin linoleate (RUT-L), as flavonoid precursors with improved physicochemical and biological properties. Following the enzymatic synthesis in the presence of Novozyme® 435, the two bioconjugates were obtained, their formation being confirmed by RAMAN and FT-IR spectroscopy. The in vitro and in ovo toxicological assessment of RUT bioconjugates (1–100 µM) was performed using 2D consecrated cell lines (cardiomyoblasts - H9c2(2-1), hepatocytes—HepaRG, and keratinocytes—HaCaT), 3D reconstructed human epidermis tissue (EpiDerm™), and chick chorioallantoic membranes, respectively. The results obtained were test compound, concentration—and cell-type dependent, as follows: RUT-O reduced the viability of H9c2(2-1), HepaRG, and HaCaT cells at 100 µM (to 77.53%, 83.17%, and 78.32%, respectively), and induced cell rounding and floating, as well as apoptotic-like features in the nuclei of all cell lines, whereas RUT-L exerted no signs of cytotoxicity in all cell lines in terms of cell viability, morphology, and nuclear integrity. Both RUT esters impaired the migration of HepaRG cells (at 25 µM) and lack irritative potential (at 100 µM) in vitro (tissue viability >50%) and in ovo (irritation scores of 0.70 for RUT-O, and 0.49 for RUT-L, respectively). Computational predictions revealed an increased lipophilicity, and reduced solubility, drug-likeness and drug score of RUT-O and RUT-L compared to their parent compounds—RUT, OA, and LA. In conclusion, we report a favorable toxicological profile for RUT-L, while RUT-O is dosage-limited since at high concentrations were noticed cytotoxic effects.
Collapse
Affiliation(s)
- Cristina Adriana Dehelean
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, “Victor Babes” University of Medicine and Pharmacy Timisoara, Timisoara, Romania
- Faculty of Food Engineering, Banat's University of Agricultural Sciences and Veterinary Medicine “King Michael I of România”, Timişoara, Romania
| | - Dorina Coricovac
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, “Victor Babes” University of Medicine and Pharmacy Timisoara, Timisoara, Romania
| | - Iulia Pinzaru
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, “Victor Babes” University of Medicine and Pharmacy Timisoara, Timisoara, Romania
- *Correspondence: Iulia Pinzaru,
| | - Iasmina Marcovici
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, “Victor Babes” University of Medicine and Pharmacy Timisoara, Timisoara, Romania
| | - Ioana Gabriela Macasoi
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, “Victor Babes” University of Medicine and Pharmacy Timisoara, Timisoara, Romania
| | - Alexandra Semenescu
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, “Victor Babes” University of Medicine and Pharmacy Timisoara, Timisoara, Romania
| | - Geza Lazar
- ”Ioan Ursu” Institute of the Faculty of Physics, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Simona Cinta Pinzaru
- ”Ioan Ursu” Institute of the Faculty of Physics, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Isidora Radulov
- Faculty of Agriculture, Banat's University of Agricultural Sciences and Veterinary Medicine “King Michael I of România”, Timişoara, Romania
| | - Ersilia Alexa
- Faculty of Food Engineering, Banat's University of Agricultural Sciences and Veterinary Medicine “King Michael I of România”, Timişoara, Romania
| | - Octavian Cretu
- Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy Timisoara, Timisoara, Romania
| |
Collapse
|
9
|
Wuerger LT, Hammer HS, Hofmann U, Kudiabor F, Sieg H, Braeuning A. Okadaic acid influences xenobiotic metabolism in HepaRG cells. EXCLI JOURNAL 2022; 21:1053-1065. [PMID: 36172076 PMCID: PMC9489895 DOI: 10.17179/excli2022-5033] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/28/2022] [Indexed: 11/10/2022]
Abstract
Okadaic acid (OA) is an algae-produced lipophilic marine biotoxin that accumulates in the fatty tissue of filter-feeding shellfish. Ingestion of contaminated shellfish leads to the diarrheic shellfish poisoning syndrome. Furthermore, several other effects of OA like genotoxicity, liver toxicity and tumor-promoting properties have been observed, probably linked to the phosphatase-inhibiting properties of the toxin. It has been shown that at high doses OA can disrupt the physical barrier of the intestinal epithelium. As the intestine and the liver do not only constitute a physical, but also a metabolic barrier against xenobiotic exposure, we here investigated the impact of OA on the expression of cytochrome P450 (CYP) enzymes and transporter proteins in human HepaRG cells liver cells in vitro at non-cytotoxic concentrations. The interplay of OA with known CYP inducers was also studied. Data show that the expression of various xenobiotic-metabolizing CYPs was downregulated after exposure to OA. Moreover, OA was able to counteract the activation of CYPs by their inducers. A number of transporters were also mainly downregulated. Overall, we demonstrate that OA has a significant effect on xenobiotic metabolism barrier in liver cells, highlighting the possibility for interactions of OA exposure with the metabolism of drugs and xenobiotics.
Collapse
Affiliation(s)
- Leonie T.D. Wuerger
- German Federal Institute for Risk Assessment, Department of Food Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| | - Helen S. Hammer
- SIGNATOPE GmbH, Markwiesenstraße 55, 72770 Reutlingen, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstr. 112, 70376 Stuttgart, and University of Tübingen, 72074 Tübingen, Germany
| | - Felicia Kudiabor
- German Federal Institute for Risk Assessment, Department of Food Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| | - Holger Sieg
- German Federal Institute for Risk Assessment, Department of Food Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany,*To whom correspondence should be addressed: Holger Sieg, German Federal Institute for Risk Assessment, Department of Food Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany, E-mail:
| | - Albert Braeuning
- German Federal Institute for Risk Assessment, Department of Food Safety, Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| |
Collapse
|
10
|
Three-Dimensional Liver Culture Systems to Maintain Primary Hepatic Properties for Toxicological Analysis In Vitro. Int J Mol Sci 2021; 22:ijms221910214. [PMID: 34638555 PMCID: PMC8508724 DOI: 10.3390/ijms221910214] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/15/2021] [Accepted: 09/19/2021] [Indexed: 12/13/2022] Open
Abstract
Drug-induced liver injury (DILI) is the major reason for failures in drug development and withdrawal of approved drugs from the market. Two-dimensional cultures of hepatocytes often fail to reliably predict DILI: hepatoma cell lines such as HepG2 do not reflect important primary-like hepatic properties and primary human hepatocytes (pHHs) dedifferentiate quickly in vitro and are, therefore, not suitable for long-term toxicity studies. More predictive liver in vitro models are urgently required in drug development and compound safety evaluation. This review discusses available human hepatic cell types for in vitro toxicology analysis and their usage in established and emerging three-dimensional (3D) culture systems. Generally, 3D cultures maintain or improve primary hepatic functions (including expression of drug-metabolizing enzymes) of different liver cells for several weeks of culture, thus allowing long-term and repeated-dose toxicity studies. Spheroid cultures of pHHs have been comprehensively tested, but also other cell types such as HepaRG benefit from 3D culture systems. Emerging 3D culture techniques include usage of induced pluripotent stem-cell-derived hepatocytes and primary-like upcyte cells, as well as advanced culture techniques such as microfluidic liver-on-a-chip models. In-depth characterization of existing and emerging 3D hepatocyte technologies is indispensable for successful implementation of such systems in toxicological analysis.
Collapse
|
11
|
Duivenvoorde LPM, Louisse J, Pinckaers NET, Nguyen T, van der Zande M. Comparison of gene expression and biotransformation activity of HepaRG cells under static and dynamic culture conditions. Sci Rep 2021; 11:10327. [PMID: 33990636 PMCID: PMC8121841 DOI: 10.1038/s41598-021-89710-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 04/27/2021] [Indexed: 11/08/2022] Open
Abstract
Flow conditions have been shown to be important in improving longevity and functionality of primary hepatocytes, but the impact of flow on HepaRG cells is largely unknown. We studied the expression of genes encoding CYP enzymes and transporter proteins and CYP1 and CYP3A4 activity during 8 weeks of culture in HepaRG cells cultured under static conditions (conventional 24-/96-well plate culture with common bicarbonate/CO2 buffering) and under flow conditions in an organ-on-chip (OOC) device. Since the OOC-device is a closed system, bicarbonate/CO2 buffering was not possible, requiring application of another buffering agent, such as HEPES. In order to disentangle the effects of HEPES from the effects of flow, we also applied HEPES-supplemented medium in static cultures and studied gene expression and CYP activity. We found that cells cultured under flow conditions in the OOC-device, as well as cells cultured under static conditions with HEPES-supplemented medium, showed more stable gene expression levels. Furthermore, only cells cultured in the OOC-device showed relatively high baseline CYP1 activity, and their gene expression levels of selected CYPs and transporters were most similar to gene expression levels in human primary hepatocytes. However, there was a decrease in baseline CYP3A4 activity under flow conditions compared to HepaRG cells cultured under static conditions. Altogether, the present study shows that HepaRG cells cultured in the OOC-device were more stable than in static cultures, being a promising in vitro model to study hepatoxicity of chemicals upon chronic exposure.
Collapse
Affiliation(s)
- Loes P M Duivenvoorde
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands.
| | - Jochem Louisse
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| | - Nicole E T Pinckaers
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| | - Tien Nguyen
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| | - Meike van der Zande
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| |
Collapse
|
12
|
Qiao S, Feng S, Wu Z, He T, Ma C, Peng Z, Tian E, Pan G. Functional Proliferating Human Hepatocytes: In Vitro Hepatocyte Model for Drug Metabolism, Excretion, and Toxicity. Drug Metab Dispos 2021; 49:305-313. [PMID: 33526515 DOI: 10.1124/dmd.120.000275] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
To develop a functional alternative hepatocyte model for primary human hepatocytes (PHHs) with proliferative property, essential drug metabolic, and transporter functions, proliferating human hepatocytes (ProliHHs) expanded from PHHs were fully characterized in vitro. Herein, ProliHHs generated from multiple PHHs donors could be expanded more than 200-fold within four passages and maintained their metabolic or transporter capacities partially. Furthermore, ProliHHs were able to regain the mature hepatic property after three-dimensional (3D) culture. Particularly, the downregulated mRNA expression and function of three major cytochrome P450 (P450) enzymes (CYP1A2, CYP2B6, and CYP3A4) in the proliferating process (ProliHHs-P) could be recovered by 3D culture. The metabolic variabilities across different PHHs donors could be inherited to their matured ProliHHs (ProliHHs-M). The intrinsic clearances of seven major P450 enzymes in ProliHHs-M correlated well (r = 0.87) with those in PHHs. Also, bile canaliculi structures could be observed in sandwich-cultured ProliHHs (SC-ProliHHs), and the biliary excretion index of four probe compounds [cholyl-lys-fluorescein, 5-(and-6)-carboxy-2', 7'-dichlorofluorescein diacetate (CDF), deuterium-labeled sodium taurocholate acid, and rosuvastatin] in SC-ProliHHs (>10%) were close to sandwich-cultured PHHs. More importantly, both ProliHHs-P and ProliHHs-M could be used to evaluate hepatotoxicity. Therefore, these findings demonstrated that the 3D and sandwich culture system could be used to recover the metabolic and transporter functions in ProliHHs for clearance prediction and cholestasis risk assessment, respectively. Together, ProliHHs could be a promising substitute for PHHs in drug metabolism, transport, and hepatotoxicity screening. SIGNIFICANCE STATEMENT: This report describes the study of drug metabolic capacities, efflux transporter functions, and toxicity assessments of proliferating human hepatocytes (ProliHHs). The metabolic variability in different primary human hepatocyte donors could be inherited by their matured ProliHHs derivatives. Also, ProliHHs could form canalicular networks in sandwich culture and display biliary excretion capacities. More importantly, both the proliferative and maturation statuses of ProliHHs could be used to evaluate hepatotoxicity. Together, ProliHHs were feasible to support drug candidate screening in hepatic metabolism, disposition, and toxicity.
Collapse
Affiliation(s)
- Shida Qiao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (S.Q., Z.W., C.M., Z.P., G.P.); University of Chinese Academy of Sciences, Beijing, China (S.Q., Z.W., C.M., Z.P., G.P.); Shanghai Hexaell Biotech Co., Ltd, Shanghai, China (S.F., E.T.); Nanjing University of Chinese Medicine, Nanjing, China (Z.W.); and Nanjing Tech University, Nanjing, China (T.H.)
| | - Sisi Feng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (S.Q., Z.W., C.M., Z.P., G.P.); University of Chinese Academy of Sciences, Beijing, China (S.Q., Z.W., C.M., Z.P., G.P.); Shanghai Hexaell Biotech Co., Ltd, Shanghai, China (S.F., E.T.); Nanjing University of Chinese Medicine, Nanjing, China (Z.W.); and Nanjing Tech University, Nanjing, China (T.H.)
| | - Zhitao Wu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (S.Q., Z.W., C.M., Z.P., G.P.); University of Chinese Academy of Sciences, Beijing, China (S.Q., Z.W., C.M., Z.P., G.P.); Shanghai Hexaell Biotech Co., Ltd, Shanghai, China (S.F., E.T.); Nanjing University of Chinese Medicine, Nanjing, China (Z.W.); and Nanjing Tech University, Nanjing, China (T.H.)
| | - Ting He
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (S.Q., Z.W., C.M., Z.P., G.P.); University of Chinese Academy of Sciences, Beijing, China (S.Q., Z.W., C.M., Z.P., G.P.); Shanghai Hexaell Biotech Co., Ltd, Shanghai, China (S.F., E.T.); Nanjing University of Chinese Medicine, Nanjing, China (Z.W.); and Nanjing Tech University, Nanjing, China (T.H.)
| | - Chen Ma
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (S.Q., Z.W., C.M., Z.P., G.P.); University of Chinese Academy of Sciences, Beijing, China (S.Q., Z.W., C.M., Z.P., G.P.); Shanghai Hexaell Biotech Co., Ltd, Shanghai, China (S.F., E.T.); Nanjing University of Chinese Medicine, Nanjing, China (Z.W.); and Nanjing Tech University, Nanjing, China (T.H.)
| | - Zhaoliang Peng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (S.Q., Z.W., C.M., Z.P., G.P.); University of Chinese Academy of Sciences, Beijing, China (S.Q., Z.W., C.M., Z.P., G.P.); Shanghai Hexaell Biotech Co., Ltd, Shanghai, China (S.F., E.T.); Nanjing University of Chinese Medicine, Nanjing, China (Z.W.); and Nanjing Tech University, Nanjing, China (T.H.)
| | - E Tian
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (S.Q., Z.W., C.M., Z.P., G.P.); University of Chinese Academy of Sciences, Beijing, China (S.Q., Z.W., C.M., Z.P., G.P.); Shanghai Hexaell Biotech Co., Ltd, Shanghai, China (S.F., E.T.); Nanjing University of Chinese Medicine, Nanjing, China (Z.W.); and Nanjing Tech University, Nanjing, China (T.H.)
| | - Guoyu Pan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (S.Q., Z.W., C.M., Z.P., G.P.); University of Chinese Academy of Sciences, Beijing, China (S.Q., Z.W., C.M., Z.P., G.P.); Shanghai Hexaell Biotech Co., Ltd, Shanghai, China (S.F., E.T.); Nanjing University of Chinese Medicine, Nanjing, China (Z.W.); and Nanjing Tech University, Nanjing, China (T.H.)
| |
Collapse
|
13
|
Schmidt K, Berg J, Roehrs V, Kurreck J, Al-Zeer MA. 3D-bioprinted HepaRG cultures as a model for testing long term aflatoxin B1 toxicity in vitro. Toxicol Rep 2020; 7:1578-1587. [PMID: 33304827 PMCID: PMC7708771 DOI: 10.1016/j.toxrep.2020.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/27/2022] Open
Abstract
In recent years 3D-bioprinting technology has been developed as an alternative to animal testing. It possesses a great potential for in vitro testing as it aims to mimic human organs and physiology. In the present study, an alginate-gelatin-Matrigel based hydrogel was used to prepare 3D-bioprinted HepaRG cultures using a pneumatic extrusion printer. These 3D models were tested for viability and metabolic functions. Using 3D-bioprinted HepaRG cultures, we tested the toxicity of aflatoxin B1 (10 or 20 μM) in vitro and compared the results with 2D HepaRG cultures. There was a dose-dependent toxicity effect on cell viability, reduction of metabolic activity and albumin production. We found that 3D-bioprinted HepaRG cultures are more resistant to aflatoxin B1 treatment than 2D cultures. Although the metabolic activities were reduced upon treatment with aflatoxin B1, the 3D models were still viable and survived longer, up to 3 weeks, than the 2D culture, as visualized by fluorescence microscopy. Furthermore, albumin production recovered slightly in 3D models after one and two weeks of treatment. Taken together, we consider using 3D-bioprinting technology to generate 3D tissue models as an alternative way to study toxicity in vitro and this could also provide a suitable alternative for chronic hepatotoxicity studies in vitro.
Collapse
Affiliation(s)
- Konrad Schmidt
- Department of Applied Biochemistry, Institute of Biotechnology, 4/3-2, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Johanna Berg
- Department of Applied Biochemistry, Institute of Biotechnology, 4/3-2, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Viola Roehrs
- Department of Applied Biochemistry, Institute of Biotechnology, 4/3-2, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Jens Kurreck
- Department of Applied Biochemistry, Institute of Biotechnology, 4/3-2, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Munir A. Al-Zeer
- Department of Applied Biochemistry, Institute of Biotechnology, 4/3-2, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| |
Collapse
|
14
|
Dietrich J, Schindler M, Lampen A, Braeuning A, Hessel-Pras S. Comparison of long-term versus short-term effects of okadaic acid on the apoptotic status of human HepaRG cells. Chem Biol Interact 2020; 317:108937. [PMID: 31926150 DOI: 10.1016/j.cbi.2020.108937] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/14/2019] [Accepted: 01/06/2020] [Indexed: 01/08/2023]
Abstract
The biotoxin okadaic acid (OA) is a lipophilic secondary metabolite of marine microalgae. Therefore, OA accumulates in the fatty tissue of various shellfish and may thus enter the food chain. The ingestion of OA via contaminated marine species can lead to the diarrhetic shellfish poisoning syndrome characterized by the occurrence of a series of acute gastrointestinal symptoms in humans. In addition, genotoxicity and tumor-promoting properties of OA might constitute a long-term threat to human health. In order to deepen our understanding of the molecular effects of OA, we compared long-term (14 d) and short-term (24 h and 48 h) apoptotic effects of the compound on human HepaRG hepatocarcinoma cells. Cells were treated either with single doses for 24 and 48 h, respectively, or seven times over a period of 14 d, so that the cumulated quantities of OA in the long-term approach were equal to the single doses upon short-term treatment. Both short-term treatment scenarios led to the induction of apoptosis. Specific caspase activation assays and transcriptional analysis of mRNAs encoding proteins involved in the regulation of apoptosis suggest that OA-induced apoptosis occurs presumably by activation of the intrinsic apoptotic pathway. In contrast, effects were much less pronounced in case of long-term treatment. This is possibly linked to cellular protective mechanisms against low amounts of toxins, e.g. transporter-mediated efflux. In conclusion, our results show a clear concentration- and time-dependency of OA-mediated apoptotic effects in HepaRG cells and contribute to the elucidation of molecular effects of OA.
Collapse
Affiliation(s)
- Jessica Dietrich
- German Federal Institute for Risk Assessment, Department of Food Safety, 10589, Berlin, Germany
| | - Magdalena Schindler
- German Federal Institute for Risk Assessment, Department of Food Safety, 10589, Berlin, Germany
| | - Alfonso Lampen
- German Federal Institute for Risk Assessment, Department of Food Safety, 10589, Berlin, Germany
| | - Albert Braeuning
- German Federal Institute for Risk Assessment, Department of Food Safety, 10589, Berlin, Germany
| | - Stefanie Hessel-Pras
- German Federal Institute for Risk Assessment, Department of Food Safety, 10589, Berlin, Germany.
| |
Collapse
|
15
|
Ning J, Chen L, Rietjens IM. Role of toxicokinetics and alternative testing strategies in pyrrolizidine alkaloid toxicity and risk assessment; state-of-the-art and future perspectives. Food Chem Toxicol 2019; 131:110572. [DOI: 10.1016/j.fct.2019.110572] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 06/05/2019] [Accepted: 06/07/2019] [Indexed: 01/31/2023]
|
16
|
Okadaic acid activates Wnt/β-catenin-signaling in human HepaRG cells. Arch Toxicol 2019; 93:1927-1939. [PMID: 31115591 DOI: 10.1007/s00204-019-02489-4] [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: 05/06/2019] [Accepted: 05/16/2019] [Indexed: 12/13/2022]
Abstract
The lipophilic phycotoxin okadaic acid (OA) occurs in the fatty tissue and hepatopancreas of filter-feeding shellfish. The compound provokes the diarrhetic shellfish poisoning (DSP) syndrome after intake of seafood contaminated with high levels of the DSP toxin. In animal experiments, long-term exposure to OA is associated with an elevated risk for tumor formation in different organs including the liver. Although OA is a known inhibitor of the serine/threonine protein phosphatase 2A, the mechanisms behind OA-induced carcinogenesis are not fully understood. Here, we investigated the influence of OA on the β-catenin-dependent Wnt-signaling pathway, addressing a major oncogenic pathway relevant for tumor development. We analyzed OA-mediated effects on β-catenin and its biological function, cellular localization, post-translational modifications, and target gene expression in human HepaRG hepatocarcinoma cells treated with non-cytotoxic concentrations up to 50 nM. We detected concentration- and time-dependent effects of OA on the phosphorylation state, cellular redistribution as well as on the amount of transcriptionally active β-catenin. These findings were confirmed by quantitative live-cell imaging of U2OS cells stably expressing a green fluorescent chromobody which specifically recognize hypophosphorylated β-catenin. Finally, we demonstrated that nuclear translocation of β-catenin mediated by non-cytotoxic OA concentrations results in an upregulation of Wnt-target genes. In conclusion, our results show a significant induction of the canonical Wnt/β-catenin-signaling pathway by OA in human liver cells. Our data contribute to a better understanding of the molecular mechanisms underlying OA-induced carcinogenesis.
Collapse
|
17
|
Ahn J, Lee HJ, Oh SJ, Kim W, Mun SJ, Lee JH, Jung CR, Cho HS, Kim DS, Son MJ, Chung KS. Developing scalable cultivation systems of hepatic spheroids for drug metabolism via genomic and functional analyses. Biotechnol Bioeng 2019; 116:1496-1508. [PMID: 30737956 DOI: 10.1002/bit.26954] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 01/18/2019] [Accepted: 02/07/2019] [Indexed: 12/21/2022]
Abstract
Spheroids, a widely used three-dimensional (3D) culture model, are standard in hepatocyte culture as they preserve long-term hepatocyte functionality and enhance survivability. In this study, we investigated the effects of three operation modes in 3D culture - static, orbital shaking, and under vertical bidirectional flow using spheroid forming units (SFUs) on hepatic differentiation and drug metabolism to propose the best for mass production of functionally enhanced spheroids. Spheroids in SFUs exhibited increased hepatic gene expression, albumin secretion, and cytochrome P450 3A4 (CYP3A4) activity during the differentiation period (12 days). SFUs advantages include facilitated mass production and a relatively earlier peak of CYP3A4 activity. However, CYP3A4 activity was not well maintained under dimethyl sulfoxide (DMSO)-free conditions (13-18 days), dramatically reducing drug metabolism capability. Continued shear stimulation without differentiation stimuli in assay conditions markedly attenuated CYP3A4 activity, which was less severe in static conditions. In this condition, SFU spheroids exhibited dedifferentiation characteristics, such as increased proliferation and Notch signaling genes. We found that the dedifferentiation could be overcome by using the serum-free medium formulation. Therefore, we suggest that SFUs represent the best option for the mass production of functionally improved spheroids and so the serum-free conditions should be maintained during drug metabolism analysis.
Collapse
Affiliation(s)
- Jiwon Ahn
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Ho-Joon Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Soo Jin Oh
- New Drug Development Center, Asan Medical Center and Convergence Medicine, University of Ulsan, Seoul, Republic of Korea
| | - Wantae Kim
- Biomedical Translational Research Center, KRIBB, Daejeon, Republic of Korea
| | - Seon Ju Mun
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Jae-Hye Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Cho-Rock Jung
- Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea.,Gene Therapy Unit, KRIBB, Daejeon, Republic of Korea
| | - Hyun-Soo Cho
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Dae-Soo Kim
- Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea.,Genome Research Center, KRIBB, Daejeon, Republic of Korea
| | - Myung Jin Son
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Kyung-Sook Chung
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Biomedical Translational Research Center, KRIBB, Daejeon, Republic of Korea.,Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
18
|
Tascher G, Burban A, Camus S, Plumel M, Chanon S, Le Guevel R, Shevchenko V, Van Dorsselaer A, Lefai E, Guguen-Guillouzo C, Bertile F. In-Depth Proteome Analysis Highlights HepaRG Cells as a Versatile Cell System Surrogate for Primary Human Hepatocytes. Cells 2019; 8:E192. [PMID: 30795634 PMCID: PMC6406872 DOI: 10.3390/cells8020192] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/16/2019] [Accepted: 02/18/2019] [Indexed: 12/12/2022] Open
Abstract
Of the hepatic cell lines developed for in vitro studies of hepatic functions as alternatives to primary human hepatocytes, many have lost major liver-like functions, but not HepaRG cells. The increasing use of the latter worldwide raises the need for establishing the reference functional status of early biobanked HepaRG cells. Using deep proteome and secretome analyses, the levels of master regulators of the hepatic phenotype and of the structural elements ensuring biliary polarity were found to be close to those in primary hepatocytes. HepaRG cells proved to be highly differentiated, with functional mitochondria, hepatokine secretion abilities, and an adequate response to insulin. Among differences between primary human hepatocytes and HepaRG cells, the factors that possibly support HepaRG transdifferentiation properties are discussed. The HepaRG cell system thus appears as a robust surrogate for primary hepatocytes, which is versatile enough to study not only xenobiotic detoxification, but also the control of hepatic energy metabolism, secretory function and disease-related mechanisms.
Collapse
Affiliation(s)
- Georg Tascher
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS, IPHC UMR 7178, Université de Strasbourg, F-67087 Strasbourg, France.
- Institute of Biochemistry II, Goethe University Hospital, D-60590 Frankfurt am Main, Germany.
| | - Audrey Burban
- INSERM U1241 NuMeCan, Université de Rennes 1, F-35033 Rennes, France.
| | - Sandrine Camus
- Biopredic International, Parc d'Affaires de la Bretêche, F-35760 St Grégoire, France.
| | - Marine Plumel
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS, IPHC UMR 7178, Université de Strasbourg, F-67087 Strasbourg, France.
| | - Stéphanie Chanon
- CarMeN Laboratory, INSERM, INRA, University of Lyon, F-69310 Pierre-Bénite, France.
| | - Remy Le Guevel
- ImPACcell platform, Biosit, Université de Rennes 1, F-35043 Rennes, France.
| | - Valery Shevchenko
- Biopredic International, Parc d'Affaires de la Bretêche, F-35760 St Grégoire, France.
| | - Alain Van Dorsselaer
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS, IPHC UMR 7178, Université de Strasbourg, F-67087 Strasbourg, France.
| | - Etienne Lefai
- CarMeN Laboratory, INSERM, INRA, University of Lyon, F-69310 Pierre-Bénite, France.
| | - Christiane Guguen-Guillouzo
- INSERM U1241 NuMeCan, Université de Rennes 1, F-35033 Rennes, France.
- Biopredic International, Parc d'Affaires de la Bretêche, F-35760 St Grégoire, France.
| | - Fabrice Bertile
- Laboratoire de Spectrométrie de Masse BioOrganique, CNRS, IPHC UMR 7178, Université de Strasbourg, F-67087 Strasbourg, France.
| |
Collapse
|
19
|
A serum-free medium suitable for maintaining cell morphology and liver-specific function in induced human hepatocytes. Cytotechnology 2019; 71:329-344. [PMID: 30603919 DOI: 10.1007/s10616-018-0289-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 12/05/2018] [Indexed: 12/15/2022] Open
Abstract
hiHep is a new type of hepatocyte-like cell that is predicted to be a potential unlimited source of hepatocytes for a bioartificial liver. However, hiHep cannot currently be used in clinical settings because serum must be added during the culture process. Thus, a defined medium is required. Because serum is complex, an efficient statistical approach based on the Plackett-Burman design was used. In this manner, an original medium and several significant cell growth factors were identified. These factors include insulin, VH, and VE, and the original medium was optimized based on these significant factors. Additionally, hiHep liver-specific functions and metabolism in the optimized serum-free medium were measured. Results showed that hiHep functions, such as glycogen storage, albumin secretion, and urea production, were well maintained in our optimized serum-free medium. In summary, we created a chemically defined, serum-free medium in which cell growth, proliferation, metabolism, and function were well maintained. This medium has the potential to support the clinical use of hiHep.
Collapse
|
20
|
Han W, Wu Q, Zhang X, Duan Z. Innovation for hepatotoxicity in vitro research models: A review. J Appl Toxicol 2018; 39:146-162. [PMID: 30182494 DOI: 10.1002/jat.3711] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/11/2018] [Accepted: 07/13/2018] [Indexed: 12/18/2022]
Abstract
Many categories of drugs can induce hepatotoxicity, so improving the prediction of toxic drugs is important. In vitro models using human hepatocytes are more accurate than in vivo animal models. Good in vitro models require an abundance of metabolic enzyme activities and normal cellular polarity. However, none of the in vitro models can completely simulate hepatocytes in the human body. There are two ways to overcome this limitation: enhancing the metabolic function of hepatocytes and changing the cultural environment. In this review, we summarize the current state of research, including the main characteristics of in vitro models and their limitations, as well as improved technology and developmental prospects. We hope that this review provides some new ideas for hepatotoxicity research.
Collapse
Affiliation(s)
- Weijia Han
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| | - Qiao Wu
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| | - Xiaohui Zhang
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| | - Zhongping Duan
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| |
Collapse
|
21
|
Coll M, Perea L, Boon R, Leite SB, Vallverdú J, Mannaerts I, Smout A, El Taghdouini A, Blaya D, Rodrigo-Torres D, Graupera I, Aguilar-Bravo B, Chesne C, Najimi M, Sokal E, Lozano JJ, van Grunsven LA, Verfaillie CM, Sancho-Bru P. Generation of Hepatic Stellate Cells from Human Pluripotent Stem Cells Enables In Vitro Modeling of Liver Fibrosis. Cell Stem Cell 2018; 23:101-113.e7. [PMID: 30049452 DOI: 10.1016/j.stem.2018.05.027] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 11/14/2017] [Accepted: 05/30/2018] [Indexed: 12/14/2022]
Abstract
The development of complex in vitro hepatic systems and artificial liver devices has been hampered by the lack of reliable sources for relevant cell types, such as hepatic stellate cells (HSCs). Here we report efficient differentiation of human pluripotent stem cells into HSC-like cells (iPSC-HSCs). iPSC-HSCs closely resemble primary human HSCs at the transcriptional, cellular, and functional levels and possess a gene expression profile intermediate between that of quiescent and activated HSCs. Functional analyses revealed that iPSC-HSCs accumulate retinyl esters in lipid droplets and are activated in response to mediators of wound healing, similar to their in vivo counterparts. When maintained as 3D spheroids with HepaRG hepatocytes, iPSC-HSCs exhibit a quiescent phenotype but mount a fibrogenic response and secrete pro-collagen in response to known stimuli and hepatocyte toxicity. Thus, this protocol provides a robust in vitro system for studying HSC development, modeling liver fibrosis, and drug toxicity screening.
Collapse
Affiliation(s)
- Mar Coll
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Luis Perea
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ruben Boon
- Stem Cell Institute Leuven, Leuven, Belgium
| | - Sofia B Leite
- Liver Cell Biology Lab, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Julia Vallverdú
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Inge Mannaerts
- Liver Cell Biology Lab, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Ayla Smout
- Liver Cell Biology Lab, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Adil El Taghdouini
- Liver Cell Biology Lab, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Delia Blaya
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Daniel Rodrigo-Torres
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Isabel Graupera
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain; Liver Unit, Hospital Clínic, Barcelona, Spain
| | - Beatriz Aguilar-Bravo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCL), Leuven, Belgium
| | - Etienne Sokal
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCL), Leuven, Belgium
| | - Juan José Lozano
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Leo A van Grunsven
- Liver Cell Biology Lab, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium.
| | | | - Pau Sancho-Bru
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain.
| |
Collapse
|
22
|
Cuykx M, Claes L, Rodrigues RM, Vanhaecke T, Covaci A. Metabolomics profiling of steatosis progression in HepaRG ® cells using sodium valproate. Toxicol Lett 2018; 286:22-30. [PMID: 29355688 DOI: 10.1016/j.toxlet.2017.12.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 12/12/2017] [Accepted: 12/20/2017] [Indexed: 02/07/2023]
Abstract
Non-alcoholic Fatty Liver Disease (NAFLD) is a frequently encountered Drug-Induced Liver Injury (DILI). Although this stage of the disease is reversible, it can lead to irreversible damage provoked by non-alcoholic steatohepatitis (NASH), fibrosis and cirrhosis. Therefore, the assessment of NAFLD is a paramount objective in toxicological screenings of new drug candidates. In this study, a metabolomic fingerprint of NAFLD induced in HepaRG® cells at four dosing schemes by a reference toxicant, sodium valproate (NaVPA), was obtained using liquid-liquid extraction followed by liquid chromatography and accurate mass-mass spectrometry (LC-AM/MS). The combination of a strict design of experiment with a robust detection method, applied on sodium valproate, validated the possibilities of untargeted metabolomics in hepatic toxicological research. Distinctive patterns between exposed and control cells were consistently observed, multivariate analyses selected up to 200 features of interest, revealing hallmark NAFLD-biomarkers, such as diacylglycerol and triglyceride accumulation and carnitine deficiency. Initial toxic responses show increased levels of S-adenosylmethionine and mono-acetylspermidine in combination with only a moderate increase in triglycerides. New specific markers of toxicity have been observed, such as spermidines, creatine, and acetylcholine. The described design of experiment provides a valuable metabolomics platform for mechanistic research of toxicological hazards and identified new markers for steatotic progression.
Collapse
Affiliation(s)
- Matthias Cuykx
- Toxicological Centre, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.
| | - Leen Claes
- Toxicological Centre, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Robim M Rodrigues
- Research group In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Jette, Belgium
| | - Tamara Vanhaecke
- Research group In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Jette, Belgium
| | - Adrian Covaci
- Toxicological Centre, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.
| |
Collapse
|
23
|
Ren Z, Chen S, Ning B, Guo L. Use of Liver-Derived Cell Lines for the Study of Drug-Induced Liver Injury. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2018. [DOI: 10.1007/978-1-4939-7677-5_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
24
|
Evaluation of HepaRG cells for the assessment of indirect drug-induced hepatotoxicity using INH as a model substance. Hum Cell 2017; 30:267-278. [PMID: 28527127 DOI: 10.1007/s13577-017-0175-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 05/04/2017] [Indexed: 01/13/2023]
Abstract
HepaRG cells are widely used as an in vitro model to assess drug-induced hepatotoxicity. However, only few studies exist so far regarding their suitability to detect the effects of drugs requiring a preceding activation via the cytochrome P450 (CYP) system. A prototypic substance is the anti-tuberculosis agent INH, which is metabolized into N-acetylhydrazine, which then triggers hepatotoxicity. Therefore, the aim of the present study was to test if this effect can also be detected in HepaRG cells and if it can be counteracted by the known hepatoprotectant silibinin. For this purpose, differentiated HepaRG cells were treated with increasing concentrations of INH (0.1-100 mM) or 10 mM INH plus escalating concentrations of silibinin (1-100 µM). After 48 h of treatment, cell morphology and parameters indicating cell vitality, oxidative stress, and liver cell function were assessed. High concentrations of INH led to severe histopathological changes, reduced cell vitality and glutathione content, increased LDH and ASAT release into the medium, enhanced lipid peroxidation, and elevated cleaved caspase-3 expression. Additionally, glycogen depletion and reduced biotransformation capacity were seen at high INH concentrations, whereas at low concentrations an induction of biotransformation enzymes was noticed. Silibinin caused clear-cut protective effects, but with few parameters INH toxicity was even aggravated, most probably due to increased metabolization of INH into its toxic metabolite. In conclusion, HepaRG cells are excellently suited to evaluate the effects of substances requiring prior toxification via the CYP system, such as INH. They additionally enable the identification of complex substance interactions.
Collapse
|
25
|
Richter LHJ, Flockerzi V, Maurer HH, Meyer MR. Pooled human liver preparations, HepaRG, or HepG2 cell lines for metabolism studies of new psychoactive substances? A study using MDMA, MDBD, butylone, MDPPP, MDPV, MDPB, 5-MAPB, and 5-API as examples. J Pharm Biomed Anal 2017; 143:32-42. [PMID: 28601767 DOI: 10.1016/j.jpba.2017.05.028] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/05/2017] [Accepted: 05/12/2017] [Indexed: 01/16/2023]
Abstract
Metabolism studies play an important role in clinical and forensic toxicology. Because of potential species differences in metabolism, human samples are best suitable for elucidating metabolism. However, in the case of new psychoactive substances (NPS), human samples of controlled studies are not available. Primary human hepatocytes have been described as gold standard for in vitro metabolism studies, but there are some disadvantages such as high costs, limited availability, and variability of metabolic enzymes. Therefore, the aim of our study was to investigate and compare the metabolism of six methylenedioxy derivatives (MDMA, MDBD, butylone, MDPPP, MDPV, MDPB) and two bioisosteric analogues (5-MAPB, 5-API) using pooled human liver microsomes (pHLM) combined with cytosol (pHLC) or pooled human liver S9 fraction (pS9) all after addition of co-substrates for six phase I and II reactions. In addition, HepaRG and HepG2 cell lines were used. Results of the different in vitro tools were compared to each other, to corresponding published data, and to metabolites identified in human urine after consumption of MDMA, MDPV, or 5-MAPB. Incubations with pHLM plus pHLC showed similar results as pS9. A more cost efficient model for prediction of targets for toxicological screening procedures in human urine should be identified. As expected, the incubations with HepaRG provided better results than those with HepG2 concerning number and signal abundance of the metabolites. Due to easy handling without special equipment, incubations with pooled liver preparations should be the most suitable alternative to find targets for toxicological screening procedures for methylenedioxy derivatives and bioisosteric analogues.
Collapse
Affiliation(s)
- Lilian H J Richter
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, 66421 Homburg, Germany
| | - Veit Flockerzi
- Department of Experimental and Clinical Pharmacology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, 66421 Homburg, Germany
| | - Hans H Maurer
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, 66421 Homburg, Germany
| | - Markus R Meyer
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, 66421 Homburg, Germany.
| |
Collapse
|
26
|
Bell CC, Lauschke VM, Vorrink SU, Palmgren H, Duffin R, Andersson TB, Ingelman-Sundberg M. Transcriptional, Functional, and Mechanistic Comparisons of Stem Cell-Derived Hepatocytes, HepaRG Cells, and Three-Dimensional Human Hepatocyte Spheroids as Predictive In Vitro Systems for Drug-Induced Liver Injury. Drug Metab Dispos 2017; 45:419-429. [PMID: 28137721 PMCID: PMC5363699 DOI: 10.1124/dmd.116.074369] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/25/2017] [Indexed: 12/15/2022] Open
Abstract
Reliable and versatile hepatic in vitro systems for the prediction of drug pharmacokinetics and toxicity are essential constituents of preclinical safety assessment pipelines for new medicines. Here, we compared three emerging cell systems—hepatocytes derived from induced pluripotent stem cells, HepaRG cells, and three-dimensional primary human hepatocyte (PHH) spheroids—at transcriptional and functional levels in a multicenter study to evaluate their potential as predictive models for drug-induced hepatotoxicity. Transcriptomic analyses revealed widespread gene expression differences between the three cell models, with 8148 of 17,462 analyzed genes (47%) being differentially expressed. Expression levels of genes involved in the metabolism of endogenous as well as xenobiotic compounds were significantly elevated in PHH spheroids, whereas genes involved in cell division and endocytosis were significantly upregulated in HepaRG cells and hepatocytes derived from induced pluripotent stem cells, respectively. Consequently, PHH spheroids were more sensitive to a panel of drugs with distinctly different toxicity mechanisms, an effect that was amplified by long-term exposure using repeated treatments. Importantly, toxicogenomic analyses revealed that transcriptomic changes in PHH spheroids were in compliance with cholestatic, carcinogenic, or steatogenic in vivo toxicity mechanisms at clinically relevant drug concentrations. Combined, the data reveal important phenotypic differences between the three cell systems and suggest that PHH spheroids can be used for functional investigations of drug-induced liver injury in vivo in humans.
Collapse
Affiliation(s)
- Catherine C Bell
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (C.C.B., V.M.L., S.U.V., T.B.A., M.I.-S.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (H.P., T.B.A.); and CXR Biosciences Ltd., Dundee, United Kingdom (R.D.)
| | - Volker M Lauschke
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (C.C.B., V.M.L., S.U.V., T.B.A., M.I.-S.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (H.P., T.B.A.); and CXR Biosciences Ltd., Dundee, United Kingdom (R.D.)
| | - Sabine U Vorrink
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (C.C.B., V.M.L., S.U.V., T.B.A., M.I.-S.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (H.P., T.B.A.); and CXR Biosciences Ltd., Dundee, United Kingdom (R.D.)
| | - Henrik Palmgren
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (C.C.B., V.M.L., S.U.V., T.B.A., M.I.-S.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (H.P., T.B.A.); and CXR Biosciences Ltd., Dundee, United Kingdom (R.D.)
| | - Rodger Duffin
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (C.C.B., V.M.L., S.U.V., T.B.A., M.I.-S.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (H.P., T.B.A.); and CXR Biosciences Ltd., Dundee, United Kingdom (R.D.)
| | - Tommy B Andersson
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (C.C.B., V.M.L., S.U.V., T.B.A., M.I.-S.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (H.P., T.B.A.); and CXR Biosciences Ltd., Dundee, United Kingdom (R.D.)
| | - Magnus Ingelman-Sundberg
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (C.C.B., V.M.L., S.U.V., T.B.A., M.I.-S.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden (H.P., T.B.A.); and CXR Biosciences Ltd., Dundee, United Kingdom (R.D.)
| |
Collapse
|
27
|
Richter LHJ, Kaminski YR, Noor F, Meyer MR, Maurer HH. Metabolic fate of desomorphine elucidated using rat urine, pooled human liver preparations, and human hepatocyte cultures as well as its detectability using standard urine screening approaches. Anal Bioanal Chem 2016; 408:6283-94. [PMID: 27372715 DOI: 10.1007/s00216-016-9740-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/14/2016] [Accepted: 06/22/2016] [Indexed: 11/29/2022]
Abstract
Desomorphine is an opioid misused as "crocodile", a cheaper alternative to heroin. It is a crude synthesis product homemade from codeine with toxic byproducts. The aim of the present work was to investigate the metabolic fate of desomorphine in vivo using rat urine and in vitro using pooled human liver microsomes and cytosol as well as human liver cell lines (HepG2 and HepaRG) by Orbitrap-based liquid chromatography-high resolution-tandem mass spectrometry or hydrophilic interaction liquid chromatography. According to the identified metabolites, the following metabolic steps could be proposed: N-demethylation, hydroxylation at various positions, N-oxidation, glucuronidation, and sulfation. The cytochrome P450 (CYP) initial activity screening revealed CYP3A4 to be the only CYP involved in all phase I steps. UDP-glucuronyltransferase (UGT) initial activity screening showed that UGT1A1, UGT1A8, UGT1A9, UGT1A10, UGT2B4, UGT2B7, UGT2B15, and UGT2B17 formed desomorphine glucuronide. Among the tested in vitro models, HepaRG cells were identified to be the most suitable tool for prediction of human hepatic phase I and II metabolism of drugs of abuse. Finally, desomorphine (crocodile) consumption should be detectable by all standard urine screening approaches mainly via the parent compound and/or its glucuronide assuming similar kinetics in rats and humans.
Collapse
Affiliation(s)
- Lilian H J Richter
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Saarland University, Kirrberger Strasse 100, 66421, Homburg (Saar), Germany
| | - Yeda Rumi Kaminski
- Biochemical Engineering Institute, Saarland University, Campus A1 5, 66123, Saarbrücken, Germany
| | - Fozia Noor
- Biochemical Engineering Institute, Saarland University, Campus A1 5, 66123, Saarbrücken, Germany
| | - Markus R Meyer
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Saarland University, Kirrberger Strasse 100, 66421, Homburg (Saar), Germany.,Department of Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Hans H Maurer
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Saarland University, Kirrberger Strasse 100, 66421, Homburg (Saar), Germany.
| |
Collapse
|
28
|
Oleaga C, Bernabini C, Smith AS, Srinivasan B, Jackson M, McLamb W, Platt V, Bridges R, Cai Y, Santhanam N, Berry B, Najjar S, Akanda N, Guo X, Martin C, Ekman G, Esch MB, Langer J, Ouedraogo G, Cotovio J, Breton L, Shuler ML, Hickman JJ. Multi-Organ toxicity demonstration in a functional human in vitro system composed of four organs. Sci Rep 2016; 6:20030. [PMID: 26837601 PMCID: PMC4738272 DOI: 10.1038/srep20030] [Citation(s) in RCA: 282] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 12/18/2015] [Indexed: 12/26/2022] Open
Abstract
We report on a functional human model to evaluate multi-organ toxicity in a 4-organ system under continuous flow conditions in a serum-free defined medium utilizing a pumpless platform for 14 days. Computer simulations of the platform established flow rates and resultant shear stress within accepted ranges. Viability of the system was demonstrated for 14 days as well as functional activity of cardiac, muscle, neuronal and liver modules. The pharmacological relevance of the integrated modules were evaluated for their response at 7 days to 5 drugs with known side effects after a 48 hour drug treatment regime. The results of all drug treatments were in general agreement with published toxicity results from human and animal data. The presented phenotypic culture model exhibits a multi-organ toxicity response, representing the next generation of in vitro systems, and constitutes a step towards an in vitro "human-on-a-chip" assay for systemic toxicity screening.
Collapse
Affiliation(s)
- Carlota Oleaga
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Catia Bernabini
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Alec S.T. Smith
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Balaji Srinivasan
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Max Jackson
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - William McLamb
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Vivien Platt
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Richard Bridges
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Yunqing Cai
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Navaneetha Santhanam
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Bonnie Berry
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Sarah Najjar
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Nesar Akanda
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Xiufang Guo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Candace Martin
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Gail Ekman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Mandy B. Esch
- Department of Biomedical Engineering, Cornell University, 115 and 305 Weill Hall, Ithaca, NY 14853
| | - Jessica Langer
- L’Oreal Research and Innovation, Clark, NJ, 07666/ Aulnay sous Bois, France, 93600
| | | | - Jose Cotovio
- L’Oreal Research and Innovation, Aulnay sous Bois, France
| | - Lionel Breton
- L’Oreal Research and Innovation, Aulnay sous Bois, France
| | - Michael L. Shuler
- Department of Biomedical Engineering, Cornell University, 115 and 305 Weill Hall, Ithaca, NY 14853
| | - James J. Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| |
Collapse
|
29
|
Herzog N, Hansen M, Miethbauer S, Schmidtke KU, Anderer U, Lupp A, Sperling S, Seehofer D, Damm G, Scheibner K, Küpper JH. Primary-like human hepatocytes genetically engineered to obtain proliferation competence display hepatic differentiation characteristics in monolayer and organotypical spheroid cultures. Cell Biol Int 2016; 40:341-53. [PMID: 26715207 DOI: 10.1002/cbin.10574] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 12/23/2015] [Indexed: 12/27/2022]
Abstract
Primary human hepatocytes are in great demand during drug development and in hepatology. However, both scarcity of tissue supply and donor variability of primary cells create a need for the development of alternative hepatocyte systems. By using a lentivirus vector system to transfer coding sequences of Upcyte® proliferation genes, we generated non-transformed stable hepatocyte cultures from human liver tissue samples. Here, we show data on newly generated proliferation-competent HepaFH3 cells investigated as conventional two-dimensional monolayer and as organotypical three-dimensional (3D) spheroid culture. In monolayer culture, HepaFH3 cells show typical cobblestone-like hepatocyte morphology and anchorage-dependent growth for at least 20 passages. Immunofluorescence staining revealed that characteristic hepatocyte marker proteins cytokeratin 8, human serum albumin, and cytochrome P450 (CYP) 3A4 were expressed. Quantitative real-time PCR analyses showed that expression levels of analyzed phase I CYP enzymes were at similar levels compared to those of cultured primary human hepatocytes and considerably higher than in the liver carcinoma cell line HepG2. Additionally, transcripts for phase II liver enzymes and transporter proteins OATP-C, MRP2, Oct1, and BSEP were present in HepaFH3. The cells produced urea and converted model compounds such as testosterone, diclofenac, and 7-OH-coumarin into phases I and II metabolites. Interestingly, phases I and II enzymes were expressed at about the same levels in convenient monolayer cultures and complex 3D spheroids. In conclusion, HepaFH3 cells and related primary-like hepatocyte lines seem to be promising tools for in vitro research of liver functions and as test system in drug development and toxicology analysis.
Collapse
Affiliation(s)
- Natalie Herzog
- Faculty of Science, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Max Hansen
- Faculty of Science, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Sebastian Miethbauer
- Faculty of Science, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Kai-Uwe Schmidtke
- Faculty of Science, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Ursula Anderer
- Faculty of Science, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Sebastian Sperling
- Department of General, Visceral and Transplantation Surgery, Charité University Medicine, Berlin, Germany
| | - Daniel Seehofer
- Department of General, Visceral and Transplantation Surgery, Charité University Medicine, Berlin, Germany
| | - Georg Damm
- Department of General, Visceral and Transplantation Surgery, Charité University Medicine, Berlin, Germany
| | - Katrin Scheibner
- Faculty of Science, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Jan-Heiner Küpper
- Faculty of Science, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| |
Collapse
|
30
|
Leite SB, Roosens T, El Taghdouini A, Mannaerts I, Smout AJ, Najimi M, Sokal E, Noor F, Chesne C, van Grunsven LA. Novel human hepatic organoid model enables testing of drug-induced liver fibrosis in vitro. Biomaterials 2015; 78:1-10. [PMID: 26618472 DOI: 10.1016/j.biomaterials.2015.11.026] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 10/30/2015] [Accepted: 11/13/2015] [Indexed: 12/14/2022]
Abstract
Current models for in vitro fibrosis consist of simple mono-layer cultures of rodent hepatic stellate cells (HSC), ignoring the role of hepatocyte injury. We aimed to develop a method allowing the detection of hepatocyte-mediated and drug-induced liver fibrosis. We used HepaRG (Hep) and primary human HSCs cultured as 3D spheroids in 96-well plates. These resulting scaffold-free organoids were characterized for CYP induction, albumin secretion, and hepatocyte and HSC-specific gene expression by qPCR. The metabolic competence of the organoid over 21 days allows activation of HSCs in the organoid in a drug- and hepatocyte-dependent manner. After a single dose or repeated exposure for 14 days to the pro-fibrotic compounds Allyl alcohol and Methotrexate, hepatic organoids display fibrotic features such as HSC activation, collagen secretion and deposition. Acetaminophen was identified by these organoids as an inducer of hepatotoxic-mediated HSC activation which was confirmed in vivo in mice. This novel hepatic organoid culture model is the first that can detect hepatocyte-dependent and compound-induced HSC activation, thereby representing an important step forward towards in vitro compound testing for drug-induced liver fibrosis.
Collapse
Affiliation(s)
- Sofia B Leite
- Liver Cell Biology Laboratory, Vrije Universiteit Brussel (VUB), Belgium.
| | - Tiffany Roosens
- Liver Cell Biology Laboratory, Vrije Universiteit Brussel (VUB), Belgium
| | - Adil El Taghdouini
- Liver Cell Biology Laboratory, Vrije Universiteit Brussel (VUB), Belgium
| | - Inge Mannaerts
- Liver Cell Biology Laboratory, Vrije Universiteit Brussel (VUB), Belgium
| | - Ayla J Smout
- Liver Cell Biology Laboratory, Vrije Universiteit Brussel (VUB), Belgium
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCL), Belgium
| | - Etienne Sokal
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCL), Belgium
| | - Fozia Noor
- Biochemical Engineering Institute, Saarland University, Germany
| | | | - Leo A van Grunsven
- Liver Cell Biology Laboratory, Vrije Universiteit Brussel (VUB), Belgium.
| |
Collapse
|
31
|
Klein S, Maggioni S, Bucher J, Mueller D, Niklas J, Shevchenko V, Mauch K, Heinzle E, Noor F. In Silico Modeling for the Prediction of Dose and Pathway-Related Adverse Effects in Humans From In Vitro Repeated-Dose Studies. Toxicol Sci 2015; 149:55-66. [PMID: 26420750 DOI: 10.1093/toxsci/kfv218] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Long-term repeated-dose toxicity is mainly assessed in animals despite poor concordance of animal data with human toxicity. Nowadays advanced human in vitro systems, eg, metabolically competent HepaRG cells, are used for toxicity screening. Extrapolation of in vitro toxicity to in vivo effects is possible by reverse dosimetry using pharmacokinetic modeling. We assessed long-term repeated-dose toxicity of bosentan and valproic acid (VPA) in HepaRG cells under serum-free conditions. Upon 28-day exposure, the EC50 values for bosentan and VPA decreased by 21- and 33-fold, respectively. Using EC(10) as lowest threshold of toxicity in vitro, we estimated the oral equivalent doses for both test compounds using a simplified pharmacokinetic model for the extrapolation of in vitro toxicity to in vivo effect. The model predicts that bosentan is safe at the considered dose under the assumed conditions upon 4 weeks exposure. For VPA, hepatotoxicity is predicted for 4% and 47% of the virtual population at the maximum recommended daily dose after 3 and 4 weeks of exposure, respectively. We also investigated the changes in the central carbon metabolism of HepaRG cells exposed to orally bioavailable concentrations of both drugs. These concentrations are below the 28-day EC(10) and induce significant changes especially in glucose metabolism and urea production. These metabolic changes may have a pronounced impact in susceptible patients such as those with compromised liver function and urea cycle deficiency leading to idiosyncratic toxicity. We show that the combination of modeling based on in vitro repeated-dose data and metabolic changes allows the prediction of human relevant in vivo toxicity with mechanistic insights.
Collapse
Affiliation(s)
- Sebastian Klein
- *Biochemical Engineering, Saarland University, 66123 Saarbruecken, Germany
| | - Silvia Maggioni
- IRCCS - Instituto di Ricerche Farmacologiche "Mario Negri," 20156 Milan, Italy
| | - Joachim Bucher
- Insilico Biotechnology AG, 70563 Stuttgart, Germany, and
| | - Daniel Mueller
- *Biochemical Engineering, Saarland University, 66123 Saarbruecken, Germany
| | - Jens Niklas
- Insilico Biotechnology AG, 70563 Stuttgart, Germany, and
| | | | - Klaus Mauch
- Insilico Biotechnology AG, 70563 Stuttgart, Germany, and
| | - Elmar Heinzle
- *Biochemical Engineering, Saarland University, 66123 Saarbruecken, Germany
| | - Fozia Noor
- *Biochemical Engineering, Saarland University, 66123 Saarbruecken, Germany,
| |
Collapse
|
32
|
Merrick BA, Paules RS, Tice RR. Intersection of toxicogenomics and high throughput screening in the Tox21 program: an NIEHS perspective. ACTA ACUST UNITED AC 2015; 14:7-27. [PMID: 27122658 DOI: 10.1504/ijbt.2015.074797] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Humans are exposed to thousands of chemicals with inadequate toxicological data. Advances in computational toxicology, robotic high throughput screening (HTS), and genome-wide expression have been integrated into the Tox21 program to better predict the toxicological effects of chemicals. Tox21 is a collaboration among US government agencies initiated in 2008 that aims to shift chemical hazard assessment from traditional animal toxicology to target-specific, mechanism-based, biological observations using in vitro assays and lower organism models. HTS uses biocomputational methods for probing thousands of chemicals in in vitro assays for gene-pathway response patterns predictive of adverse human health outcomes. In 1999, NIEHS began exploring the application of toxicogenomics to toxicology and recent advances in NextGen sequencing should greatly enhance the biological content obtained from HTS platforms. We foresee an intersection of new technologies in toxicogenomics and HTS as an innovative development in Tox21. Tox21 goals, priorities, progress, and challenges will be reviewed.
Collapse
Affiliation(s)
- B Alex Merrick
- Biomolecular Screening Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Richard S Paules
- Biomolecular Screening Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Raymond R Tice
- Biomolecular Screening Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| |
Collapse
|
33
|
Alépée N, Bahinski A, Daneshian M, De Wever B, Fritsche E, Goldberg A, Hansmann J, Hartung T, Haycock J, Hogberg H, Hoelting L, Kelm JM, Kadereit S, McVey E, Landsiedel R, Leist M, Lübberstedt M, Noor F, Pellevoisin C, Petersohn D, Pfannenbecker U, Reisinger K, Ramirez T, Rothen-Rutishauser B, Schäfer-Korting M, Zeilinger K, Zurich MG. State-of-the-art of 3D cultures (organs-on-a-chip) in safety testing and pathophysiology. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2014. [PMID: 25027500 DOI: 10.14573/altex1406111] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Integrated approaches using different in vitro methods in combination with bioinformatics can (i) increase the success rate and speed of drug development; (ii) improve the accuracy of toxicological risk assessment; and (iii) increase our understanding of disease. Three-dimensional (3D) cell culture models are important building blocks of this strategy which has emerged during the last years. The majority of these models are organotypic, i.e., they aim to reproduce major functions of an organ or organ system. This implies in many cases that more than one cell type forms the 3D structure, and often matrix elements play an important role. This review summarizes the state of the art concerning commonalities of the different models. For instance, the theory of mass transport/metabolite exchange in 3D systems and the special analytical requirements for test endpoints in organotypic cultures are discussed in detail. In the next part, 3D model systems for selected organs--liver, lung, skin, brain--are presented and characterized in dedicated chapters. Also, 3D approaches to the modeling of tumors are presented and discussed. All chapters give a historical background, illustrate the large variety of approaches, and highlight up- and downsides as well as specific requirements. Moreover, they refer to the application in disease modeling, drug discovery and safety assessment. Finally, consensus recommendations indicate a roadmap for the successful implementation of 3D models in routine screening. It is expected that the use of such models will accelerate progress by reducing error rates and wrong predictions from compound testing.
Collapse
|
34
|
Alépée N, Bahinski A, Daneshian M, De Wever B, Fritsche E, Goldberg A, Hansmann J, Hartung T, Haycock J, Hogberg HT, Hoelting L, Kelm JM, Kadereit S, McVey E, Landsiedel R, Leist M, Lübberstedt M, Noor F, Pellevoisin C, Petersohn D, Pfannenbecker U, Reisinger K, Ramirez T, Rothen-Rutishauser B, Schäfer-Korting M, Zeilinger K, Zurich MG. State-of-the-art of 3D cultures (organs-on-a-chip) in safety testing and pathophysiology. ALTEX 2014; 31:441-77. [PMID: 25027500 PMCID: PMC4783151 DOI: 10.14573/altex.1406111] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 06/30/2014] [Indexed: 02/02/2023]
Abstract
Integrated approaches using different in vitro methods in combination with bioinformatics can (i) increase the success rate and speed of drug development; (ii) improve the accuracy of toxicological risk assessment; and (iii) increase our understanding of disease. Three-dimensional (3D) cell culture models are important building blocks of this strategy which has emerged during the last years. The majority of these models are organotypic, i.e., they aim to reproduce major functions of an organ or organ system. This implies in many cases that more than one cell type forms the 3D structure, and often matrix elements play an important role. This review summarizes the state of the art concerning commonalities of the different models. For instance, the theory of mass transport/metabolite exchange in 3D systems and the special analytical requirements for test endpoints in organotypic cultures are discussed in detail. In the next part, 3D model systems for selected organs--liver, lung, skin, brain--are presented and characterized in dedicated chapters. Also, 3D approaches to the modeling of tumors are presented and discussed. All chapters give a historical background, illustrate the large variety of approaches, and highlight up- and downsides as well as specific requirements. Moreover, they refer to the application in disease modeling, drug discovery and safety assessment. Finally, consensus recommendations indicate a roadmap for the successful implementation of 3D models in routine screening. It is expected that the use of such models will accelerate progress by reducing error rates and wrong predictions from compound testing.
Collapse
Affiliation(s)
| | - Anthony Bahinski
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, USA
| | - Mardas Daneshian
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany
| | | | - Ellen Fritsche
- Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Alan Goldberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - Jan Hansmann
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Thomas Hartung
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany,Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - John Haycock
- Department of Materials Science of Engineering, University of Sheffield, Sheffield, UK
| | - Helena T. Hogberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - Lisa Hoelting
- Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | | | - Suzanne Kadereit
- Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Emily McVey
- Board for the Authorization of Plant Protection Products and Biocides, Wageningen, The Netherlands
| | | | - Marcel Leist
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany,Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Marc Lübberstedt
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Fozia Noor
- Biochemical Engineering, Saarland University, Saarbruecken, Germany
| | | | | | | | | | - Tzutzuy Ramirez
- BASF SE, Experimental Toxicology and Ecology, Ludwigshafen, Germany
| | | | - Monika Schäfer-Korting
- Institute for Pharmacy (Pharmacology and Toxicology), Freie Universität Berlin, Berlin, Germany
| | - Katrin Zeilinger
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Marie-Gabriele Zurich
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland,Swiss Center for Applied Human Toxicology (SCAHT), University of Lausanne, Lausanne, Switzerland
| |
Collapse
|