51
|
Rose S, Cuvellier M, Ezan F, Carteret J, Bruyère A, Legagneux V, Nesslany F, Baffet G, Langouët S. DMSO-free highly differentiated HepaRG spheroids for chronic toxicity, liver functions and genotoxicity studies. Arch Toxicol 2021; 96:243-258. [PMID: 34762139 DOI: 10.1007/s00204-021-03178-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/06/2021] [Indexed: 12/23/2022]
Abstract
The liver is essential in the elimination of environmental and food contaminants. Given the interspecies differences between rodents and humans, the development of relevant in vitro human models is crucial to investigate liver functions and toxicity in cells that better reflect pathophysiological processes. Classically, the differentiation of the hepatic HepaRG cell line requires high concentration of dimethyl sulfoxide (DMSO), which restricts its usefulness for drug-metabolism studies. Herein, we describe undifferentiated HepaRG cells embedded in a collagen matrix in DMSO-free conditions that rapidly organize into polarized hollow spheroids of differentiated hepatocyte-like cells (Hepoid-HepaRG). Our conditions allow concomitant proliferation with high levels of liver-specific functions and xenobiotic metabolism enzymes expression and activities after a few days of culture and for at least 4 weeks. By studying the toxicity of well-known injury-inducing drugs by treating cells with 1- to 100-fold of their plasmatic concentrations, we showed appropriate responses and demonstrate the sensitivity to drugs known to induce various degrees of liver injury. Our results also demonstrated that the model is well suited to estimate cholestasis and steatosis effects of drugs following chronic treatment. Additionally, DNA alterations caused by four genotoxic compounds (Aflatoxin B1 (AFB1), Benzo[a]Pyrene (B[a]P), Cyclophosphamide (CPA) and Methyl methanesulfonate (MMS)) were quantified in a dose-dependent manner by the comet and micronucleus assays. Their genotoxic effects were significantly increased after either an acute 24 h treatment (AFB1: 1.5-6 μM, CPA: 2.5-10 μM, B[a]P: 12.5-50 μM, MMS: 90-450 μM) or after a 14-day treatment at much lower concentrations (AFB1: 0.05-0.2 μM, CPA: 0.125-0.5 μM, B[a]P: 0.125-0.5 μM) representative to human exposure. Altogether, the DMSO-free 3D culture of Hepoid-HepaRG provides highly differentiated and proliferating cells relevant for various toxicological in vitro assays, especially for drug-preclinical studies and environmental chemicals risk assessment.
Collapse
Affiliation(s)
- Sophie Rose
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Marie Cuvellier
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Frédéric Ezan
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Jennifer Carteret
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Arnaud Bruyère
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Vincent Legagneux
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Fabrice Nesslany
- Genotoxicology Department, Institut Pasteur de Lille, 1, Rue du Professeur Calmette, 59000, Lille, France
| | - Georges Baffet
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France.
| | - Sophie Langouët
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France.
| |
Collapse
|
52
|
The utility of endogenous glycochenodeoxycholate-3-sulfate and 4β-hydroxycholesterol to evaluate the hepatic disposition of atorvastatin in rats. Asian J Pharm Sci 2021; 16:519-529. [PMID: 34703500 PMCID: PMC8520055 DOI: 10.1016/j.ajps.2021.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 01/06/2021] [Accepted: 03/07/2021] [Indexed: 11/22/2022] Open
Abstract
The liver is an important organ for drugs disposition, and thus how to accurately evaluate hepatic clearance is essential for proper drug dosing. However, there are many limitations in drug dosage adjustment based on liver function and pharmacogenomic testing. In this study, we evaluated the ability of endogenous glycochenodeoxycholate-3-sulfate (GCDCA-S) and 4β-hydroxycholesterol (4β-HC) plasma levels to evaluate organic anion-transporting polypeptide (Oatps)-mediated hepatic uptake and Cyp3a-meidated metabolism of atorvastatin (ATV) in rats. The concentration of ATV and its metabolites, 2-OH ATV and 4-OH ATV, was markedly increased after a single injection of rifampicin (RIF), an inhibitor of Oatps. Concurrently, plasma GCDCA-S levels were also elevated. After a single injection of the Cyp3a inhibitor ketoconazole (KTZ), plasma ATV concentrations were significantly increased and 2-OH ATV concentrations were decreased, consistent with the metabolism of ATV by Cyp3a. However, plasma 4β-HC was not affected by KTZ treatment despite it being a Cyp3a metabolite of cholesterol. After repeated oral administration of RIF, plasma concentrations of ATV, 2-OH ATV and 4-OH ATV were markedly increased and the hepatic uptake ratio of ATV and GCDCA-S was decreased. KTZ did not affect plasma concentrations of ATV, 2-OH ATV and 4-OH ATV, but significantly decreased the metabolic ratio of total and 4-OH ATV. However, the plasma level and hepatic metabolism of 4β-HC were not changed by KTZ. The inhibition of hepatic uptake of GCDCA-S by RIF was fully reversed after a 7-d washout of RIF. Plasma concentration and hepatic uptake ratio of GCDCA-S were correlated with the plasma level and hepatic uptake of ATV in rats with ANIT-induced liver injury, respectively. These results demonstrate that plasma GCDCA-S is a sensitive probe for the assessment of Oatps-mediated hepatic uptake of ATV. However, Cyp3a-mediated metabolism of ATV was not predicted by plasma 4β-HC levels in rats.
Collapse
|
53
|
Abstract
There are many factors which are known to cause variability in human in vitro enzyme kinetic data. Factors such as the source of enzyme and how it was prepared, the genetics and background of the donor, how the in vitro studies are designed, and how the data are analyzed contribute to variability in the resulting kinetic parameters. It is important to consider not only the factors which cause variability within an experiment, such as selection of a probe substrate, but also those that cause variability when comparing kinetic data across studies and laboratories. For example, the artificial nature of the microsomal lipid membrane and microenvironment in some recombinantly expressed enzymes, relative to those found in native tissue microsomes, has been shown to influence enzyme activity and thus can be a source of variability when comparing across the two different systems. All of these factors, and several others, are discussed in detail in the chapter below. In addition, approaches which can be used to visualize the uncertainty arising from the use of enzyme kinetic data within the context of predicting human pharmacokinetics are discussed.
Collapse
|
54
|
Polidoro MA, Ferrari E, Marzorati S, Lleo A, Rasponi M. Experimental liver models: From cell culture techniques to microfluidic organs-on-chip. Liver Int 2021; 41:1744-1761. [PMID: 33966344 DOI: 10.1111/liv.14942] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022]
Abstract
The liver is one of the most studied organs of the human body owing to its central role in xenobiotic and drug metabolism. In recent decades, extensive research has aimed at developing in vitro liver models able to mimic liver functions to study pathophysiological clues in high-throughput and reproducible environments. Two-dimensional (2D) models have been widely used in screening potential toxic compounds but have failed to accurately reproduce the three-dimensionality (3D) of the liver milieu. To overcome these limitations, improved 3D culture techniques have been developed to recapitulate the hepatic native microenvironment. These models focus on reproducing the liver architecture, representing both parenchymal and nonparenchymal cells, as well as cell interactions. More recently, Liver-on-Chip (LoC) models have been developed with the aim of providing physiological fluid flow and thus achieving essential hepatic functions. Given their unprecedented ability to recapitulate critical features of the liver cellular environments, LoC have been extensively adopted in pathophysiological modelling and currently represent a promising tool for tissue engineering and drug screening applications. In this review, we discuss the evolution of experimental liver models, from the ancient 2D hepatocyte models, widely used for liver toxicity screening, to 3D and LoC culture strategies adopted for mirroring a more physiological microenvironment for the study of liver diseases.
Collapse
Affiliation(s)
- Michela Anna Polidoro
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Erika Ferrari
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Simona Marzorati
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| |
Collapse
|
55
|
Preiss LC, Liu R, Hewitt P, Thompson D, Georgi K, Badolo L, Lauschke VM, Petersson C. Deconvolution of Cytochrome P450 Induction Mechanisms in HepaRG Nuclear Hormone Receptor Knockout Cells. Drug Metab Dispos 2021; 49:668-678. [PMID: 34035124 DOI: 10.1124/dmd.120.000333] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/10/2021] [Indexed: 12/11/2022] Open
Abstract
Pregnane X receptor (PXR), constitutive androstane receptor (CAR), and PXR/CAR knockout (KO) HepaRG cells, as well as a PXR reporter gene assay, were used to investigate the mechanism of CYP3A4 and CYP2B6 induction by prototypical substrates and a group of compounds from the Merck KGaA oncology drug discovery pipeline. The basal and inducible gene expression of CYP3A4 and CYP2B6 of nuclear hormone receptor (NHR) KO HepaRG relative to control HepaRG was characterized. The basal expression of CYP3A4 was markedly higher in the PXR (10-fold) and CAR (11-fold) KO cell lines compared with control HepaRG, whereas inducibility was substantially lower. Inversely, basal expression of CYP3A4 in PXR/CAR double KO (dKO) was low (10-fold reduction). Basal CYP2B6 expression was high in PXR KO (9-fold) cells which showed low inducibility, whereas the basal expression remained unchanged in CAR and dKO cell lines compared with control cells. Most of the test compounds induced CYP3A4 and CYP2B6 via PXR and, to a lesser extent, via CAR. Furthermore, other non-NHR-driven induction mechanisms were implicated, either alone or in addition to NHRs. Notably, 5 of the 16 compounds (31%) that were PXR inducers in HepaRG did not activate PXR in the reporter gene assay, illustrating the limitations of this system. This study indicates that HepaRG is a highly sensitive system fit for early screening of cytochrome P450 (P450) induction in drug discovery. Furthermore, it shows the applicability of HepaRG NHR KO cells as tools to deconvolute mechanisms of P450 induction using novel compounds representative for oncology drug discovery. SIGNIFICANCE STATEMENT: This work describes the identification of induction mechanisms of CYP3A4 and CYP2B6 for an assembly of oncology drug candidates using HepaRG nuclear hormone receptor knockout and displays its advantages compared to a pregnane X receptor reporter gene assay. With this study, risk assessment of drug candidates in early drug development can be improved.
Collapse
Affiliation(s)
- Lena C Preiss
- Departments of Drug Metabolism and Pharmacokinetics (L.C.P., R.L., K.G., L.B., C.P.) and Early Chemical and Preclinical Safety (P.H.), Merck KGaA, Darmstadt, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); and Research & Development, In Vitro Safety Systems, MilliporeSigma, St. Louis, Missouri (D.T.)
| | - Ruoqi Liu
- Departments of Drug Metabolism and Pharmacokinetics (L.C.P., R.L., K.G., L.B., C.P.) and Early Chemical and Preclinical Safety (P.H.), Merck KGaA, Darmstadt, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); and Research & Development, In Vitro Safety Systems, MilliporeSigma, St. Louis, Missouri (D.T.)
| | - Philip Hewitt
- Departments of Drug Metabolism and Pharmacokinetics (L.C.P., R.L., K.G., L.B., C.P.) and Early Chemical and Preclinical Safety (P.H.), Merck KGaA, Darmstadt, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); and Research & Development, In Vitro Safety Systems, MilliporeSigma, St. Louis, Missouri (D.T.)
| | - David Thompson
- Departments of Drug Metabolism and Pharmacokinetics (L.C.P., R.L., K.G., L.B., C.P.) and Early Chemical and Preclinical Safety (P.H.), Merck KGaA, Darmstadt, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); and Research & Development, In Vitro Safety Systems, MilliporeSigma, St. Louis, Missouri (D.T.)
| | - Katrin Georgi
- Departments of Drug Metabolism and Pharmacokinetics (L.C.P., R.L., K.G., L.B., C.P.) and Early Chemical and Preclinical Safety (P.H.), Merck KGaA, Darmstadt, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); and Research & Development, In Vitro Safety Systems, MilliporeSigma, St. Louis, Missouri (D.T.)
| | - Lassina Badolo
- Departments of Drug Metabolism and Pharmacokinetics (L.C.P., R.L., K.G., L.B., C.P.) and Early Chemical and Preclinical Safety (P.H.), Merck KGaA, Darmstadt, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); and Research & Development, In Vitro Safety Systems, MilliporeSigma, St. Louis, Missouri (D.T.)
| | - Volker M Lauschke
- Departments of Drug Metabolism and Pharmacokinetics (L.C.P., R.L., K.G., L.B., C.P.) and Early Chemical and Preclinical Safety (P.H.), Merck KGaA, Darmstadt, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); and Research & Development, In Vitro Safety Systems, MilliporeSigma, St. Louis, Missouri (D.T.)
| | - Carl Petersson
- Departments of Drug Metabolism and Pharmacokinetics (L.C.P., R.L., K.G., L.B., C.P.) and Early Chemical and Preclinical Safety (P.H.), Merck KGaA, Darmstadt, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); and Research & Development, In Vitro Safety Systems, MilliporeSigma, St. Louis, Missouri (D.T.)
| |
Collapse
|
56
|
Choudhuri S, Kaur T, Jain S, Sharma C, Asthana S. A review on genotoxicity in connection to infertility and cancer. Chem Biol Interact 2021; 345:109531. [PMID: 34058178 DOI: 10.1016/j.cbi.2021.109531] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/22/2021] [Accepted: 05/17/2021] [Indexed: 10/21/2022]
Abstract
Genotoxicity has been identified as the main cause of infertility and a variety of cancers. The mechanisms affect the structure, quality of the information or the segregation of DNA and are not inherently correlated with mutagenicity. The concept of genotoxicity, the chemical classes that cause genetic damage and the associated mechanisms of action are discussed here. Hazardous effects of pharmaceuticals, cosmetics, agrochemicals, industrial compounds, food additives, natural toxins and nanomaterials are, in large part, identified by genotoxicity and mutagenicity tests. These are critical and early steps in industrial and regulatory health assessment. Though several in vitro experiments are commonly used and approval by regulatory agencies for commercial licensing of drugs, their accuracy in human predictions for genotoxic and mutagenic effects is frequently questioned. Treatment of real and functional genetic toxicity problems depends in detail on the knowledge of mechanisms of DNA damage in the molecular, subcellular, cellular and tissue or organ system levels. Current strategies for risk assessment of human health need revisions to achieve robust and reliable results for optimizing their effectiveness. Additionally, computerized methods, neo-biomarkers leveraging '-omics' approaches, all of which can provide a convincing genotoxicity evaluation to reduce infertility and cancer risk.
Collapse
Affiliation(s)
- Sharmistha Choudhuri
- Department of Biochemistry, R. G. Kar Medical College and Hospital, Kolkata, West Bengal, India
| | - Taruneet Kaur
- Animal Biochemistry Division, National Dairy Research Institute, Karnal, Haryana, India
| | - Sapna Jain
- Multidisciplinary Clinical Translational Research, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Chandresh Sharma
- Multidisciplinary Clinical Translational Research, Translational Health Science and Technology Institute, Faridabad, Haryana, India.
| | - Shailendra Asthana
- Non-Communicable Disease, Translational Health Science and Technology Institute, Faridabad, Haryana, India.
| |
Collapse
|
57
|
The Food Contaminants Pyrrolizidine Alkaloids Disturb Bile Acid Homeostasis Structure-Dependently in the Human Hepatoma Cell Line HepaRG. Foods 2021; 10:foods10051114. [PMID: 34069968 PMCID: PMC8157858 DOI: 10.3390/foods10051114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 11/18/2022] Open
Abstract
Pyrrolizidine alkaloids (PAs) are a group of secondary plant metabolites being contained in various plant species. The consumption of contaminated food can lead to acute intoxications in humans and exert severe hepatotoxicity. The development of jaundice and elevated bile acid concentrations in blood have been reported in acute human PA intoxication, indicating a connection between PA exposure and the induction of cholestasis. Additionally, it is considered that differences in toxicity of individual PAs is based on their individual chemical structures. Therefore, we aimed to elucidate the structure-dependent disturbance of bile acid homeostasis by PAs in the human hepatoma cell line HepaRG. A set of 14 different PAs, including representatives of all major structural characteristics, namely, the four different necine bases retronecine, heliotridine, otonecine and platynecine and different grades of esterification, was analyzed in regard to the expression of genes involved in bile acid synthesis, metabolism and transport. Additionally, intra- and extracellular bile acid levels were analyzed after PA treatment. In summary, our data show significant structure-dependent effects of PAs on bile acid homeostasis. Especially PAs of diester type caused the strongest dysregulation of expression of genes associated with cholestasis and led to a strong decrease of intra- and extracellular bile acid concentrations.
Collapse
|
58
|
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: 2.0] [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
|
59
|
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: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
60
|
Franzosa JA, Bonzo JA, Jack J, Baker NC, Kothiya P, Witek RP, Hurban P, Siferd S, Hester S, Shah I, Ferguson SS, Houck KA, Wambaugh JF. High-throughput toxicogenomic screening of chemicals in the environment using metabolically competent hepatic cell cultures. NPJ Syst Biol Appl 2021; 7:7. [PMID: 33504769 PMCID: PMC7840683 DOI: 10.1038/s41540-020-00166-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 10/15/2020] [Indexed: 01/30/2023] Open
Abstract
The ToxCast in vitro screening program has provided concentration-response bioactivity data across more than a thousand assay endpoints for thousands of chemicals found in our environment and commerce. However, most ToxCast screening assays have evaluated individual biological targets in cancer cell lines lacking integrated physiological functionality (such as receptor signaling, metabolism). We evaluated differentiated HepaRGTM cells, a human liver-derived cell model understood to effectively model physiologically relevant hepatic signaling. Expression of 93 gene transcripts was measured by quantitative polymerase chain reaction using Fluidigm 96.96 dynamic arrays in response to 1060 chemicals tested in eight-point concentration-response. A Bayesian framework quantitatively modeled chemical-induced changes in gene expression via six transcription factors including: aryl hydrocarbon receptor, constitutive androstane receptor, pregnane X receptor, farnesoid X receptor, androgen receptor, and peroxisome proliferator-activated receptor alpha. For these chemicals the network model translates transcriptomic data into Bayesian inferences about molecular targets known to activate toxicological adverse outcome pathways. These data also provide new insights into the molecular signaling network of HepaRGTM cell cultures.
Collapse
Affiliation(s)
- Jill A Franzosa
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. EPA, Research Triangle Park, NC, 27711, USA
| | - Jessica A Bonzo
- Cell Biology, Biosciences Division, Thermo Fisher Scientific, Frederick, MD, 21703, USA
| | - John Jack
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. EPA, Research Triangle Park, NC, 27711, USA
| | | | - Parth Kothiya
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. EPA, Research Triangle Park, NC, 27711, USA
| | - Rafal P Witek
- Cell Biology, Biosciences Division, Thermo Fisher Scientific, Frederick, MD, 21703, USA
| | | | | | - Susan Hester
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. EPA, Research Triangle Park, NC, 27711, USA
| | - Imran Shah
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. EPA, Research Triangle Park, NC, 27711, USA
| | - Stephen S Ferguson
- Division of National Toxicology Program, National Institutes of Environmental Health Sciences of National Institutes of Health, Durham, NC, 27709, USA
| | - Keith A Houck
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. EPA, Research Triangle Park, NC, 27711, USA
| | - John F Wambaugh
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. EPA, Research Triangle Park, NC, 27711, USA.
| |
Collapse
|
61
|
Jones SW, Penman SL, French NS, Park BK, Chadwick AE. Investigating dihydroorotate dehydrogenase inhibitor mediated mitochondrial dysfunction in hepatic in vitro models. Toxicol In Vitro 2021; 72:105096. [PMID: 33460737 DOI: 10.1016/j.tiv.2021.105096] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/17/2020] [Accepted: 01/12/2021] [Indexed: 01/13/2023]
Abstract
Inhibition of dihydroorotate dehydrogenase (DHODH), the rate-limiting enzymatic step in de novo pyrimidine synthesis, has broad immunosuppressive effects in vivo and shows promise as a therapeutic target for the treatment of malignancies, viral infections and auto-immune diseases. Whilst there are numerous DHODH inhibitors under development, leflunomide and teriflunomide are the only FDA approved compounds on the market, each of which have been issued with black-box warnings for hepatotoxicity. Mitochondrial dysfunction is a putative mechanism by which teriflunomide and leflunomide elicit their hepatotoxic effects, however it is as yet unclear whether this is shared by other nascent DHODH inhibitors. The present study aimed to evaluate the propensity for DHODH inhibitors to mediate mitochondrial dysfunction in two hepatic in vitro models. Initial comparisons of cytotoxicity and ATP content in HepaRG® cells primed for oxidative metabolism, in tandem with mechanistic evaluations by extracellular flux analysis identified multifactorial toxicity and moderate indications of respiratory chain dysfunction or uncoupling. Further investigations using HepG2 cells, a hepatic line with limited capability for phase I xenobiotic metabolism, identified leflunomide and brequinar as positive mitochondrial toxicants. Taken together, biotransformation of some DHODH inhibitor species may play a role in mediating or masking hepatic mitochondrial liabilities.
Collapse
Affiliation(s)
- Samantha W Jones
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Ashton street Liverpool, L69 3GE, UK
| | - Sophie L Penman
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Ashton street Liverpool, L69 3GE, UK
| | - Neil S French
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Ashton street Liverpool, L69 3GE, UK
| | - B Kevin Park
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Ashton street Liverpool, L69 3GE, UK
| | - Amy E Chadwick
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Ashton street Liverpool, L69 3GE, UK.
| |
Collapse
|
62
|
Heintze T, Klein K, Hofmann U, Zanger UM. Differential effects on human cytochromes P450 by CRISPR/Cas9-induced genetic knockout of cytochrome P450 reductase and cytochrome b5 in HepaRG cells. Sci Rep 2021; 11:1000. [PMID: 33441761 PMCID: PMC7806635 DOI: 10.1038/s41598-020-79952-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
HepaRG cells are increasingly accepted as model for human drug metabolism and other hepatic functions. We used lentiviral transduction of undifferentiated HepaRG cells to deliver Cas9 and two alternative sgRNAs targeted at NADPH:cytochrome P450 oxidoreductase (POR), the obligate electron donor for microsomal cytochromes P450 (CYP). Cas9-expressing HepaRGVC (vector control) cells were phenotypically similar to wild type HepaRG cells and could be differentiated into hepatocyte-like cells by DMSO. Genetic POR-knockout resulted in phenotypic POR knockdown of up to 90% at mRNA, protein, and activity levels. LC–MS/MS measurement of seven CYP-activities showed differential effects of POR-knockdown with CYP2C8 being least and CYP2C9 being most affected. Further studies on cytochrome b5 (CYB5), an alternative NADH-dependent electron donor indicated particularly strong support of CYP2C8-dependent amodiaquine N-deethylation by CYB5 and this was confirmed by genetic CYB5 single- and POR/CYB5 double-knockout. POR-knockdown also affected CYP expression on mRNA and protein level, with CYP1A2 being induced severalfold, while CYP2C9 was strongly downregulated. In summary our results show that POR/NADPH- and CYB5/NADH-electron transport systems influence human drug metabolizing CYPs differentially and differently than mouse Cyps. Our Cas9-expressing HepaRGVC cells should be suitable to study the influence of diverse genes on drug metabolism and other hepatic functions.
Collapse
Affiliation(s)
- Tamara Heintze
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Kathrin Klein
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Ulrich M Zanger
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany. .,Eberhard Karls University Tuebingen, Tuebingen, Germany.
| |
Collapse
|
63
|
Hayes CN, Chayama K. Unmet Needs in Basic Research of Hepatitis B Virus Infection: In Vitro and In Vivo Models. HEPATITIS B VIRUS AND LIVER DISEASE 2021:29-49. [DOI: 10.1007/978-981-16-3615-8_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
64
|
Chen S, Wu Q, Li X, Li D, Mei N, Ning B, Puig M, Ren Z, Tolleson WH, Guo L. Characterization of cytochrome P450s (CYP)-overexpressing HepG2 cells for assessing drug and chemical-induced liver toxicity. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2021; 39:68-86. [PMID: 33576714 PMCID: PMC7931144 DOI: 10.1080/26896583.2021.1880242] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Hepatic metabolism catalyzed by the cytochrome P450 (CYP) superfamily affects liver toxicity associated with exposures to natural compounds and xenobiotic agents. Previously we generated a battery of HepG2-derived stable cell lines that individually express 14 CYPs (1A1, 1A2, 1B1, 2A6, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6, 2E1, 3A4, 3A5, and 3A7). In this study, we comprehensively characterized each cell line for its CYP expression and enzyme activity. Specifically, we measured the mRNA expression, protein expression, and metabolite formation. Using CYP3A4, 2D6, and 2C9-overexpressing cells as representatives, we examined the stability of these cells in long-term cultures for up to 10 passages. The results showed that CYPs can be stably overexpressed for up to 10 cell culture passages without losing their activities. The robustness of responses to stimuli among the cells at different passages was also investigated in CYP3A4-overexpressing cells and the response to amiodarone and dronedarone showed no difference between the cells at the passage 2 and 10. Moreover, the mRNA expression level of most CYPs was higher in CYP-overexpressing HepG2 cells than that in HepaRG cells and primary human hepatocytes. This study confirmed the stability of CYP-overexpressing HepG2 cell lines and provided useful information for a broader use of these cells in pharmacologic and toxicologic research.
Collapse
Affiliation(s)
- Si Chen
- Division of Biochemical Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| | - Qiangen Wu
- Division of Biochemical Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| | - Xilin Li
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| | - Dongying Li
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| | - Nan Mei
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| | - Baitang Ning
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| | - Montserrat Puig
- Division of Biotechnology Review and Research III, Office of Biotechnology Products, Center for Drug Evaluation and Research/U.S. FDA, Silver Spring, Maryland, USA
| | - Zhen Ren
- Division of Biochemical Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| | - William H. Tolleson
- Division of Biochemical Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| | - Lei Guo
- Division of Biochemical Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| |
Collapse
|
65
|
HepG2 (C3A) spheroids show higher sensitivity compared to HepaRG spheroids for drug-induced liver injury (DILI). Toxicol Appl Pharmacol 2020; 408:115279. [DOI: 10.1016/j.taap.2020.115279] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/25/2020] [Accepted: 10/11/2020] [Indexed: 12/20/2022]
|
66
|
Milner E, Ainsworth M, McDonough M, Stevens B, Buehrer J, Delzell R, Wilson C, Barnhill J. Emerging Three-Dimensional Hepatic Models in Relation to Traditional Two-Dimensional In Vitro Assays for Evaluating Drug Metabolism and Hepatoxicity. MEDICINE IN DRUG DISCOVERY 2020. [DOI: 10.1016/j.medidd.2020.100060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
67
|
Cox CR, Lynch S, Goldring C, Sharma P. Current Perspective: 3D Spheroid Models Utilizing Human-Based Cells for Investigating Metabolism-Dependent Drug-Induced Liver Injury. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:611913. [PMID: 35047893 PMCID: PMC8757888 DOI: 10.3389/fmedt.2020.611913] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/04/2020] [Indexed: 12/21/2022] Open
Abstract
Drug-induced liver injury (DILI) remains a leading cause for the withdrawal of approved drugs. This has significant financial implications for pharmaceutical companies, places increasing strain on global health services, and causes harm to patients. For these reasons, it is essential that in-vitro liver models are capable of detecting DILI-positive compounds and their underlying mechanisms, prior to their approval and administration to patients or volunteers in clinical trials. Metabolism-dependent DILI is an important mechanism of drug-induced toxicity, which often involves the CYP450 family of enzymes, and is associated with the production of a chemically reactive metabolite and/or inefficient removal and accumulation of potentially toxic compounds. Unfortunately, many of the traditional in-vitro liver models fall short of their in-vivo counterparts, failing to recapitulate the mature hepatocyte phenotype, becoming metabolically incompetent, and lacking the longevity to investigate and detect metabolism-dependent DILI and those associated with chronic and repeat dosing regimens. Nevertheless, evidence is gathering to indicate that growing cells in 3D formats can increase the complexity of these models, promoting a more mature-hepatocyte phenotype and increasing their longevity, in vitro. This review will discuss the use of 3D in vitro models, namely spheroids, organoids, and perfusion-based systems to establish suitable liver models to investigate metabolism-dependent DILI.
Collapse
Affiliation(s)
- Christopher R. Cox
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- *Correspondence: Christopher R. Cox
| | - Stephen Lynch
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Christopher Goldring
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Parveen Sharma
- Department of Pharmacology and Experimental Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- Liverpool Centre for Cardiovascular Science, Liverpool, United Kingdom
| |
Collapse
|
68
|
Štampar M, Breznik B, Filipič M, Žegura B. Characterization of In Vitro 3D Cell Model Developed from Human Hepatocellular Carcinoma (HepG2) Cell Line. Cells 2020; 9:E2557. [PMID: 33260628 PMCID: PMC7759933 DOI: 10.3390/cells9122557] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022] Open
Abstract
In genetic toxicology, there is a trend against the increased use of in vivo models as highlighted by the 3R strategy, thus encouraging the development and implementation of alternative models. Two-dimensional (2D) hepatic cell models, which are generally used for studying the adverse effects of chemicals and consumer products, are prone to giving misleading results. On the other hand, newly developed hepatic three-dimensional (3D) cell models provide an attractive alternative, which, due to improved cell interactions and a higher level of liver-specific functions, including metabolic enzymes, reflect in vivo conditions more accurately. We developed an in vitro 3D cell model from the human hepatocellular carcinoma (HepG2) cell line. The spheroids were cultured under static conditions and characterised by monitoring their growth, morphology, and cell viability during the time of cultivation. A time-dependent suppression of cell division was observed. Cell cycle analysis showed time-dependent accumulation of cells in the G0/G1 phase. Moreover, time-dependent downregulation of proliferation markers was shown at the mRNA level. Genes encoding hepatic markers, metabolic phase I/II enzymes, were time-dependently deregulated compared to monolayers. New knowledge on the characteristics of the 3D cell model is of great importance for its further development and application in the safety assessment of chemicals, food products, and complex mixtures.
Collapse
Affiliation(s)
- Martina Štampar
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia; (M.Š.); (B.B.); (M.F.)
- Jozef Stefan International Postgraduate School, 1000 Ljubljana, Slovenia
| | - Barbara Breznik
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia; (M.Š.); (B.B.); (M.F.)
| | - Metka Filipič
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia; (M.Š.); (B.B.); (M.F.)
- Jozef Stefan International Postgraduate School, 1000 Ljubljana, Slovenia
| | - Bojana Žegura
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia; (M.Š.); (B.B.); (M.F.)
- Jozef Stefan International Postgraduate School, 1000 Ljubljana, Slovenia
| |
Collapse
|
69
|
Hartman GD, Kuduk SD, Espiritu C, Lam AM. P450s under Restriction (PURE) Screen Using HepaRG and Primary Human Hepatocytes for Discovery of Novel HBV Antivirals. ACS Med Chem Lett 2020; 11:1919-1927. [PMID: 33062174 DOI: 10.1021/acsmedchemlett.9b00630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 02/26/2020] [Indexed: 12/13/2022] Open
Abstract
Herein is reported a novel screening paradigm PURE (P450s under restriction) for the identification and optimization of hits as part of a hepatitis B virus (HBV) antiviral discovery program. To closely represent in vivo hepatocytes, differentiated HepaRG cells (dHRGs) and primary human hepatocytes (PHHs) were used as the basis for an HBV infection system. However, a significant challenge arose during potency evaluation in using cultured dHRGs and PHHs as screening platforms because, as with hepatocytes in vivo, these cells express active cytochrome P450 enzymes and thus can metabolize test compounds. The observed antiviral effects may be the cumulative result of a dynamic pool of parent compound and metabolites thus confounding structure activity relationship (SAR) interpretation and subsequent optimization design initiatives. We show here that PURE methodology restricts metabolism of HBV-infected dHRGs and PHHs and thus provides highly informative potency data for decision-making on key representative antiviral compounds.
Collapse
Affiliation(s)
- George D. Hartman
- Novira Therapeutics, a Janssen Pharmaceutical Company, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Scott D. Kuduk
- Novira Therapeutics, a Janssen Pharmaceutical Company, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Christine Espiritu
- Novira Therapeutics, a Janssen Pharmaceutical Company, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Angela M. Lam
- Novira Therapeutics, a Janssen Pharmaceutical Company, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| |
Collapse
|
70
|
Noh KH, Kang HM, Oh SJ, Lee JY, Kim DH, Kim M, Chung KS, Son MY, Kim DS, Cho HS, Lee J, Lee DG, Lim JH, Jung CR. A new experimental model to study human drug responses. Biofabrication 2020; 12:045029. [PMID: 32975216 DOI: 10.1088/1758-5090/abb652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Accurate prediction of pharmacokinetic (PK) and pharmacodynamic (PD) characteristics is critical for drug development. Oral drugs are particularly difficult because they are absorbed by the intestine and metabolized in the liver before systemic metabolism in vivo; this is called the first-pass effect and is a critical factor for predicting oral bioavailability (BA). Here, we fabricated a new networking and circulating cell culture system (NCCS), mimicking the circulatory system and interaction of organs for studying the pharmacokinetic and pharmacodynamics of oral drugs in vitro. NCCS consisted of a micro-pump for circulating fluids, two types of multi-insert culture dishes for culturing different cell types, and an orbital shaker for mixing; flow rate and shaking-speed were controlled by weight-sensors and drivers. A first-pass effect test was performed using functionally differentiated HepaRG and Caco-2 cell lines, using a new modified spheroid forming unit (SFU) protocol. To verify the similarity of PK (first-pass effect) data of NCCS with the data from the human body, 15 reference drugs were chosen and their associated data were obtained by liquid chromatography-mass spectrometry analysis. NCCS generated absorption and metabolism data showed >70% similarity to human data respectively. NCCS can also be used to demonstrate species differences. Animal models are the primary basis for drug discovery, development, and testing. However, the weak correlation between humans and animals, particularly regarding absorption and metabolism, is a substantial limitation for the use of animal models. Here we compare human and mouse acetaminophen (APAP) metabolism using NCCS, and its application can be extended to assess cellular responses, such as efficacy and toxicity, simultaneously.
Collapse
Affiliation(s)
- Kyung Hee Noh
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Acito M, Bartolini D, Ceccarini MR, Russo C, Vannini S, Dominici L, Codini M, Villarini M, Galli F, Beccari T, Moretti M. Imbalance in the antioxidant defence system and pro-genotoxic status induced by high glucose concentrations: In vitro testing in human liver cells. Toxicol In Vitro 2020; 69:105001. [PMID: 32942007 DOI: 10.1016/j.tiv.2020.105001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 08/18/2020] [Accepted: 09/12/2020] [Indexed: 12/19/2022]
Abstract
It has been hypothesized that high glucose concentrations might contribute to the overall intracellular oxidative stress either by the direct generation of reactive oxygen species (ROS) or by altering the redox balance. Moreover, it has also been suggested that high glucose concentration can increase the susceptibility of DNA to genotoxic effects of xenobiotics. The aim of this approach was to test high glucose concentrations for pro-genotoxicity in human liver cells by setting up an in vitro model for hyperglycaemia. The experimental design included performing of tests on both human HepG2 tumour cells and HepaRG immortalized cells. Increased cell susceptibility to genotoxic xenobiotics was tested by challenging cell cultures with 4-nitroquinoline-N-oxide (4NQO) and evaluating the extent of primary DNA damage by comet assay. Moreover, we evaluated the relationship between glucose concentration and intracellular ROS, as well as the effects of glucose concentration on the induction of Nrf2-dependent genes such as Glutathione S-transferases, Heme‑oxygenase-1, and Glutathione peroxidase-4. To investigate the involvement of ROS in the induced pro-genotoxic activity, parallel experimental sets were set up by considering co-treatment of cells with the model mutagen 4NQO and the antioxidant, glutathione precursor N-acetyl-L-cysteine. High glucose concentrations caused a significant increase in the levels of primary DNA damage, with a pro-genotoxic condition closely related to the concentration of glucose in the culture medium when cells were exposed to 4NQO. High glucose concentrations also stimulated the production of ROS and down-regulated genes involved in contrasting of the effects of oxidative stress. In conclusion, in the presence of high concentrations of glucose, the cells are in unfavourable conditions for the maintenance of genome integrity.
Collapse
Affiliation(s)
- Mattia Acito
- Department of Pharmaceutical Sciences, Unit of Public Health, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Desirée Bartolini
- Department of Pharmaceutical Sciences, Unit of Nutrition and Clinical Biochemistry, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Maria Rachele Ceccarini
- Department of Pharmaceutical Sciences, Unit of Food Chemistry, Biochemistry, Physiology and Nutrition, University of Perugia, Via San Costanzo, 06126 Perugia, Italy
| | - Carla Russo
- Department of Pharmaceutical Sciences, Unit of Public Health, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Samuele Vannini
- Department of Pharmaceutical Sciences, Unit of Public Health, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Luca Dominici
- Department of Pharmaceutical Sciences, Unit of Public Health, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Michela Codini
- Department of Pharmaceutical Sciences, Unit of Food Chemistry, Biochemistry, Physiology and Nutrition, University of Perugia, Via San Costanzo, 06126 Perugia, Italy
| | - Milena Villarini
- Department of Pharmaceutical Sciences, Unit of Public Health, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Francesco Galli
- Department of Pharmaceutical Sciences, Unit of Nutrition and Clinical Biochemistry, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Tommaso Beccari
- Department of Pharmaceutical Sciences, Unit of Food Chemistry, Biochemistry, Physiology and Nutrition, University of Perugia, Via San Costanzo, 06126 Perugia, Italy
| | - Massimo Moretti
- Department of Pharmaceutical Sciences, Unit of Public Health, University of Perugia, Via del Giochetto, 06122 Perugia, Italy.
| |
Collapse
|
72
|
3D In Vitro Human Organ Mimicry Devices for Drug Discovery, Development, and Assessment. ADVANCES IN POLYMER TECHNOLOGY 2020. [DOI: 10.1155/2020/6187048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The past few decades have shown significant advancement as complex in vitro humanized systems have substituted animal trials and 2D in vitro studies. 3D humanized platforms mimic the organs of interest with their stimulations (physical, electrical, chemical, and mechanical). Organ-on-chip devices, including in vitro modelling of 3D organoids, 3D microfabrication, and 3D bioprinted platforms, play an essential role in drug discovery, testing, and assessment. In this article, a thorough review is provided of the latest advancements in the area of organ-on-chip devices targeting liver, kidney, lung, gut, heart, skin, and brain mimicry devices for drug discovery, development, and/or assessment. The current strategies, fabrication methods, and the specific application of each device, as well as the advantages and disadvantages, are presented for each reported platform. This comprehensive review also provides some insights on the challenges and future perspectives for the further advancement of each organ-on-chip device.
Collapse
|
73
|
Kunst RF, Niemeijer M, van der Laan LJW, Spee B, van de Graaf SFJ. From fatty hepatocytes to impaired bile flow: Matching model systems for liver biology and disease. Biochem Pharmacol 2020; 180:114173. [PMID: 32717228 DOI: 10.1016/j.bcp.2020.114173] [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: 06/01/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 02/08/2023]
Abstract
A large variety of model systems are used in hepatobiliary research. In this review, we aim to provide an overview of established and emerging models for specific research questions. We specifically discuss the value and limitations of these models for research on metabolic associated fatty liver disease (MAFLD), (previously named non-alcoholic fatty liver diseases/non-alcoholic steatohepatitis (NAFLD/NASH)) and cholestasis-related diseases such as primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC). The entire range of models is discussed varying from immortalized cell lines, mature or pluripotent stem cell-based models including organoids/spheroids, to animal models and human ex vivo models such as normothermic machine perfusion of livers and living liver slices. Finally, the pros and cons of each model are discussed as well as the need in the scientific community for continuous innovation in model development to better mimic the human (patho)physiology.
Collapse
Affiliation(s)
- Roni F Kunst
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Marije Niemeijer
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands; Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Bart Spee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Stan F J van de Graaf
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
74
|
Schimek K, Frentzel S, Luettich K, Bovard D, Rütschle I, Boden L, Rambo F, Erfurth H, Dehne EM, Winter A, Marx U, Hoeng J. Human multi-organ chip co-culture of bronchial lung culture and liver spheroids for substance exposure studies. Sci Rep 2020; 10:7865. [PMID: 32398725 PMCID: PMC7217973 DOI: 10.1038/s41598-020-64219-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 04/01/2020] [Indexed: 01/05/2023] Open
Abstract
Extrapolation of cell culture-based test results to in vivo effects is limited, as cell cultures fail to emulate organ complexity and multi-tissue crosstalk. Biology-inspired microphysiological systems provide preclinical insights into absorption, distribution, metabolism, excretion, and toxicity of substances in vitro by using human three-dimensional organotypic cultures. We co-cultured a human lung equivalent from the commercially available bronchial MucilAir culture and human liver spheroids from HepaRG cells to assess the potential toxicity of inhaled substances under conditions that permit organ crosstalk. We designed a new HUMIMIC Chip with optimized medium supply and oxygenation of the organ cultures and cultivated them on-chip for 14 days in separate culture compartments of a closed circulatory perfusion system, demonstrating the viability and homeostasis of the tissue cultures. A single-dose treatment of the hepatotoxic and carcinogenic aflatoxin B1 impaired functionality in bronchial MucilAir tissues in monoculture but showed a protective effect when the tissues were co-cultured with liver spheroids, indicating that crosstalk can be achieved in this new human lung–liver co-culture. The setup described here may be used to determine the effects of exposure to inhaled substances on a systemic level.
Collapse
Affiliation(s)
| | - Stefan Frentzel
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Karsta Luettich
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - David Bovard
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | | | - Laura Boden
- TissUse GmbH, Oudenarder Str. 16, 13347, Berlin, Germany
| | - Felix Rambo
- TissUse GmbH, Oudenarder Str. 16, 13347, Berlin, Germany
| | | | | | - Annika Winter
- TissUse GmbH, Oudenarder Str. 16, 13347, Berlin, Germany
| | - Uwe Marx
- TissUse GmbH, Oudenarder Str. 16, 13347, Berlin, Germany
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| |
Collapse
|
75
|
Lichtenstein D, Luckert C, Alarcan J, de Sousa G, Gioutlakis M, Katsanou ES, Konstantinidou P, Machera K, Milani ES, Peijnenburg A, Rahmani R, Rijkers D, Spyropoulou A, Stamou M, Stoopen G, Sturla SJ, Wollscheid B, Zucchini-Pascal N, Braeuning A, Lampen A. An adverse outcome pathway-based approach to assess steatotic mixture effects of hepatotoxic pesticides in vitro. Food Chem Toxicol 2020; 139:111283. [DOI: 10.1016/j.fct.2020.111283] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/06/2020] [Accepted: 03/16/2020] [Indexed: 12/29/2022]
|
76
|
In Vitro Systems for Studying Different Genotypes/Sub-Genotypes of Hepatitis B Virus: Strengths and Limitations. Viruses 2020; 12:v12030353. [PMID: 32210021 PMCID: PMC7150782 DOI: 10.3390/v12030353] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022] Open
Abstract
Hepatitis B virus (HBV) infects the liver resulting in end stage liver disease, cirrhosis, and hepatocellular carcinoma. Despite an effective vaccine, HBV poses a serious health problem globally, accounting for 257 million chronic carriers. Unique features of HBV, including its narrow virus-host range and its hepatocyte tropism, have led to major challenges in the development of suitable in vivo and in vitro model systems to recapitulate the HBV replication cycle and to test various antiviral strategies. Moreover, HBV is classified into at least nine genotypes and 35 sub-genotypes with distinct geographical distributions and prevalence, which have different natural histories of infection, clinical manifestation, and response to current antiviral agents. Here, we review various in vitro systems used to study the molecular biology of the different (sub)genotypes of HBV and their response to antiviral agents, and we discuss their strengths and limitations. Despite the advances made, no system is ideal for pan-genotypic HBV research or drug development and therefore further improvement is required. It is necessary to establish a centralized repository of HBV-related generated materials, which are readily accessible to HBV researchers, with international collaboration toward advancement and development of in vitro model systems for testing new HBV antivirals to ensure their pan-genotypic and/or customized activity.
Collapse
|
77
|
Cellular levels and molecular dynamics simulations of estragole DNA adducts point at inefficient repair resulting from limited distortion of the double-stranded DNA helix. Arch Toxicol 2020; 94:1349-1365. [PMID: 32185416 PMCID: PMC7225201 DOI: 10.1007/s00204-020-02695-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 03/02/2020] [Indexed: 10/25/2022]
Abstract
Estragole, naturally occurring in a variety of herbs and spices, can form DNA adducts after bioactivation. Estragole DNA adduct formation and repair was studied in in vitro liver cell models, and a molecular dynamics simulation was used to investigate the conformation dependent (in)efficiency of N2-(trans-isoestragol-3'-yl)-2'-deoxyguanosine (E-3'-N2-dG) DNA adduct repair. HepG2, HepaRG cells, primary rat hepatocytes and CHO cells (including CHO wild-type and three NER-deficient mutants) were exposed to 50 μM estragole or 1'-hydroxyestragole and DNA adduct formation was quantified by LC-MS immediately following exposure and after a period of repair. Results obtained from CHO cell lines indicated that NER plays a role in repair of E-3'-N2-dG adducts, however, with limited efficiency since in the CHO wt cells 80% DNA adducts remained upon 24 h repair. Inefficiency of DNA repair was also found in HepaRG cells and primary rat hepatocytes. Changes in DNA structure resulting from E-3'-N2-dG adduct formation were investigated by molecular dynamics simulations. Results from molecular dynamics simulations revealed that conformational changes in double-stranded DNA by E-3'-N2-dG adduct formation are small, providing a possible explanation for the restrained repair, which may require larger distortions in the DNA structure. NER-mediated enzymatic repair of E-3'-N2-dG DNA adducts upon exposure to estragole will be limited, providing opportunities for accumulation of damage upon repeated daily exposure. The inability of this enzymatic repair is likely due to a limited distortion of the DNA double-stranded helix resulting in inefficient activation of nucleotide excision repair.
Collapse
|
78
|
Ngo LP, Owiti NA, Swartz C, Winters J, Su Y, Ge J, Xiong A, Han J, Recio L, Samson LD, Engelward B. Sensitive CometChip assay for screening potentially carcinogenic DNA adducts by trapping DNA repair intermediates. Nucleic Acids Res 2020; 48:e13. [PMID: 31822921 PMCID: PMC7026589 DOI: 10.1093/nar/gkz1077] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 10/08/2019] [Accepted: 11/19/2019] [Indexed: 12/27/2022] Open
Abstract
Genotoxicity testing is critical for predicting adverse effects of pharmaceutical, industrial, and environmental chemicals. The alkaline comet assay is an established method for detecting DNA strand breaks, however, the assay does not detect potentially carcinogenic bulky adducts that can arise when metabolic enzymes convert pro-carcinogens into a highly DNA reactive products. To overcome this, we use DNA synthesis inhibitors (hydroxyurea and 1-β-d-arabinofuranosyl cytosine) to trap single strand breaks that are formed during nucleotide excision repair, which primarily removes bulky lesions. In this way, comet-undetectable bulky lesions are converted into comet-detectable single strand breaks. Moreover, we use HepaRG™ cells to recapitulate in vivo metabolic capacity, and leverage the CometChip platform (a higher throughput more sensitive comet assay) to create the 'HepaCometChip', enabling the detection of bulky genotoxic lesions that are missed by current genotoxicity screens. The HepaCometChip thus provides a broadly effective approach for detection of bulky DNA adducts.
Collapse
Affiliation(s)
- Le P Ngo
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Norah A Owiti
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Carol Swartz
- Toxicology Program, Integrated Laboratory Systems, Inc., Research Triangle Park, NC 27560, USA
| | - John Winters
- Toxicology Program, Integrated Laboratory Systems, Inc., Research Triangle Park, NC 27560, USA
| | - Yang Su
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jing Ge
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Aoli Xiong
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology, 138602 Singapore
| | - Jongyoon Han
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology, 138602 Singapore
- Department of Electrical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Leslie Recio
- Toxicology Program, Integrated Laboratory Systems, Inc., Research Triangle Park, NC 27560, USA
| | - Leona D Samson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bevin P Engelward
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
79
|
Braeuning A, Mentz A, Schmidt FF, Albaum SP, Planatscher H, Kalinowski J, Joos TO, Poetz O, Lichtenstein D. RNA-protein correlation of liver toxicity markers in HepaRG cells. EXCLI JOURNAL 2020; 19:135-153. [PMID: 32194361 PMCID: PMC7068204 DOI: 10.17179/excli2019-2005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/15/2020] [Indexed: 12/23/2022]
Abstract
The liver is a main target organ for the toxicity of many different compounds. While in general, in vivo testing is still routinely used for assessing the hepatotoxic potential of test chemicals, the use of in vitro models offers advantages with regard to throughput, consumption of resources, and animal welfare aspects. Using the human hepatoma cell line HepaRG, we performed a comparative evaluation of a panel of hepatotoxicity marker mRNAs and proteins after exposure of the cells to 30 different pesticidal active compounds comprising herbizides, fungicides, insecticides, and others. The panel of hepatotoxicity markers included nuclear receptor target genes, key players of fatty acid and bile acid metabolism-related pathways, as well as recently identified biomarkers of drug-induced liver injury. Moreover, marker genes and proteins were identified, for example, S100P, ANXA10, CYP1A1, and CYP7A1. These markers respond with high sensitivity to stimulation with chemically diverse test compounds already at non-cytotoxic concentrations. The potency of the test compounds, determined as an overall parameter of their ability to deregulate marker expression in vitro, was very similar between the mRNA and protein levels. Thus, this study does not only characterize the response of human liver cells to 30 different pesticides but also demonstrates that hepatotoxicity testing in human HepaRG cells yields well comparable results at the mRNA and protein levels. Furthermore, robust hepatotoxicity marker genes and proteins were identified in HepaRG cells.
Collapse
Affiliation(s)
- Albert Braeuning
- German Federal Institute for Risk Assessment, Dept. Food Safety, Berlin, Germany
| | - Almut Mentz
- Center for Biotechnology (CeBiTec), Universität Bielefeld, Bielefeld, Germany
| | | | - Stefan P. Albaum
- Center for Biotechnology (CeBiTec), Universität Bielefeld, Bielefeld, Germany
| | | | - Jörn Kalinowski
- Center for Biotechnology (CeBiTec), Universität Bielefeld, Bielefeld, Germany
| | - Thomas O. Joos
- Signatope GmbH, Reutlingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Tübingen,Germany
| | - Oliver Poetz
- Signatope GmbH, Reutlingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Tübingen,Germany
| | - Dajana Lichtenstein
- German Federal Institute for Risk Assessment, Dept. Food Safety, Berlin, Germany
| |
Collapse
|
80
|
Ishida S. Requirements for designing organ-on-a-chip platforms to model the pathogenesis of liver disease. ORGAN-ON-A-CHIP 2020:181-213. [DOI: 10.1016/b978-0-12-817202-5.00005-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
81
|
Franco ME, Sutherland GE, Fernandez-Luna MT, Lavado R. Altered expression and activity of phase I and II biotransformation enzymes in human liver cells by perfluorooctanoate (PFOA) and perfluorooctane sulfonate (PFOS). Toxicology 2020; 430:152339. [DOI: 10.1016/j.tox.2019.152339] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 11/26/2019] [Accepted: 12/02/2019] [Indexed: 01/19/2023]
|
82
|
Thompson KJ, Wessling-Resnick M. ZIP14 is degraded in response to manganese exposure. Biometals 2019; 32:829-843. [PMID: 31541377 PMCID: PMC7755095 DOI: 10.1007/s10534-019-00216-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/10/2019] [Indexed: 12/16/2022]
Abstract
Manganese (Mn) is an essential element necessary for proper development and brain function. Circulating Mn levels are regulated by hepatobiliary clearance to limit toxic levels and prevent tissue deposition. To characterize mechanisms involved in hepatocyte Mn uptake, polarized human HepaRG cells were used for this study. Western blot analysis and immunofluorescence microscopy showed the Mn transporter ZIP14 was expressed and localized to the basolateral surface of polarized HepaRG cells. HepaRG cells took up 54Mn in a time- and temperature-dependent manner but uptake was reduced after exposure to Mn. This loss in transport activity was associated with decreased ZIP14 protein levels in response to Mn exposure. Mn-induced degradation of ZIP14 was blocked by bafilomycin A1, which increased localization of the transporter in Lamp1-positive vesicles. Mn exposure also down-regulated the Golgi proteins TMEM165 and GPP130 while the ER stress marker BiP was induced. These results indicate that Mn exposure decreases ZIP14 protein levels to limit subsequent uptake of Mn as a cytoprotective response. Thus, high levels of Mn may compromise first-pass-hepatic clearance mechanisms.
Collapse
Affiliation(s)
- Khristy J Thompson
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.
| | - Marianne Wessling-Resnick
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
| |
Collapse
|
83
|
Corbett JL, Duncan SA. iPSC-Derived Hepatocytes as a Platform for Disease Modeling and Drug Discovery. Front Med (Lausanne) 2019; 6:265. [PMID: 31803747 PMCID: PMC6873655 DOI: 10.3389/fmed.2019.00265] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/30/2019] [Indexed: 12/15/2022] Open
Abstract
The liver is one of the largest organs in the body and is responsible for a diverse repertoire of metabolic processes. Such processes include the secretion of serum proteins, carbohydrate and lipid metabolism, bile acid and urea synthesis, detoxification of drugs and metabolic waste products, and vitamin and carbohydrate storage. Currently, liver disease is one of the most prevalent causes of mortality in the USA with congenital liver defects contributing to a significant proportion of these deaths. Historically the study of liver disease has been hampered by a shortage of organ donors, the subsequent scarcity of healthy tissue, and the failure of animal models to fully recapitulate human liver function. In vitro culture of hepatocytes has also proven difficult because primary hepatocytes rapidly de-differentiate in culture. Recent advances in stem cell technology have facilitated the generation of induced pluripotent stem cells (iPSCs) from various somatic cell types from patients. Such cells can be differentiated to a liver cell fate, essentially providing a limitless supply of cells with hepatocyte characteristics that can mimic the pathophysiology of liver disease. Furthermore, development of the CRISPR-Cas9 system, as well as advancement of miniaturized differentiation platforms has facilitated the development of high throughput models for the investigation of hepatocyte differentiation and drug discovery. In this review, we will explore the latest advances in iPSC-based disease modeling and drug screening platforms and examine how this technology is being used to identify new pharmacological interventions, and to advance our understanding of liver development and mechanisms of disease. We will cover how iPSC technology is being used to develop predictive models for rare diseases and how information gained from large in vitro screening experiments can be used to directly inform clinical investigation.
Collapse
Affiliation(s)
| | - Stephen A. Duncan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
84
|
Leblanc AF, Attignon EA, Distel E, Karakitsios SP, Sarigiannis DA, Bortoli S, Barouki R, Coumoul X, Aggerbeck M, Blanc EB. A dual mixture of persistent organic pollutants modifies carbohydrate metabolism in the human hepatic cell line HepaRG. ENVIRONMENTAL RESEARCH 2019; 178:108628. [PMID: 31520823 DOI: 10.1016/j.envres.2019.108628] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 07/12/2019] [Accepted: 08/04/2019] [Indexed: 06/10/2023]
Abstract
Individuals as well as entire ecosystems are exposed to mixtures of Persistent Organic Pollutants (POPs). Previously, we showed, by a non-targeted approach, that the expression of several genes involved in carbohydrate metabolism was almost completely inhibited in the human hepatic cell line HepaRG following exposure to a mixture of the organochlorine insecticide alpha-endosulfan and 2,3,7,8 tetrachlorodibenzo-p-dioxin. In this European HEALS project, which studies the effects of the exposome on human health, we used a Physiologically Based BioKinetic model to compare the concentrations previously used in vitro with in vivo exposures for humans. We investigated the effects of these POPs on the levels of proteins, on glycogen content, glucose production and the oxidation of glucose into CO2 and correlated them to the expression of genes involved in carbohydrate metabolism as measured by RT-qPCR. Exposure to individual POPs and the mixture decreased the expression of the proteins investigated as well as glucose output (up to 82%), glucose oxidation (up to 29%) and glycogen content (up to 48%). siRNAs that specifically inhibit the expression of several xenobiotic receptors were used to assess receptor involvement in the effects of the POPs. In the HepaRG model, we demonstrate that the effects are mediated by the aryl hydrocarbon receptor and the estrogen receptor alpha, but not the pregnane X receptor or the constitutive androstane receptor. These results provide evidence that exposure to combinations of POPs, acting through different signaling pathways, may affect, more profoundly than single pollutants alone, metabolic pathways such as carbohydrate/energy metabolism and play a potential role in pollutant associated metabolic disorders.
Collapse
Affiliation(s)
- Alix F Leblanc
- INSERM UMR-S 1124, Toxicité Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, 45 rue des Saints Pères, 75006, Paris, France; Université de Paris, Université Paris Descartes, 45 rue des Saints Pères, 75006, Paris, France.
| | - Eléonore A Attignon
- INSERM UMR-S 1124, Toxicité Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, 45 rue des Saints Pères, 75006, Paris, France; Université de Paris, Université Paris Descartes, 45 rue des Saints Pères, 75006, Paris, France.
| | - Emilie Distel
- INSERM UMR-S 1124, Toxicité Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, 45 rue des Saints Pères, 75006, Paris, France; Université de Paris, Université Paris Descartes, 45 rue des Saints Pères, 75006, Paris, France.
| | - Spyros P Karakitsios
- Aristotle University of Thessaloniki, Department of Chemical Engineering, 54 124, Thessaloniki, Greece.
| | - Dimosthenis A Sarigiannis
- Aristotle University of Thessaloniki, Department of Chemical Engineering, 54 124, Thessaloniki, Greece; Environmental Health Engineering, Institute for Advanced Study, Pavia, Italy.
| | - Sylvie Bortoli
- INSERM UMR-S 1124, Toxicité Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, 45 rue des Saints Pères, 75006, Paris, France; Université de Paris, Université Paris Descartes, 45 rue des Saints Pères, 75006, Paris, France.
| | - Robert Barouki
- INSERM UMR-S 1124, Toxicité Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, 45 rue des Saints Pères, 75006, Paris, France; Université de Paris, Université Paris Descartes, 45 rue des Saints Pères, 75006, Paris, France; AP-HP, Hôpital Necker-Enfants Malades, Service de Biochimie Métabolique, 149, rue de Sèvres, 75743, Paris, France.
| | - Xavier Coumoul
- INSERM UMR-S 1124, Toxicité Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, 45 rue des Saints Pères, 75006, Paris, France; Université de Paris, Université Paris Descartes, 45 rue des Saints Pères, 75006, Paris, France.
| | - Martine Aggerbeck
- INSERM UMR-S 1124, Toxicité Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, 45 rue des Saints Pères, 75006, Paris, France; Université de Paris, Université Paris Descartes, 45 rue des Saints Pères, 75006, Paris, France.
| | - Etienne B Blanc
- INSERM UMR-S 1124, Toxicité Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, 45 rue des Saints Pères, 75006, Paris, France; Université de Paris, Université Paris Descartes, 45 rue des Saints Pères, 75006, Paris, France.
| |
Collapse
|
85
|
Qi Y, Yao X, Du X. Midazolam inhibits proliferation and accelerates apoptosis of hepatocellular carcinoma cells by elevating microRNA-124-3p and suppressing PIM-1. IUBMB Life 2019; 72:452-464. [PMID: 31651086 DOI: 10.1002/iub.2171] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 09/05/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Recently, the impact of microRNAs (miRNAs) has been identified in hepatocellular carcinoma (HCC), this study was designed to assess the effects of miR-124-3p and midazolam (MDZ) in HCC with the involvement of PIM-1. METHODS HepG2 human HCC cells were selected for our study, which were treated with different concentrations of MDZ. The gain- and loss-of-function experiments were performed to elucidate the migration, invasion, proliferation, colony formation ability, cell cycle, and apoptosis of HepG2 cells upon treatment of MDZ, miR-124-3p mimics, or miR-124-3p inhibitor. The expression levels of miR-124-3p, PIM-1, Bax, Bcl-2, P21, and Ki-67 in HepG2 cells were assessed by reverse transcription quantitative polymerase chain reaction and western blot analysis. Moreover, HepG2 cell growth in vivo was measured by subcutaneous tumorigenesis in nude mice, and the target relation between miR-124-3p and PIM-1 was evaluated using dual luciferase reporter gene assay. RESULTS We have found that after treated with overexpression of miR-124-3p and MDZ, there exhibited elevated miR-124-3p, declined expression of PIM-1, attenuated migration, invasion, proliferation and colony formation ability, and promoted apoptosis of HepG2 cells. Additionally, it could be observed that the tumor volume and weight were all reduced upon treatment of overexpression of miR-124-3p and MDZ. Meanwhile, the results in the HepG2 cells that treated with down-regulated miR-124-3p were the opposite. Furthermore, PIM-1 was found to be a target gene of miR-124-3p. CONCLUSION Our study found that MDZ could inhibit proliferation and accelerate apoptosis of HCC cells by elevation of miR-124-3p and suppressing PIM-1, which may be an effective method in the treatment of HCC.
Collapse
Affiliation(s)
- Yanyan Qi
- Anesthesiology Department, Henan Province People's hospital, Zhengzhou, Henan, People's Republic of China
| | - Xiangyan Yao
- Anesthesiology Department, Henan Province People's hospital, Zhengzhou, Henan, People's Republic of China
| | - Xianhui Du
- Anesthesiology Department, Henan Province People's hospital, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
86
|
Forsch K, Schöning V, Assmann GM, Moser C, Siewert B, Butterweck V, Drewe J. In vitro hepatotoxicity of Petasites hybridus extract (Ze 339) depends on the concentration, the cytochrome activity of the cell system, and the species used. Phytother Res 2019; 34:184-192. [PMID: 31631423 PMCID: PMC7004140 DOI: 10.1002/ptr.6516] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/19/2019] [Accepted: 09/05/2019] [Indexed: 12/24/2022]
Abstract
Ze 339, a CO2 extract prepared from the leaves of Petasites hybridus, possesses antispasmodic and anti‐inflammatory effects and is proven to be effective in the treatment of allergic rhinitis. To study possible hepatotoxic effects of Ze 339, its main constituents and metabolites, a series of in vitro investigations were performed. Furthermore, different reconstituted fractions of extract (petasins and fatty acid fraction) were examined in three in vitro test systems using hepatocytes: Two human cell lines, with lower and higher activity of cytochrome P450 enzymes (HepG2, HepaRG) as well as a rodent cell line with high cytochrome P450 activity (H‐4‐II‐E), were used. Metabolic activity, assessed by the WST‐1 assay, was chosen as indicator of cytotoxicity. To assess potential bioactivation of Ze 339 compounds, metabolic experiments using S9 fractions from rats, dogs, and humans and isolated cytochromes (human/rat) were performed, and the formation of reactive metabolites was assessed by measuring cellular concentrations of glutathione and glutathione disulphide. Our data revealed that the cytotoxicity of Ze 339, its single constituents, and main metabolites depends on the concentration, the cytochrome activity of the cell system, and the species used.
Collapse
Affiliation(s)
- Kristina Forsch
- Preclinical Research, Max Zeller Söhne AG, Romanshorn, Switzerland
| | - Verena Schöning
- Preclinical Research, Max Zeller Söhne AG, Romanshorn, Switzerland
| | | | - Christin Moser
- Preclinical Research, Max Zeller Söhne AG, Romanshorn, Switzerland
| | - Beate Siewert
- Preclinical Research, Max Zeller Söhne AG, Romanshorn, Switzerland
| | | | - Jürgen Drewe
- Preclinical Research, Max Zeller Söhne AG, Romanshorn, Switzerland
| |
Collapse
|
87
|
Deferm N, De Vocht T, Qi B, Van Brantegem P, Gijbels E, Vinken M, de Witte P, Bouillon T, Annaert P. Current insights in the complexities underlying drug-induced cholestasis. Crit Rev Toxicol 2019; 49:520-548. [PMID: 31589080 DOI: 10.1080/10408444.2019.1635081] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Drug-induced cholestasis (DIC) poses a major challenge to the pharmaceutical industry and regulatory agencies. It causes both drug attrition and post-approval withdrawal of drugs. DIC represents itself as an impaired secretion and flow of bile, leading to the pathological hepatic and/or systemic accumulation of bile acids (BAs) and their conjugate bile salts. Due to the high number of mechanisms underlying DIC, predicting a compound's cholestatic potential during early stages of drug development remains elusive. A profound understanding of the different molecular mechanisms of DIC is, therefore, of utmost importance. Although many knowledge gaps and caveats still exist, it is generally accepted that alterations of certain hepatobiliary membrane transporters and changes in hepatocellular morphology may cause DIC. Consequently, liver models, which represent most of these mechanisms, are valuable tools to predict human DIC. Some of these models, such as membrane-based in vitro models, are exceptionally well-suited to investigate specific mechanisms (i.e. transporter inhibition) of DIC, while others, such as liver slices, encompass all relevant biological processes and, therefore, offer a better representation of the in vivo situation. In the current review, we highlight the principal molecular mechanisms associated with DIC and offer an overview and critical appraisal of the different liver models that are currently being used to predict the cholestatic potential of drugs.
Collapse
Affiliation(s)
- Neel Deferm
- Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Tom De Vocht
- Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Bing Qi
- Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Pieter Van Brantegem
- Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Eva Gijbels
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mathieu Vinken
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Peter de Witte
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Thomas Bouillon
- Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Pieter Annaert
- Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| |
Collapse
|
88
|
Mbarik M, Poirier SJ, Doiron J, Selka A, Barnett DA, Cormier M, Touaibia M, Surette ME. Phenolic acid phenethylesters and their corresponding ketones: Inhibition of 5-lipoxygenase and stability in human blood and HepaRG cells. Pharmacol Res Perspect 2019; 7:e00524. [PMID: 31523435 PMCID: PMC6743424 DOI: 10.1002/prp2.524] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 08/27/2019] [Accepted: 08/27/2019] [Indexed: 02/07/2023] Open
Abstract
5-lipoxygenase (5-LO) catalyzes the biosynthesis of leukotrienes, potent lipid mediators involved in inflammatory diseases, and both 5-LO and the leukotrienes are validated therapeutic targets. Caffeic acid phenethyl ester (CAPE) is an effective inhibitor of 5-LO and leukotriene biosynthesis but is susceptible to hydrolysis by esterases. In this study a number of CAPE analogues were synthesized with modifications to the caffeoyl moiety and the replacement of the ester linkage with a ketone. Several new molecules showed better inhibition of leukotriene biosynthesis than CAPE in isolated human neutrophils and in whole blood with IC50 values in the nanomolar (290-520 nmol/L) and low micromolar (1.0-2.3 µmol/L) ranges, respectively. Sinapic acid and 2,5-dihydroxy derivatives were more stable than CAPE in whole blood, and ketone analogues were degraded more slowly in HepaRG hepatocyte cultures than esters. All compounds underwent modification consistent with glucuronidation in HepaRG cultures as determined using LC-MS/MS analysis, though the modified sinapoyl ketone (10) retained 50% of its inhibitory activity after up to one hour of incubation. This study has identified at least one CAPE analogue, compound 10, that shows favorable properties that warrant further in vivo investigation as an antiinflammatory compound.
Collapse
Affiliation(s)
- Maroua Mbarik
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNBCanada
| | - Samuel J. Poirier
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNBCanada
| | - Jérémie Doiron
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNBCanada
| | - Ayyoub Selka
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNBCanada
| | | | - Marc Cormier
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNBCanada
| | - Mohamed Touaibia
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNBCanada
| | - Marc E. Surette
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNBCanada
| |
Collapse
|
89
|
Mišík M, Nersesyan A, Ropek N, Huber WW, Haslinger E, Knasmueller S. Use of human derived liver cells for the detection of genotoxins in comet assays. MUTATION RESEARCH/GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2019; 845:402995. [DOI: 10.1016/j.mrgentox.2018.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 12/03/2018] [Accepted: 12/09/2018] [Indexed: 04/09/2023]
|
90
|
Chong LH, Ng C, Li H, Tian EF, Ananthanarayanan A, McMillian M, Toh YC. Hepatic Bioactivation of Skin-Sensitizing Drugs to Immunogenic Reactive Metabolites. ACS OMEGA 2019; 4:13902-13912. [PMID: 31497708 PMCID: PMC6714514 DOI: 10.1021/acsomega.9b01551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 07/30/2019] [Indexed: 06/10/2023]
Abstract
The clinical use of some drugs, such as carbamazepine, phenytoin, and allopurinol, is often associated with adverse cutaneous reactions. The bioactivation of drugs into immunologically reactive metabolites by the liver is postulated to be the first step in initiating a downstream cascade of pathological immune responses. Current mechanistic understanding and the ability to predict such adverse drug cutaneous responses have been partly limited by the lack of appropriate cutaneous drug bioactivation experimental models. Although in vitro human liver models have been extensively investigated for predicting hepatotoxicity and drug-drug interactions, their ability to model the generation of antigenic reactive drug metabolites that are capable of eliciting immunological reactions is not well understood. Here, we employed a human progenitor cell (HepaRG)-derived hepatocyte model and established highly sensitive liquid chromatography-mass spectrometry analytical assays to generate and quantify different reactive metabolite species of three paradigm skin sensitizers, namely, carbamazepine, phenytoin, and allopurinol. We found that the generation of reactive drug metabolites by the HepaRG-hepatocytes was sensitive to the medium composition. In addition, a functional assay based on the activation of U937 myeloid cells into the antigen-presenting cell (APC) phenotype was established to evaluate the immunogenicity potential of the reactive drug metabolites produced by HepaRG-derived hepatocytes. We showed that the reactive drug metabolites of known skin sensitizers could significantly upregulate IL8, IL1β, and CD86 expressions in U937 cells compared to the metabolites from a nonskin sensitizer (i.e., acetaminophen). Thus, the extent of APC activation by HepaRG-hepatocytes conditioned medium containing reactive drug metabolites can potentially be used to predict their skin sensitization potential.
Collapse
Affiliation(s)
- Lor Huai Chong
- Department
of Biomedical Engineering, National University
of Singapore, 4 Engineering Drive 3, #04-08, Singapore 117583
| | - Celine Ng
- School
of Applied Science, Temasek Polytechnic, Tampines Avenue 1, Singapore 529765
| | - Huan Li
- School
of Applied Science, Temasek Polytechnic, Tampines Avenue 1, Singapore 529765
| | - Edmund Feng Tian
- School
of Applied Science, Temasek Polytechnic, Tampines Avenue 1, Singapore 529765
| | | | - Michael McMillian
- Invitrocue
Pte Ltd, 11, Biopolis
Way, Helios #12-07/08, Singapore 138667
- Department
of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, MD9, #04-11, Singapore 117597
| | - Yi-Chin Toh
- Department
of Biomedical Engineering, National University
of Singapore, 4 Engineering Drive 3, #04-08, Singapore 117583
- Institute
for Health Innovation and Technology (iHealthtech), National University of Singapore, MD6, 14 Medical Drive, #14-01, Singapore 117599
- The
N.1 Institute for Health, 28 Medical Drive, #05-corridor, Singapore 117456
- NUS
Tissue Engineering Programme, National University
of Singapore, 28 Medical
Drive, Singapore 117456
| |
Collapse
|
91
|
Three-dimensional HepaRG spheroids as a liver model to study human genotoxicity in vitro with the single cell gel electrophoresis assay. Sci Rep 2019; 9:10548. [PMID: 31332230 PMCID: PMC6646340 DOI: 10.1038/s41598-019-47114-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/08/2019] [Indexed: 12/12/2022] Open
Abstract
Many efforts have been made in the last 30 years to develop more relevant in vitro models to study genotoxic responses of drugs and environmental contaminants. While 2D HepaRG cells are one of the most promising models for liver toxicology, a switch to 3D cultures that integrate both in vivo architecture and cell-cell interactions has occurred to achieve even more predictive models. Preliminary studies have indicated that 3D HepaRG cells are suitable for liver toxicity screening. Our study aimed to evaluate the response of HepaRG spheroids exposed to various genotoxic compounds using the single cell gel electrophoresis assay. HepaRG spheroids were used at 10 days after seeding and exposed for 24 and 48 hours to certain selected chemical compounds (methylmethansulfonate (MMS), etoposide, benzo[a]pyrene (B[a]P), cyclophosphamide (CPA), 7,12-dimethylbenz[a]anthracene (DMBA), 2-acetylaminofluorene (2-AAF), 4-nitroquinoline (4-NQO), 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), 2-amino-3-methylimidazo[4,5-f]quinolone (IQ), acrylamide, and 2-4-diaminotoluene (2,4-DAT)). After treatment, the comet assay was performed on single cell suspensions and cytotoxicity was determined by the ATP assay. Comet formation was observed for all compounds except IQ, etoposide and 2,4-DAT. Treatment of spheroids with rifampicin increased CYP3A4 activity, demonstrating the metabolic capacity of HepaRG spheroids. These data on genotoxicity in 3D HepaRG spheroids are promising, but further experiments are required to prove that this model can improve the predictivity of in vitro models to detect human carcinogens.
Collapse
|
92
|
Comparison of three human liver cell lines for in vitro drug-induced liver injury assessment: Huh7, HepaRG, and stem cell-derived hepatocytes. Mol Cell Toxicol 2019. [DOI: 10.1007/s13273-019-0031-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
93
|
Hu L, Wu F, He J, Zhong L, Song Y, Shao H. Cytotoxicity of safrole in HepaRG cells: studies on the role of CYP1A2-mediated ortho-quinone metabolic activation. Xenobiotica 2019; 49:1504-1515. [PMID: 30865484 DOI: 10.1080/00498254.2019.1590882] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Linlin Hu
- The Pharmaceutical Department, Nanjing Zhong-da Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Fei Wu
- Engineering Research Center of Modern Preparation Technology of TCM of Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Jie He
- The Pharmaceutical Department, Nanjing Zhong-da Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Lingjun Zhong
- The Pharmaceutical Department, Nanjing Zhong-da Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Yifan Song
- The Pharmaceutical Department, Nanjing Zhong-da Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Hua Shao
- The Pharmaceutical Department, Nanjing Zhong-da Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| |
Collapse
|
94
|
Koeda A, Iwao T, Nakanishi A, Mizuno S, Yamashita M, Sakai Y, Nakamura K, Matsunaga T. Comparison of mRNA expression profiles of drug-metabolizing enzymes and transporters in fresh and cryopreserved cynomolgus monkey hepatocytes. Drug Metab Pharmacokinet 2019; 34:253-263. [PMID: 31174977 DOI: 10.1016/j.dmpk.2019.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 03/12/2019] [Accepted: 04/05/2019] [Indexed: 02/08/2023]
Abstract
In this study, freshly isolated and cryopreserved cynomolgus monkey hepatocytes were seeded on Cell-able® plates with feeder cells to form spheroids and were cultured for 28 days. As a control, hepatocytes were also cultured with or without feeder cells on collagen-coated plates. We verified the mRNA expression levels of drug-metabolizing enzyme-related genes and the leakage of enzymes (AST, ALT, LDH, and γ-GTP) as indicators of cell survival. As a result, the patterns of target mRNA expression in fresh and cryopreserved hepatocytes were very similar during the culture period between culture methods. mRNA expression levels were highly maintained at day 28 using the 3D spheroid and co-culture methods, demonstrating that these methods are useful for maintenance of liver function. Leakage of AST and ALT was higher at day 3 but decreased at day 14. LDH was not detected, suggesting that the cell viability was also maintained during the culture period. Furthermore, the functional differences between fresh and cryopreserved hepatocytes were not clearly detected. The co-culture method was useful for long-term culture not requiring 3D structure, and the 3D spheroid culture method was effective as well. With these techniques, cynomolgus monkey hepatocytes are expected to exhibit smaller individual differences and high reproducibility.
Collapse
Affiliation(s)
- Akiko Koeda
- Ina Research Inc., Ina, Japan; Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | - Takahiro Iwao
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan; Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | - Anna Nakanishi
- Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | - Shota Mizuno
- Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | - Misaki Yamashita
- Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | - Yoko Sakai
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | | | - Tamihide Matsunaga
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan; Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| |
Collapse
|
95
|
Kamalian L, Douglas O, Jolly CE, Snoeys J, Simic D, Monshouwer M, Williams DP, Park BK, Chadwick AE. Acute Metabolic Switch Assay Using Glucose/Galactose Medium in HepaRG Cells to Detect Mitochondrial Toxicity. CURRENT PROTOCOLS IN TOXICOLOGY 2019; 80:e76. [PMID: 31058461 DOI: 10.1002/cptx.76] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Using galactose instead of glucose in the culture medium of hepatoma cell lines, such as HepG2 cells, has been utilized for a decade to unmask the mitochondrial liability of chemical compounds. A modified glucose-galactose assay on HepG2 cells, reducing the experimental period for screening of mitochondrial toxicity to 2 to 4 hr, has been previously reported. HepaRG cells are one of the few cell lines that retain some of the important characteristics of human hepatocytes, offering advantages of working with a cell line, therefore, are considered an alternative for HepG2 cells in drug toxicity screening. A method is described here using HepaRG cells in an acute metabolic switch assay utilizing specific glucose/galactose media, a combined ATP-protein-LDH assay measuring three endpoints from one 96-well plate, and a criteria to label a compound as a mitochondrial toxin. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Laleh Kamalian
- MRC Centre for Drug Safety Science, The Department of Clinical and Molecular Pharmacology, The University of Liverpool, Liverpool, United Kingdom
| | - Oisin Douglas
- MRC Centre for Drug Safety Science, The Department of Clinical and Molecular Pharmacology, The University of Liverpool, Liverpool, United Kingdom
| | - Carol E Jolly
- MRC Centre for Drug Safety Science, The Department of Clinical and Molecular Pharmacology, The University of Liverpool, Liverpool, United Kingdom
| | - Jan Snoeys
- Drug Metabolism and Pharmacokinetics, Janssen Research and Development, Beerse, Belgium
| | - Damir Simic
- Mechanistic and Investigative Toxicology, Janssen Research and Development, Spring House, Pennsylvania
| | - Mario Monshouwer
- Drug Metabolism and Pharmacokinetics, Janssen Research and Development, Beerse, Belgium
| | - Dominic P Williams
- Innovative Medicines and Early Development, Drug Safety and Metabolism, Translational Safety, AstraZeneca, Cambridge, United Kingdom
| | - B Kevin Park
- MRC Centre for Drug Safety Science, The Department of Clinical and Molecular Pharmacology, The University of Liverpool, Liverpool, United Kingdom
| | - Amy E Chadwick
- MRC Centre for Drug Safety Science, The Department of Clinical and Molecular Pharmacology, The University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
96
|
Vlach M, Quesnot N, Dubois-Pot-Schneider H, Ribault C, Verres Y, Petitjean K, Rauch C, Morel F, Robin MA, Corlu A, Loyer P. Cytochrome P450 1A1/2, 2B6 and 3A4 HepaRG Cell-Based Biosensors to Monitor Hepatocyte Differentiation, Drug Metabolism and Toxicity. SENSORS 2019; 19:s19102245. [PMID: 31096615 PMCID: PMC6567340 DOI: 10.3390/s19102245] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 01/20/2023]
Abstract
Human hepatoma HepaRG cells express most drug metabolizing enzymes and constitute a pertinent in vitro alternative cell system to primary cultures of human hepatocytes in order to determine drug metabolism and evaluate the toxicity of xenobiotics. In this work, we established novel transgenic HepaRG cells transduced with lentiviruses encoding the reporter green fluorescent protein (GFP) transcriptionally regulated by promoter sequences of cytochromes P450 (CYP) 1A1/2, 2B6 and 3A4 genes. Here, we demonstrated that GFP-biosensor transgenes shared similar expression patterns with the corresponding endogenous CYP genes during proliferation and differentiation in HepaRG cells. Interestingly, differentiated hepatocyte-like HepaRG cells expressed GFP at higher levels than cholangiocyte-like cells. Despite weaker inductions of GFP expression compared to the strong increases in mRNA levels of endogenous genes, we also demonstrated that the biosensor transgenes were induced by prototypical drug inducers benzo(a)pyrene and phenobarbital. In addition, we used the differentiated biosensor HepaRG cells to evidence that pesticide mancozeb triggered selective cytotoxicity of hepatocyte-like cells. Our data demonstrate that these new biosensor HepaRG cells have potential applications in the field of chemicals safety evaluation and the assessment of drug hepatotoxicity.
Collapse
Affiliation(s)
- Manuel Vlach
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Nicolas Quesnot
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | | | - Catherine Ribault
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Yann Verres
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Kilian Petitjean
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Claudine Rauch
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Fabrice Morel
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Marie-Anne Robin
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Anne Corlu
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Pascal Loyer
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
- Correspondence: ; Tel.: +33-(0)223233873; Fax: +33-(0)299540137
| |
Collapse
|
97
|
Abstract
Human hepatocytes possess a wider range of phase I and II drug-metabolizing enzyme activities than other liver tissue-derived products, such as human liver microsomes. Thus, hepatocytes may be useful for predicting the in vivo metabolic fate of new drugs of abuse in humans. Recently, new types of human hepatocytes have been made commercially available for use in drug metabolism studies, such as a liver tumor-derived cell line (HepaRG), and a human induced pluripotent stem cell-derived hepatocyte (h-iPS-HEP). In our laboratory, HepaRG has been used to elucidate the metabolic pathways of XLR-11, a synthetic cannabinoid, and its thermal degradant. In addition, the potential of h-iPS-HEP to metabolize drugs was assessed using fentanyl as a model drug, and indeed, h-iPS-HEP exhibited a pattern for fentanyl metabolite formation similar to that observed in vivo. In addition, the phase I and II drug-metabolizing enzyme activities of HepaRG, h-iPS-HEP, liver-humanized mouse-derived hepatocytes (PXB-cellsTM), and human primary hepatocytes were evaluated and compared. HepaRG showed high phase I and II drug metabolism activities; however, the CYP2D6 activity in these cells was quite low, and therefore h-iPS-HEP lacked O-methylation and conjugation activities. PXB-cells provided optimal results, i.e., these cells are extremely easy to use, and they possess higher phase I and II drug-metabolizing enzyme activities than the other cells tested. Although PXB-cells are contaminated with mouse-derived cells up to a concentration of several percent, this cell system appears to be promising for the prediction of in vivo human metabolism of new drugs of abuse.
Collapse
Affiliation(s)
- Tatsuyuki Kanamori
- First Chemistry Section, Third Department of Forensic Science, National Research Institute of Police Science
| |
Collapse
|
98
|
Joossens E, Macko P, Palosaari T, Gerloff K, Ojea-Jiménez I, Gilliland D, Novak J, Fortaner Torrent S, Gineste JM, Römer I, Briffa SM, Valsami-Jones E, Lynch I, Whelan M. A high throughput imaging database of toxicological effects of nanomaterials tested on HepaRG cells. Sci Data 2019; 6:46. [PMID: 31048742 PMCID: PMC6497662 DOI: 10.1038/s41597-019-0053-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/27/2019] [Indexed: 01/22/2023] Open
Abstract
The large amount of existing nanomaterials demands rapid and reliable methods for testing their potential toxicological effect on human health, preferably by means of relevant in vitro techniques in order to reduce testing on animals. Combining high throughput workflows with automated high content imaging techniques allows deriving much more information from cell-based assays than the typical readouts (i.e. one measurement per well) with optical plate-readers. We present here a dataset including data based on a maximum of 14 different read outs (including viable cell count, cell membrane permeability, apoptotic cell death, mitochondrial membrane potential and steatosis) of the human hepatoma HepaRG cell line treated with a large set of nanomaterials, coatings and supernatants at different concentrations. The database, given its size, can be utilized in the development of in silico hazard assessment and prediction tools or can be combined with toxicity results from other in vitro test systems.
Collapse
Affiliation(s)
| | - Peter Macko
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Taina Palosaari
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Kirsten Gerloff
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | | | - Jaroslav Novak
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | | | - Isabella Römer
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
- Human & Environmental Health & Safety Group, Materials Safety Unit, LEITAT, C/Palllars 179-185, 08005, Barcelona, Spain
| | - Sophie Marie Briffa
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Eugenia Valsami-Jones
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Iseult Lynch
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Maurice Whelan
- European Commission, Joint Research Centre (JRC), Ispra, Italy.
| |
Collapse
|
99
|
Won KJ, Park JS, Jeong H. Repression of hepatocyte nuclear factor 4 alpha by AP-1 underlies dyslipidemia associated with retinoic acid. J Lipid Res 2019; 60:794-804. [PMID: 30709899 PMCID: PMC6446710 DOI: 10.1194/jlr.m088880] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/28/2019] [Indexed: 11/20/2022] Open
Abstract
All-trans retinoic acid (atRA) is used to treat certain cancers and dermatologic diseases. A common adverse effect of atRA is hypercholesterolemia; cytochrome P450 (CYP) 7A repression is suggested as a driver. However, the underlying molecular mechanisms remain unclear. We investigated CYP7A1 expression in the presence of atRA in human hepatocytes and hepatic cell lines. In HepaRG cells, atRA increased cholesterol levels dose-dependently alongside dramatic decreases in CYP7A1 expression. Lentiviral-mediated CYP7A1 overexpression reversed atRA-induced cholesterol accumulation, suggesting that CYP7A1 repression mediated cholesterol accumulation. In CYP7A1 promoter reporter assays and gene-knockdown studies, altered binding of hepatocyte nuclear factor 4 α (HNF4α) to the proximal promoter was essential for atRA-mediated CYP7A1 repression. Pharmacologic inhibition of c-Jun N-terminal kinase (JNK) and ERK pathways attenuated atRA-mediated CYP7A1 repression and cholesterol accumulation. Overexpression of AP-1 (c-Jun/c-Fos), a downstream target of JNK and ERK, repressed CYP7A1 expression. In DNA pull-down and chromatin immunoprecipitation assays, AP-1 exhibited sequence-specific binding to the proximal CYP7A1 promoter region overlapping the HNF4α binding site, and atRA increased AP-1 but decreased HNF4α recruitment to the promoter. Collectively, these results indicate that atRA activates JNK and ERK pathways and the downstream target AP-1 represses HNF4α transactivation of the CYP7A1 promoter, potentially responsible for hypercholesterolemia.
Collapse
Affiliation(s)
- Kyoung-Jae Won
- Departments of Pharmacy Practice College of Pharmacy, University of Illinois at Chicago, Chicago, IL
| | - Joo-Seop Park
- Divisions of Pediatric Urology Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH; Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Hyunyoung Jeong
- Departments of Pharmacy Practice College of Pharmacy, University of Illinois at Chicago, Chicago, IL; Biopharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL.
| |
Collapse
|
100
|
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: 53] [Impact Index Per Article: 8.8] [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
|