1
|
Drees A, Nassiri V, Tabernilla A, Serroyen J, Gustin E, Dos Santos Rodrigues B, Moss DM, De Smedt A, Vinken M, Van Goethem F, Sanz-Serrano J. Optimization of the drug-induced cholestasis index based on advanced modeling for predicting liver toxicity. Toxicology 2025; 514:154119. [PMID: 40107378 DOI: 10.1016/j.tox.2025.154119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/11/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Cholestatic drug-induced liver injury (cDILI) is a frequent reason for drug failure and withdrawal during premarketing and postmarketing stages of drug development. Strategies for reliable detection of cDILI in early drug development are therefore urgently needed. The drug-induced cholestasis index (DICI) concept was previously introduced as a tool for assessing the cholestatic potential of drug candidates. DICI is calculated as the ratio between the viability values obtained in drug-treated liver cells in the presence and absence of bile acids. The present in vitro study was set up to investigate the applicability of DICI in a novel high-throughput and large sample setting. Furthermore, the improvement of the predictivity of the DICI by introduction of advanced modeling was explored. Fifty-eight well-documented drugs were selected and categorized as drugs inducing cDILI, non-cholestatic DILI (ncDILI), and not inducing DILI (non-DILI). Cultures of human hepatoma HepaRG cells in 3D spheroid configuration were exposed to 9 half-log concentrations of each drug for 1, 3 and 7 days in the absence or presence of a concentrated mixture of human bile acids. The highest concentration of each drug was based on solubility and the maximum concentrations in human plasma (total Cmax). DICI values were computed for all drugs and time points. In addition, the area under the curve ratio and the occurrence of a trend in the cytotoxicity profiles were included as modeling descriptors. As such, 3 time-related scenarios were considered upon modeling, while categories were modeled on a nominal or an ordinal scale. Applying DICI with a cut-off value of 0.8 resulted in a high sensitivity for the cDILI class, but in turn, a low sensitivity for the non- DILI class. From the 28 predictive models generated, the best performing models integrated all descriptors and the ordinal scale for either the 7-day time point from a 3-time-point model or the 3-day time point. While these models were unable to accurately identify ncDILI drugs, the 7-day time point identified 84 % of the cDILI drugs and the 3-day time point correctly identified 94 % of non-DILI drugs. Based on the obtained results, it can be concluded that the reported DICI modeling provides an optimized approach that could be applied in an integrated DILI testing strategy.
Collapse
Affiliation(s)
- Annika Drees
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Belgium
| | | | - Andrés Tabernilla
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Belgium
| | - Jan Serroyen
- Janssen R&D, Statistics & Decision Sciences, Belgium
| | | | | | | | - Ann De Smedt
- Janssen R&D, Preclinical Sciences and Translational Safety, Belgium
| | - Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Belgium
| | - Freddy Van Goethem
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Belgium; Janssen R&D, Preclinical Sciences and Translational Safety, Belgium
| | - Julen Sanz-Serrano
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Belgium.
| |
Collapse
|
2
|
Chowdhury RR, Rose S, Ezan F, Sovadinová I, Babica P, Langouët S. Hepatotoxicity of cyanotoxin microcystin-LR in human: Insights into mechanisms of action in the 3D culture model Hepoid-HepaRG. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 342:123047. [PMID: 38036087 DOI: 10.1016/j.envpol.2023.123047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/03/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
Microcystin-LR (MC-LR) is a potent hepatotoxin produced by harmful cyanobacterial blooms (CyanoHABs). MC-LR targets highly differentiated hepatocytes expressing organic anion transporting polypeptides OATP1B1 and OATP1B3 that are responsible for hepatocellular uptake of the toxin. The present study utilized an advanced 3D in vitro human liver model Hepoid-HepaRG based on the cultivation of collagen-matrix embedded multicellular spheroids composed of highly differentiated and polarized hepatocyte-like cells. 14-d-old Hepoid-HepaRG cultures showed increased expression of OATP1B1/1B3 and sensitivity to MC-LR cytotoxicity at concentrations >10 nM (48 h exposure, EC20 = 26 nM). MC-LR induced neither caspase 3/7 activity nor expression of the endoplasmic reticulum stress marker gene BiP/GRP78, but increased release of pro-inflammatory cytokine IL-8, indicating a necrotic type of cell death. Subcytotoxic (10 nM) and cytotoxic (≥100 nM) MC-LR concentrations disrupted hepatocyte functions, such as xenobiotic metabolism phase-I enzyme activities (cytochrome P450 1A/1B) and albumin secretion, along with reduced expression of CYP1A2 and ALB genes. MC-LR also decreased expression of HNF4A gene, a critical regulator of hepatocyte differentiation and function. Genes encoding hepatobiliary membrane transporters (OATP1B1, BSEP, NTCP), hepatocyte gap junctional gene connexin 32 and the epithelial cell marker E-cadherin were also downregulated. Simultaneous upregulation of connexin 43 gene, primarily expressed by liver progenitor and non-parenchymal cells, indicated a disruption of tissue homeostasis. This was associated with a shift in the expression ratio of E-cadherin to N-cadherin towards the mesenchymal cell marker, a process linked to epithelial-mesenchymal transition (EMT) and hepatocarcinogenesis. The effects observed in the human liver cell in vitro model revealed mechanisms that can potentially contribute to the MC-LR-induced promotion and progression of hepatocellular carcinoma (HCC). Hepoid-HepaRG cultures provide a robust, accessible and versatile in vitro model, capable of sensitively detecting hepatotoxic effects at toxicologically relevant concentrations, allowing for assessing hepatotoxicity mechanisms, human health hazards and impacts of environmental hepatotoxins, such as MC-LR.
Collapse
Affiliation(s)
- Riju R Chowdhury
- Masaryk University, Faculty of Science, RECETOX, Kotlářská 2, 61137, Brno, Czech Republic
| | - Sophie Rose
- University of Rennes, Inserm, EHESP, Irset (Institut de Recherche en santé, environnement et travail), UMR_S 1085, 35000, Rennes, France
| | - Frédéric Ezan
- University of Rennes, Inserm, EHESP, Irset (Institut de Recherche en santé, environnement et travail), UMR_S 1085, 35000, Rennes, France
| | - Iva Sovadinová
- Masaryk University, Faculty of Science, RECETOX, Kotlářská 2, 61137, Brno, Czech Republic
| | - Pavel Babica
- Masaryk University, Faculty of Science, RECETOX, Kotlářská 2, 61137, Brno, Czech Republic
| | - Sophie Langouët
- University of Rennes, Inserm, EHESP, Irset (Institut de Recherche en santé, environnement et travail), UMR_S 1085, 35000, Rennes, France.
| |
Collapse
|
3
|
Lynch KD, Iverson DT, Bachhav NK, Call MR, Yue GE, Prasad B, Clarke JD. Involvement of the p38/MK2 Pathway in MCLR Hepatotoxicity Revealed through MAPK Pharmacological Inhibition and Phosphoproteomics in HepaRG Cells. Int J Mol Sci 2023; 24:11168. [PMID: 37446360 PMCID: PMC10342686 DOI: 10.3390/ijms241311168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Microcystin-leucine arginine (MCLR) is one of the most common and toxic microcystin variants, a class of cyanotoxins produced by cyanobacteria. A major molecular mechanism for MCLR-elicited liver toxicity involves the dysregulation of protein phosphorylation through protein phosphatase (PP) inhibition and mitogen-activated protein kinase (MAPK) modulation. In this study, specific pharmacological MAPK inhibitors were used in HepaRG cells to examine the pathways associated with MCLR cytotoxicity. SB203580 (SB), a p38 inhibitor, rescued HepaRG cell viability, whereas treatment with SP600125 (JNK inhibitor), MK2206 (AKT inhibitor), or N-acetylcysteine (reactive oxygen species scavenger) did not. Phosphoproteomic analysis revealed that phosphosites-which were altered by the addition of SB compared to MCLR treatment alone-included proteins involved in RNA processing, cytoskeletal stability, DNA damage response, protein degradation, and cell death. A closer analysis of specific proteins in some of these pathways indicated that SB reversed the MCLR-mediated phosphorylation of the necroptosis-associated proteins, the mixed lineage kinase domain-like protein (MLKL), receptor-interacting serine/threonine kinase 1 (RIP1), DNA damage response proteins, ataxia telangiectasia and Rad3-related kinase (ATR), and checkpoint kinase 1 (CHK1). Overall, these data implicate p38/MK2, DNA damage, and necroptosis in MCLR-mediated hepatotoxicity, and suggest these pathways may be targets for prevention prior to, or treatment after, MCLR toxicity.
Collapse
Affiliation(s)
- Katherine D. Lynch
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA; (K.D.L.); (B.P.)
| | | | | | | | | | | | - John D. Clarke
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA; (K.D.L.); (B.P.)
| |
Collapse
|
4
|
Fragki S, Louisse J, Bokkers B, Luijten M, Peijnenburg A, Rijkers D, Piersma AH, Zeilmaker MJ. New approach methodologies: A quantitative in vitro to in vivo extrapolation case study with PFASs. Food Chem Toxicol 2023; 172:113559. [PMID: 36535450 PMCID: PMC9890272 DOI: 10.1016/j.fct.2022.113559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/23/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
PER: and polyfluoroalkyl substances (PFASs) have been associated with increased blood lipids in humans. Perfluorooctanoic acid (PFOA) has been also linked with elevated alanine transferase (ALT) serum levels in humans, and in rodents the liver is a main target organ for many PFASs. With the focus on New Approach Methodologies, the chronic oral equivalent effect doses were calculated for PFOA, PFNA (perfluorononanoic acid), PFHxS (perfluorohexanesulfonic acid) and PFOS (perfluorooctane sulfonic acid) based on in vitro effects measured in the HepaRG cell line. Selected in vitro readouts were considered biomarkers for lipid disturbances and hepatotoxicity. Concentration-response data obtained from HepaRG cells on triglyceride (TG) accumulation and expression changes of 12 selected genes (some involved in cholesterol homeostasis) were converted into corresponding human dose-response data, using physiologically based kinetic (PBK) model-facilitated reverse dosimetry. Next to this, the biokinetics of the chemicals were studied in the cell system. The current European dietary PFASs exposure overlaps with the calculated oral equivalent effect doses, indicating that the latter may lead to interference with hepatic gene expression and lipid metabolism. These findings illustrate an in vitro-in silico methodology, which can be applied for more PFASs, to select those that should be prioritized for further hazard characterization.
Collapse
Affiliation(s)
- Styliani Fragki
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands.
| | - Jochem Louisse
- Wageningen Food Safety Research (WFSR), Wageningen, the Netherlands
| | - Bas Bokkers
- Centre for Safety of Substances and Products, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Mirjam Luijten
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Ad Peijnenburg
- Wageningen Food Safety Research (WFSR), Wageningen, the Netherlands
| | - Deborah Rijkers
- Wageningen Food Safety Research (WFSR), Wageningen, the Netherlands
| | - Aldert H Piersma
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences, Utrecht University, P.O. Box 80178, 3508 TD, Utrecht, the Netherlands
| | - Marco J Zeilmaker
- Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| |
Collapse
|
5
|
The next frontier in ADME science: Predicting transporter-based drug disposition, tissue concentrations and drug-drug interactions in humans. Pharmacol Ther 2022; 238:108271. [DOI: 10.1016/j.pharmthera.2022.108271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/05/2022] [Accepted: 08/17/2022] [Indexed: 12/25/2022]
|
6
|
Tenge VR, Murakami K, Salmen W, Lin SC, Crawford SE, Neill FH, Prasad BVV, Atmar RL, Estes MK. Bile Goes Viral. Viruses 2021; 13:998. [PMID: 34071855 PMCID: PMC8227374 DOI: 10.3390/v13060998] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/12/2022] Open
Abstract
Laboratory cultivation of viruses is critical for determining requirements for viral replication, developing detection methods, identifying drug targets, and developing antivirals. Several viruses have a history of recalcitrance towards robust replication in laboratory cell lines, including human noroviruses and hepatitis B and C viruses. These viruses have tropism for tissue components of the enterohepatic circulation system: the intestine and liver, respectively. The purpose of this review is to discuss how key enterohepatic signaling molecules, bile acids (BAs), and BA receptors are involved in the replication of these viruses and how manipulation of these factors was useful in the development and/or optimization of culture systems for these viruses. BAs have replication-promoting activities through several key mechanisms: (1) affecting cellular uptake, membrane lipid composition, and endocytic acidification; (2) directly interacting with viral capsids to influence binding to cells; and (3) modulating the innate immune response. Additionally, expression of the Na+-taurocholate cotransporting polypeptide BA receptor in continuous liver cell lines is critical for hepatitis B virus entry and robust replication in laboratory culture. Viruses are capable of hijacking normal cellular functions, and understanding the role of BAs and BA receptors, components of the enterohepatic system, is valuable for expanding our knowledge on the mechanisms of norovirus and hepatitis B and C virus replication.
Collapse
Affiliation(s)
- Victoria R. Tenge
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (V.R.T.); (W.S.); (S.-C.L.); (S.E.C.); (F.H.N.); (B.V.V.P.); (R.L.A.)
| | - Kosuke Murakami
- Department of Virology II, National Institute of Infectious Diseases, Musashi-Murayama, Tokyo 208-0011, Japan;
| | - Wilhelm Salmen
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (V.R.T.); (W.S.); (S.-C.L.); (S.E.C.); (F.H.N.); (B.V.V.P.); (R.L.A.)
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shih-Ching Lin
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (V.R.T.); (W.S.); (S.-C.L.); (S.E.C.); (F.H.N.); (B.V.V.P.); (R.L.A.)
| | - Sue E. Crawford
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (V.R.T.); (W.S.); (S.-C.L.); (S.E.C.); (F.H.N.); (B.V.V.P.); (R.L.A.)
| | - Frederick H. Neill
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (V.R.T.); (W.S.); (S.-C.L.); (S.E.C.); (F.H.N.); (B.V.V.P.); (R.L.A.)
| | - B. V. Venkataram Prasad
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (V.R.T.); (W.S.); (S.-C.L.); (S.E.C.); (F.H.N.); (B.V.V.P.); (R.L.A.)
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Robert L. Atmar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (V.R.T.); (W.S.); (S.-C.L.); (S.E.C.); (F.H.N.); (B.V.V.P.); (R.L.A.)
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (V.R.T.); (W.S.); (S.-C.L.); (S.E.C.); (F.H.N.); (B.V.V.P.); (R.L.A.)
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
7
|
In vivo and in vitro α-amanitin metabolism studies using molecular networking. Toxicol Lett 2021; 346:1-6. [PMID: 33872745 DOI: 10.1016/j.toxlet.2021.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/17/2021] [Accepted: 04/13/2021] [Indexed: 01/09/2023]
Abstract
Amanitin poisonings are among the most life-threatening mushroom poisonings, and are mainly caused by the genus Amanita. Hepatotoxicity is the hallmark of amanitins, powerful toxins contained in these mushrooms, and can require liver transplant. Among amatoxins, α-amanitin is the most studied. However, the hypothesis of a possible metabolism of amanitins is still controversial in this pathophysiology. Therefore, there is a need of clarification using cutting-edge tools allowing metabolism study. Molecular network has emerged as powerful tool allowing metabolism study through organization and representation of untargeted tandem mass spectrometry (MS/MS) data in a graphical form. The aim of this study is to investigate amanitin metabolism using molecular networking. In vivo (four positive amanitin urine samples) and in vitro (differentiated HepaRG cells supernatant incubated with α-amanitin 2 μM for 24 h) samples were extracted and analyzed by LC-HRMS/MS using a Q Exactive™ Orbitrap mass spectrometer. Using molecular networking on both in vitro and in vivo, we have demonstrated that α-amanitin does not undergo metabolism in human. Thus, we provide solid evidence that a possible production of amanitin metabolites cannot be involved in its toxicity pathways. These findings can help to settle the debate on amanitin metabolism and toxicity.
Collapse
|
8
|
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]
|
9
|
Tajiri A, Hirota T, Kawano S, Yonamine A, Ieiri I. Regulation of Organic Anion Transporting Polypeptide 2B1 Expression by MicroRNA in the Human Liver. Mol Pharm 2020; 17:2821-2830. [PMID: 32602343 DOI: 10.1021/acs.molpharmaceut.0c00193] [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] [Indexed: 01/23/2023]
Abstract
Organic anion transporting polypeptide 2B1 (OATP2B1, SLCO2B1) is an uptake transporter expressed in several tissues, including the liver, intestine, brain, kidney, and skeletal muscle. Hepatocyte nuclear factor 4 alpha (HNF4α) is known as an important transcriptional factor of OATP2B1 in the liver. It has been reported that there are large interindividual differences in OATP2B1 mRNA and protein expressions in human livers. The mechanism causing the interindividual differences in OATP2B1 expression is still unclear. MicroRNAs (miRNAs) control gene expression by leading translational repression and/or degradation of the target mRNA. There is no significant correlation between OATP2B1 mRNA and protein expression, suggesting that post-transcriptional regulating mechanisms, such as miRNAs, play an important role in the interindividual differences in OATP2B1 expression. In this study, we hypothesized that certain miRNAs cause the interindividual differences in OATP2B1 expression in the human liver. In silico analysis showed that miR-24 was a candidate miRNA regulating OATP2B1 expression. It has been reported that miR-24 degrades HNF4α mRNA expression. We revealed that the miR-24 expression level was negatively correlated with OATP2B1 mRNA, protein, and HNF4α mRNA expression levels in human livers. Transfection by the miR-24 precursor decreased the luciferase activity in the transfected cells with the vector containing the OATP2B1 3' untranslated region (3'UTR) or SLCO2B1 promoter region. In HepaRG cells, miR-24 decreased the OATP2B1 and HNF4α expression levels. These results suggest that miR-24 represses not only the translation of OATP2B1 but also the transcription of OATP2B1 by HNF4α mRNA degradation.
Collapse
Affiliation(s)
- Ayaka Tajiri
- Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takeshi Hirota
- Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Sasagu Kawano
- Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Akira Yonamine
- Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ichiro Ieiri
- Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
10
|
Nozaki Y, Izumi S. Recent advances in preclinical in vitro approaches towards quantitative prediction of hepatic clearance and drug-drug interactions involving organic anion transporting polypeptide (OATP) 1B transporters. Drug Metab Pharmacokinet 2020; 35:56-70. [DOI: 10.1016/j.dmpk.2019.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 09/29/2019] [Accepted: 11/02/2019] [Indexed: 12/26/2022]
|
11
|
Dietrich J, Schindler M, Lampen A, Braeuning A, Hessel-Pras S. Comparison of long-term versus short-term effects of okadaic acid on the apoptotic status of human HepaRG cells. Chem Biol Interact 2020; 317:108937. [PMID: 31926150 DOI: 10.1016/j.cbi.2020.108937] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/14/2019] [Accepted: 01/06/2020] [Indexed: 01/08/2023]
Abstract
The biotoxin okadaic acid (OA) is a lipophilic secondary metabolite of marine microalgae. Therefore, OA accumulates in the fatty tissue of various shellfish and may thus enter the food chain. The ingestion of OA via contaminated marine species can lead to the diarrhetic shellfish poisoning syndrome characterized by the occurrence of a series of acute gastrointestinal symptoms in humans. In addition, genotoxicity and tumor-promoting properties of OA might constitute a long-term threat to human health. In order to deepen our understanding of the molecular effects of OA, we compared long-term (14 d) and short-term (24 h and 48 h) apoptotic effects of the compound on human HepaRG hepatocarcinoma cells. Cells were treated either with single doses for 24 and 48 h, respectively, or seven times over a period of 14 d, so that the cumulated quantities of OA in the long-term approach were equal to the single doses upon short-term treatment. Both short-term treatment scenarios led to the induction of apoptosis. Specific caspase activation assays and transcriptional analysis of mRNAs encoding proteins involved in the regulation of apoptosis suggest that OA-induced apoptosis occurs presumably by activation of the intrinsic apoptotic pathway. In contrast, effects were much less pronounced in case of long-term treatment. This is possibly linked to cellular protective mechanisms against low amounts of toxins, e.g. transporter-mediated efflux. In conclusion, our results show a clear concentration- and time-dependency of OA-mediated apoptotic effects in HepaRG cells and contribute to the elucidation of molecular effects of OA.
Collapse
Affiliation(s)
- Jessica Dietrich
- German Federal Institute for Risk Assessment, Department of Food Safety, 10589, Berlin, Germany
| | - Magdalena Schindler
- German Federal Institute for Risk Assessment, Department of Food Safety, 10589, Berlin, Germany
| | - Alfonso Lampen
- German Federal Institute for Risk Assessment, Department of Food Safety, 10589, Berlin, Germany
| | - Albert Braeuning
- German Federal Institute for Risk Assessment, Department of Food Safety, 10589, Berlin, Germany
| | - Stefanie Hessel-Pras
- German Federal Institute for Risk Assessment, Department of Food Safety, 10589, Berlin, Germany.
| |
Collapse
|
12
|
Cheng X, Wang Y, Tian J, Zhou L, Chen X, Guo H, Zeng J, Shen N, Li J, Ke J, Zhu Y, Gong J, Chang J, Liu L, Zhong R. SLC10A1 S267F variant influences susceptibility to HBV infection and reduces cholesterol level by impairing bile acid uptake. J Viral Hepat 2019; 26:1178-1185. [PMID: 31177598 DOI: 10.1111/jvh.13157] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 04/07/2019] [Accepted: 05/15/2019] [Indexed: 01/05/2023]
Abstract
The SLC10A1 Ser267Phe (S267F) variant has been reported to severely inhibit hepatitis B virus (HBV) infection and taurocholate transport activity. This study aimed to clarify the effects of this variant on HBV infection and bile acid metabolism. SLC10A1 S267F was genotyped in 2907 HBV-exposed subjects (including HBV persistent carriers and spontaneously recovered subjects) and 1364 unexposed subjects (HBV marker-negative subjects), followed by replication I, comprising 914 exposed subjects and 1123 unexposed subjects, and replication II, comprising 355 children born to HBsAg-positive mothers (226 HBV-infected children and 129 controls). Intriguingly, SLC10A1 AA was observed only in the unexposed group, but not in the exposed group. The SLC10A1 A allele consistently decreased HBV infection risk compared with the G allele (OR = 0.76, 95% CI: 0.64-0.90 in combined samples). In addition, children with the SLC10A1 GA genotype had a reduced risk of perinatal transmission (OR = 0.31, 95% CI: 0.14-0.71). Moreover, unexposed subjects with the SLC10A1 AA genotype exhibited decreased serum total cholesterol and low-density lipoprotein cholesterol compared to those with the GG or GA genotypes (P = 2.975 × 10-4 and 0.004, respectively). The study highlighted the role of the SLC10A1 S267F variant in the loss of the ability to support HBV infection and taurocholate transport activity. Subjects with the AA genotype may escape from HBV infection and present decreased cholesterol levels as a consequence of impaired bile acid uptake.
Collapse
Affiliation(s)
- Xiang Cheng
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Wang
- Department of Virology, Wuhan Centers for Disease Prevention and Control, Wuhan, China
| | - Jianbo Tian
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Zhou
- Department of Epidemiology, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, China
| | - Xueqin Chen
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Guo
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junchao Zeng
- Medical Examination Center, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Shen
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaoyuan Li
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juntao Ke
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Zhu
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Gong
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiang Chang
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Rong Zhong
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Jang M, Kleber A, Ruckelshausen T, Betzholz R, Manz A. Differentiation of the human liver progenitor cell line (HepaRG) on a microfluidic-based biochip. J Tissue Eng Regen Med 2019; 13:482-494. [PMID: 30746894 DOI: 10.1002/term.2802] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/26/2018] [Accepted: 01/09/2019] [Indexed: 12/13/2022]
Abstract
HepaRG is a bipotent stem cell line that can be differentiated towards hepatocyte-like and biliary-like cells. The entire cultivation process requires 1 month and relies on the addition of 2% dimethyl sulfoxide (DMSO) to the culture. Our motivation in this research is to differentiate HepaRG cells (progenitor cells and undifferentiated cells) towards hepatocyte-like cells by minimizing the cultivation time and without using DMSO treatment by instead using a microfluidic device combined with the following strategies: (a) comparison of extracellular matrices (matrigel and collagen I), (b) types of flow (one or both sides), and (c) effects of DMSO. Our results demonstrate that matrigel promotes the differentiation of progenitor cells towards hepatocytes and biliary-like cells. Moreover, the frequent formation of HepaRG cell clusters was observed by a supply of both sides of flow, and the cell viability and liver specific functions were influenced by DMSO. Finally, differentiated HepaRG progenitor cells cultured in a microfluidic device for 14 days without DMSO treatment yielded 70% of hepatocyte-like cells with a highly polarized organization that reacted to stimulation with IL-6 to produce C-reactive protein (CRP). This culture model has high potential for investigating cell differentiation and liver pathophysiology research.
Collapse
Affiliation(s)
- Mi Jang
- Department of system engineering, Saarland University, Saarbrücken, Germany.,Microfluidics group, KIST Europe, Saarbrücken, Germany.,Department of Neuroscience, Korea University College of Medicine, Seoul, Korea
| | - Astrid Kleber
- Rhineland Palantinate Centre of Excellence for climate Change Impacts, Trippstadt, Germany
| | - Thomas Ruckelshausen
- Dynamic Biomaterial group, INM - Leibniz-Institut für Neue Materialien GmbH, Saarbrücken, Germany.,Service and Support group, PicoQuant, Rudower Chaussee 29, Berlin, Germany
| | - Ralf Betzholz
- School of Physics, Huazhong University of Science and Technology, Wuhan, China
| | - Andreas Manz
- Department of system engineering, Saarland University, Saarbrücken, Germany.,Microfluidics group, KIST Europe, Saarbrücken, Germany
| |
Collapse
|
14
|
Petrov PD, Fernández-Murga ML, López-Riera M, Goméz-Lechón MJ, Castell JV, Jover R. Predicting drug-induced cholestasis: preclinical models. Expert Opin Drug Metab Toxicol 2018; 14:721-738. [PMID: 29888962 DOI: 10.1080/17425255.2018.1487399] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION In almost 50% of patients with drug-induced liver injury (DILI), the bile flow from the liver to the duodenum is impaired, a condition known as cholestasis. However, this toxic response only appears in a small percentage of the treated patients (idiosyncrasy). Prediction of drug-induced cholestasis (DIC) is challenging and emerges as a safety issue that requires attention by professionals in clinical practice, regulatory authorities, pharmaceutical companies, and research institutions. Area covered: The current synopsis focuses on the state-of-the-art in preclinical models for cholestatic DILI prediction. These models differ in their goal, complexity, availability, and applicability, and can widely be classified in experimental animals and in vitro models. Expert opinion: Drugs are a growing cause of cholestasis, but the progress made in explaining mechanisms and differences in susceptibility is not growing at the same rate. We need reliable models able to recapitulate the features of DIC, particularly its idiosyncrasy. The homogeneity and the species-specific differences move animal models away from a fair predictability. However, in vitro human models are improving and getting closer to the real hepatocyte phenotype, and they will likely be the choice in the near future. Progress in this area will not only need reliable predictive models but also mechanistic insights.
Collapse
Affiliation(s)
- Petar D Petrov
- a Instituto de Investigación Sanitaria La Fe (IIS La Fe) , Unidad de Hepatología Experimental , Valencia , Spain.,b Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD) , Madrid , Spain
| | - M Leonor Fernández-Murga
- a Instituto de Investigación Sanitaria La Fe (IIS La Fe) , Unidad de Hepatología Experimental , Valencia , Spain
| | - Mireia López-Riera
- a Instituto de Investigación Sanitaria La Fe (IIS La Fe) , Unidad de Hepatología Experimental , Valencia , Spain
| | - M José Goméz-Lechón
- a Instituto de Investigación Sanitaria La Fe (IIS La Fe) , Unidad de Hepatología Experimental , Valencia , Spain.,b Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD) , Madrid , Spain
| | - Jose V Castell
- a Instituto de Investigación Sanitaria La Fe (IIS La Fe) , Unidad de Hepatología Experimental , Valencia , Spain.,b Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD) , Madrid , Spain.,c Departamento de Bioquímica y Biología Molecular, Facultad de Medicina , Universidad de Valencia , Valencia , Spain
| | - Ramiro Jover
- a Instituto de Investigación Sanitaria La Fe (IIS La Fe) , Unidad de Hepatología Experimental , Valencia , Spain.,b Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD) , Madrid , Spain.,c Departamento de Bioquímica y Biología Molecular, Facultad de Medicina , Universidad de Valencia , Valencia , Spain
| |
Collapse
|
15
|
Zeng Y, Li Z, Shang J, Kang Y. Efficient delivery of HBV NLS siRNAs into HepG2.2.15 cells for HBV inhibition through novel recombinant preS1‑tP proteins. Int J Mol Med 2018; 42:1181-1189. [PMID: 29786106 DOI: 10.3892/ijmm.2018.3681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/08/2018] [Indexed: 11/05/2022] Open
Abstract
Hepatitis B virus (HBV) infection and related liver complications remain severe public health problems worldwide. Previous investigations have shown that small interfering (si)RNAs can offer an effective strategy for the treatment of chronic hepatitis B. The present study aimed to develop a novel siRNA‑delivering system of therapeutic HBV nuclear localization sequence (NLS) siRNAs using the recombinant preS1‑truncated protamine (tP) proteins. The preS1 region of the LHB was used in place of scFv to construct the recombinant preS1‑tP proteins, which were applied to deliver siRNAs targeting the HBV NLS to inhibit HBV replication and infection in HepG2.2.15 cells overexpressing sodium taurocholate cotransporting polypeptide (NTCP). The results revealed that HepG2.2.15 cells with stable NTCP expression (HepG2.2.15‑NTCP cells) transfected with the recombinant lentivirus showed increased expression of NTCP genes. The HBV NLS siRNAs significantly suppressed HBV mRNA content and levels of HBsAg and HBeAg in the HepG2.2.15‑NTCP cells. Recombinant preS1‑tP proteins tagged with His and glutathione S‑transferase were found to enter into HepG2.2.15‑NTCP cells and bind with DNA. The HBV NLS siRNAs were delivered into HepG2.2.15‑NTCP cells by recombinant preS1‑tP proteins, which resulted in decreased expression of HBV mRNA, HBsAg and HBeAg, HBV DNA and covalently closed circular DNA in the HepG2.2.15‑NTCP cells. Therefore, the recombinant preS1‑tP proteins successfully delivered NLS siRNAs into HepG2.2.15 cells and repressed HBV infection and replication.
Collapse
Affiliation(s)
- Yanli Zeng
- Department of Infectious Diseases, Zhengzhou University People's Hospital, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Zixi Li
- Tongji Medical College of Huazhong University of Science and Technology Affiliated Hospital of Traditional Chinese and Western Medicine, Wuhan, Hubei 430030, P.R. China
| | - Jia Shang
- Department of Infectious Diseases, Zhengzhou University People's Hospital, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Yi Kang
- Department of Infectious Diseases, Zhengzhou University People's Hospital, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
16
|
Schaefer M, Morinaga G, Matsui A, Schänzle G, Bischoff D, Süssmuth RD. Quantitative Expression of Hepatobiliary Transporters and Functional Uptake of Substrates in Hepatic Two-Dimensional Sandwich Cultures: A Comparative Evaluation of Upcyte and Primary Human Hepatocytes. Drug Metab Dispos 2018; 46:166-177. [PMID: 29212823 DOI: 10.1124/dmd.117.078238] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/27/2017] [Indexed: 02/13/2025] Open
Abstract
Deficient functional expression of drug transporters incapacitates most hepatic cell lines as a reliable tool for evaluating transporter-mediated drug-drug interactions. Recently, genetically modified cells (referred to as upcyte hepatocytes) have emerged as an expandable, noncancerous source of human hepatic cells. Herein, we quantified mRNA and protein levels of key hepatobiliary transporters and we assessed associated uptake activity in short- and long-term cultures of upcyte human hepatocytes (UHH) in comparison to cryopreserved primary human hepatocytes (cPHH). Expression of canalicular efflux pumps, such as MRD1/ABCB1, MATE1/SLC47A1, and MRP2/ABCC2, was relatively well preserved in UHH. By contrast, long-term cultivation of UHH in a two-dimensional sandwich configuration [sandwich-cultured upcyte human hepatocytes (SCUHH)] was required to upregulate organic anion-transporting polypeptide OATP1B1/SLCO1B1, OATP2B1/SLCO2B1, NTCP/SLC10A1, and OCT1/SLC22A1 mRNA expression, which correlated well with respective protein abundances. However, mRNA and protein levels of sinusoidal solute carrier transporters, except for NTCP and OATP2B1, remained low in SCUHH compared to sandwich-cultured cPHH. OCT1- and NTCP-mediated uptake of N-methyl-4-phenylpyridinium acetate and taurocholate was demonstrated in both hepatic models, whereas active uptake of OATP1B1/1B3-selective marker substrates, paralleled by markedly reduced SLCO1B1/1B3 expression, were not detectable in SCUHH. Uptake studies under Na+-depletion and excess of taurocholate confirmed the presence of functional NTCP protein and indicated that NTCP, apart from OATP2B1, contributed substantially to the overall hepatic uptake of rosuvastatin in SCUHH. In conclusion, our data suggest that SCUHH, despite their limitation for evaluating OATP1B1/1B3-mediated transport processes, retain NTCP, OATP2B1, and OCT1 transport activities and thus may be considered as a tool for elucidating compensatory uptake pathways for OATP1B1/1B3 substrates.
Collapse
Affiliation(s)
- Michelle Schaefer
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany (M.S., G.S., D.B.); Department of Pharmacokinetics and Nonclinical Safety, Kobe Pharma Research Institute, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan (G.M., A.M.); and Institut für Chemie, Technische Universität Berlin, Berlin, Germany (R.D.S.)
| | - Gaku Morinaga
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany (M.S., G.S., D.B.); Department of Pharmacokinetics and Nonclinical Safety, Kobe Pharma Research Institute, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan (G.M., A.M.); and Institut für Chemie, Technische Universität Berlin, Berlin, Germany (R.D.S.)
| | - Akiko Matsui
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany (M.S., G.S., D.B.); Department of Pharmacokinetics and Nonclinical Safety, Kobe Pharma Research Institute, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan (G.M., A.M.); and Institut für Chemie, Technische Universität Berlin, Berlin, Germany (R.D.S.)
| | - Gerhard Schänzle
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany (M.S., G.S., D.B.); Department of Pharmacokinetics and Nonclinical Safety, Kobe Pharma Research Institute, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan (G.M., A.M.); and Institut für Chemie, Technische Universität Berlin, Berlin, Germany (R.D.S.)
| | - Daniel Bischoff
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany (M.S., G.S., D.B.); Department of Pharmacokinetics and Nonclinical Safety, Kobe Pharma Research Institute, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan (G.M., A.M.); and Institut für Chemie, Technische Universität Berlin, Berlin, Germany (R.D.S.)
| | - Roderich D Süssmuth
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany (M.S., G.S., D.B.); Department of Pharmacokinetics and Nonclinical Safety, Kobe Pharma Research Institute, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan (G.M., A.M.); and Institut für Chemie, Technische Universität Berlin, Berlin, Germany (R.D.S.)
| |
Collapse
|
17
|
Beckwitt CH, Clark AM, Wheeler S, Taylor DL, Stolz DB, Griffith L, Wells A. Liver 'organ on a chip'. Exp Cell Res 2018; 363:15-25. [PMID: 29291400 PMCID: PMC5944300 DOI: 10.1016/j.yexcr.2017.12.023] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 12/21/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022]
Abstract
The liver plays critical roles in both homeostasis and pathology. It is the major site of drug metabolism in the body and, as such, a common target for drug-induced toxicity and is susceptible to a wide range of diseases. In contrast to other solid organs, the liver possesses the unique ability to regenerate. The physiological importance and plasticity of this organ make it a crucial system of study to better understand human physiology, disease, and response to exogenous compounds. These aspects have impelled many to develop liver tissue systems for study in isolation outside the body. Herein, we discuss these biologically engineered organoids and microphysiological systems. These aspects have impelled many to develop liver tissue systems for study in isolation outside the body. Herein, we discuss these biologically engineered organoids and microphysiological systems.
Collapse
Affiliation(s)
- Colin H Beckwitt
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA; Research and Development Service, VA Pittsburgh Health System, Pittsburgh, PA 15240, USA
| | - Amanda M Clark
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sarah Wheeler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - D Lansing Taylor
- Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA; Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Donna B Stolz
- Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Linda Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute of Regenerative Medicine University of Pittsburgh, Pittsburgh, PA 15213, USA; Research and Development Service, VA Pittsburgh Health System, Pittsburgh, PA 15240, USA.
| |
Collapse
|
18
|
Ueyama T, Tsuji S, Sugiyama T, Tada M. Fluorometric evaluation of CYP3A4 expression using improved transgenic HepaRG cells carrying a dual-colour reporter for CYP3A4 and CYP3A7. Sci Rep 2017; 7:2874. [PMID: 28588200 PMCID: PMC5460180 DOI: 10.1038/s41598-017-03146-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/24/2017] [Indexed: 01/29/2023] Open
Abstract
Primary human hepatocytes are necessary to evaluate cytotoxicity, drug metabolism, and drug–drug interactions for candidate compounds in early-phase drug discovery and development. However, these analyses are often hampered by limited resources and functional or genetic variation among lots. HepaRG human hepatocellular carcinoma cells can differentiate into mature hepatocyte-like cells (HepLCs) that possess similar metabolic activity to human hepatocytes. We previously established transgenic HepaRG cells carrying a dual reporter that express red fluorescent protein (RFP) under the transcriptional regulation of CYP3A7 in the hepatoblast-like cell state and enhanced green fluorescent protein (EGFP) under the transcriptional regulation of CYP3A4 following HepLC differentiation. In this study, we successfully isolated a subclone of transgenic CYP3A4G/7R HepaRG cells with an improved HepLC differentiation potency. Midazolam metabolism by CYP3A4 in these HepLCs was comparable to that in wild-type HepLCs. The EGFP fluorescence intensity was greatly induced by rifampicin (RIF) treatment. There was a strong correlation between fluorometric and metabolic analyses. The fold change in EGFP-positive cells was comparable to those in the CYP3A4 mRNA level and luminescence of proluciferin metabolites. RIF treatment and cell proliferation increased the RFP-positive cell number. Thus, CYP3A4G/7R HepLCs provide a real-time, multiwell-based system to co-evaluate CYP3A4 induction and hepatic regeneration.
Collapse
Affiliation(s)
- Takafumi Ueyama
- Tottori Cell Laboratory, Cell Technology, KAC Co., Ltd., Yonago, Japan.,Chromosome Engineering Research Centre, Tottori University, Yonago, Japan
| | - Saori Tsuji
- Tottori Cell Laboratory, Cell Technology, KAC Co., Ltd., Yonago, Japan.,Chromosome Engineering Research Centre, Tottori University, Yonago, Japan
| | - Takemi Sugiyama
- Tottori Cell Laboratory, Cell Technology, KAC Co., Ltd., Yonago, Japan
| | - Masako Tada
- Chromosome Engineering Research Centre, Tottori University, Yonago, Japan. .,Stem Cells & Reprogramming Laboratory, Human Biology, Department of Biology, Faculty of Science, Toho University, Miyama 2-2-1, Funabashi, Chiba, 274-8510, Japan.
| |
Collapse
|
19
|
Evaluation of HepaRG cells for the assessment of indirect drug-induced hepatotoxicity using INH as a model substance. Hum Cell 2017; 30:267-278. [PMID: 28527127 DOI: 10.1007/s13577-017-0175-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 05/04/2017] [Indexed: 01/13/2023]
Abstract
HepaRG cells are widely used as an in vitro model to assess drug-induced hepatotoxicity. However, only few studies exist so far regarding their suitability to detect the effects of drugs requiring a preceding activation via the cytochrome P450 (CYP) system. A prototypic substance is the anti-tuberculosis agent INH, which is metabolized into N-acetylhydrazine, which then triggers hepatotoxicity. Therefore, the aim of the present study was to test if this effect can also be detected in HepaRG cells and if it can be counteracted by the known hepatoprotectant silibinin. For this purpose, differentiated HepaRG cells were treated with increasing concentrations of INH (0.1-100 mM) or 10 mM INH plus escalating concentrations of silibinin (1-100 µM). After 48 h of treatment, cell morphology and parameters indicating cell vitality, oxidative stress, and liver cell function were assessed. High concentrations of INH led to severe histopathological changes, reduced cell vitality and glutathione content, increased LDH and ASAT release into the medium, enhanced lipid peroxidation, and elevated cleaved caspase-3 expression. Additionally, glycogen depletion and reduced biotransformation capacity were seen at high INH concentrations, whereas at low concentrations an induction of biotransformation enzymes was noticed. Silibinin caused clear-cut protective effects, but with few parameters INH toxicity was even aggravated, most probably due to increased metabolization of INH into its toxic metabolite. In conclusion, HepaRG cells are excellently suited to evaluate the effects of substances requiring prior toxification via the CYP system, such as INH. They additionally enable the identification of complex substance interactions.
Collapse
|
20
|
Miyake T, Mizuno T, Mochizuki T, Kimura M, Matsuki S, Irie S, Ieiri I, Maeda K, Kusuhara H. Involvement of Organic Cation Transporters in the Kinetics of Trimethylamine N-oxide. J Pharm Sci 2017; 106:2542-2550. [PMID: 28479364 DOI: 10.1016/j.xphs.2017.04.067] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/21/2017] [Accepted: 04/25/2017] [Indexed: 10/19/2022]
Abstract
Recent studies suggest that trimethylamine N-oxide (TMAO) is associated with the development of chronic kidney disease and heart failure. In this study, we investigated the importance of organic cation transporters (OCTs) in the clearance and tissue distribution of TMAO. The low-affinity and high-capacity transport of TMAO by mouse and human OCT1 and OCT2 was observed. Uptake and efflux of TMAO by the mouse hepatocytes as well as TMAO uptake into mouse kidney slices were significantly decreased by the addition of tetraethylammonium or Oct1/2 double knockout (dKO). Plasma concentrations of endogenous TMAO and TMAO-d9 given by intravenous infusion was 2-fold higher in Oct1/2 dKO than in wild-type mice due to significant decrease in its renal clearance. These results indicate that OCTs have a crucial role in the kinetics of TMAO in mice. In human, however, the OCT2-mediated tubular secretion in the urinary excretion of TMAO was insignificant because the renal clearance of TMAO was similar to that of creatinine in both young and elderly subjects, suggesting the species difference in the urinary excretion mechanisms of TMAO between mouse and human.
Collapse
Affiliation(s)
- Takeshi Miyake
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tadahaya Mizuno
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tatsuki Mochizuki
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Miyuki Kimura
- Fukuoka Mirai Hospital Clinical Research Center, Fukuoka 813-0017, Japan
| | - Shunji Matsuki
- Fukuoka Mirai Hospital Clinical Research Center, Fukuoka 813-0017, Japan
| | - Shin Irie
- Fukuoka Mirai Hospital Clinical Research Center, Fukuoka 813-0017, Japan
| | - Ichiro Ieiri
- Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
21
|
van der Mark VA, Rudi de Waart D, Shevchenko V, Elferink RPJO, Chamuleau RAFM, Hoekstra R. Stable Overexpression of the Constitutive Androstane Receptor Reduces the Requirement for Culture with Dimethyl Sulfoxide for High Drug Metabolism in HepaRG Cells. Drug Metab Dispos 2017; 45:56-67. [PMID: 27780834 DOI: 10.1124/dmd.116.072603] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 10/24/2016] [Indexed: 01/08/2023] Open
Abstract
Dimethylsulfoxide (DMSO) induces cellular differentiation and expression of drug metabolic enzymes in the human liver cell line HepaRG; however, DMSO also induces cell death and interferes with cellular activities. The aim of this study was to examine whether overexpression of the constitutive androstane receptor (CAR, NR1I3), the nuclear receptor controlling various drug metabolism genes, would sufficiently promote differentiation and drug metabolism in HepaRG cells, optionally without using DMSO. By stable lentiviral overexpression of CAR, HepaRG cultures were less affected by DMSO in total protein content and obtained increased resistance to acetaminophen- and amiodarone-induced cell death. Transcript levels of CAR target genes were significantly increased in HepaRG-CAR cultures without DMSO, resulting in increased activities of cytochrome P450 (P450) enzymes and bilirubin conjugation to levels equal or surpassing those of HepaRG cells cultured with DMSO. Unexpectedly, CAR overexpression also increased the activities of non-CAR target P450s, as well as albumin production. In combination with DMSO treatment, CAR overexpression further increased transcript levels and activities of CAR targets. Induction of CYP1A2 and CYP2B6 remained unchanged, whereas CYP3A4 was reduced. Moreover, the metabolism of low-clearance compounds warfarin and prednisolone was increased. In conclusion, CAR overexpression creates a more physiologically relevant environment for studies on hepatic (drug) metabolism and differentiation in HepaRG cells without the utilization of DMSO. DMSO still may be applied to accomplish higher drug metabolism, required for sensitive assays, such as low-clearance studies and identification of (rare) metabolites, whereas reduced total protein content after DMSO culture is diminished by CAR overexpression.
Collapse
Affiliation(s)
- Vincent A van der Mark
- Department of Experimental Surgery (V.A.M., R.A.F.M.C., R.H.), and the Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (V.A.M., D.R.W., R.P.J.O.E., R.A.F.M.C., R.H.), Amsterdam, the Netherlands; and Biopredic International, Saint-Grégoire, France (V.S.)
| | - D Rudi de Waart
- Department of Experimental Surgery (V.A.M., R.A.F.M.C., R.H.), and the Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (V.A.M., D.R.W., R.P.J.O.E., R.A.F.M.C., R.H.), Amsterdam, the Netherlands; and Biopredic International, Saint-Grégoire, France (V.S.)
| | - Valery Shevchenko
- Department of Experimental Surgery (V.A.M., R.A.F.M.C., R.H.), and the Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (V.A.M., D.R.W., R.P.J.O.E., R.A.F.M.C., R.H.), Amsterdam, the Netherlands; and Biopredic International, Saint-Grégoire, France (V.S.)
| | - Ronald P J Oude Elferink
- Department of Experimental Surgery (V.A.M., R.A.F.M.C., R.H.), and the Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (V.A.M., D.R.W., R.P.J.O.E., R.A.F.M.C., R.H.), Amsterdam, the Netherlands; and Biopredic International, Saint-Grégoire, France (V.S.)
| | - Robert A F M Chamuleau
- Department of Experimental Surgery (V.A.M., R.A.F.M.C., R.H.), and the Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (V.A.M., D.R.W., R.P.J.O.E., R.A.F.M.C., R.H.), Amsterdam, the Netherlands; and Biopredic International, Saint-Grégoire, France (V.S.)
| | - Ruurdtje Hoekstra
- Department of Experimental Surgery (V.A.M., R.A.F.M.C., R.H.), and the Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (V.A.M., D.R.W., R.P.J.O.E., R.A.F.M.C., R.H.), Amsterdam, the Netherlands; and Biopredic International, Saint-Grégoire, France (V.S.)
| |
Collapse
|
22
|
Chen X, Wang Y, Chen X, Cheng K, Li J, Lou J, Ke J, Yang Y, Gong Y, Zhu Y, Wang L, Zhong R. Genetic variants in the regulatory region of SLC10A1 are not associated with the risk of hepatitis B virus infection and clearance. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2016; 44:495-500. [PMID: 27491457 DOI: 10.1016/j.meegid.2016.07.043] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 07/29/2016] [Accepted: 07/30/2016] [Indexed: 12/27/2022]
Abstract
The Na/taurocholate cotransporter NTCP (encoded by SLC10A1) was identified as a cellular entry receptor for the human hepatitis B virus (HBV), advancing our understanding of the molecular mechanism of HBV infection. An alternative hypothesis was put forward that regulatory variants in SLC10A1 might play an important role in HBV susceptibility by potentially influencing expression levels of NTCP. The three regulatory SNPs (rs8011311, rs7154439, rs111409076) were genotyped in 1023 HBV-persistent carriers, 735 subjects with HBV natural clearance and 732 HBV marker-negative subjects in a Han Chinese population. Real-time reverse transcription PCR analysis and luciferase assays have been performed to dissect the potential functionality. In logistic regression analysis, when subjects with HBV natural clearance were compared with HBV marker-negative subjects, no significant associations with the risk of HBV infection were observed for any of the three SNPs after adjusting for age, sex, smoking status and alcohol consumption (P>0.05). Similar negative results were also found for the three SNPs when HBV-persistent carriers were compared with HBV marker-negative subjects. Likewise, no significant associations with the risk of HBV clearance were observed when HBV-persistent carriers were compared with subjects with HBV natural clearance (P>0.05). Quantitative RT/PCR showed no significant difference in NTCP expression levels in normal liver tissue amongst individuals with different rs111409076 genotypes (P=0.317 for the general linear model). Moreover, no evident effect of the SLC10A1 rs111409076 AACA/- polymorphism on transcriptional activity was found by luciferase assay in either HepG2 (P=0.161) or Hep3b (P=0.129) cell lines. The present study indicated that the common variants in the regulatory region of SLC10A1 may not influence the expression of NTCP at the level of transcriptional regulation, and ultimately may not be associated with HBV susceptibility in this Chinese population.
Collapse
Affiliation(s)
- Xueqin Chen
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Quality Management, Jiangxi Provincial Cancer Hospital, Nanchang, Jiangxi, China
| | - Ying Wang
- Department of Virology, Wuhan Centers for Disease Prevention and Control, Wuhan, Hubei, China
| | - Xiaohua Chen
- Department of Laboratory Medicine, No 161 Hospital of PLA, Wuhan 430010, China
| | - Kailiang Cheng
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jiaoyuan Li
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiao Lou
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Juntao Ke
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yang Yang
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yajie Gong
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ying Zhu
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Li Wang
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China.
| | - Rong Zhong
- Department of Epidemiology and Biostatistics and Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
23
|
Cell Culture Models for the Investigation of Hepatitis B and D Virus Infection. Viruses 2016; 8:v8090261. [PMID: 27657111 PMCID: PMC5035974 DOI: 10.3390/v8090261] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/12/2016] [Accepted: 09/14/2016] [Indexed: 02/06/2023] Open
Abstract
Chronic hepatitis B virus (HBV) and hepatitis D virus (HDV) infections are major causes of liver disease and hepatocellular carcinoma worldwide. Despite the presence of an efficient preventive vaccine, more than 250 million patients are chronically infected with HBV. Current antivirals effectively control but only rarely cure chronic infection. While the molecular biology of the two viruses has been characterized in great detail, the absence of robust cell culture models for HBV and/or HDV infection has limited the investigation of virus-host interactions. Native hepatoma cell lines do not allow viral infection, and the culture of primary hepatocytes, the natural host cell for the viruses, implies a series of constraints restricting the possibilities of analyzing virus-host interactions. Recently, the discovery of the sodium taurocholate co-transporting polypeptide (NTCP) as a key HBV/HDV cell entry factor has opened the door to a new era of investigation, as NTCP-overexpressing hepatoma cells acquire susceptibility to HBV and HDV infections. In this review, we summarize the major cell culture models for HBV and HDV infection, discuss their advantages and limitations and highlight perspectives for future developments.
Collapse
|
24
|
Teng S, Tebby C, Barcellini-Couget S, De Sousa G, Brochot C, Rahmani R, Pery ARR. Analysis of real-time mixture cytotoxicity data following repeated exposure using BK/TD models. Toxicol Appl Pharmacol 2016; 305:118-126. [PMID: 27317371 DOI: 10.1016/j.taap.2016.06.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 06/06/2016] [Accepted: 06/13/2016] [Indexed: 11/23/2022]
Abstract
Cosmetic products generally consist of multiple ingredients. Thus, cosmetic risk assessment has to deal with mixture toxicity on a long-term scale which means it has to be assessed in the context of repeated exposure. Given that animal testing has been banned for cosmetics risk assessment, in vitro assays allowing long-term repeated exposure and adapted for in vitro - in vivo extrapolation need to be developed. However, most in vitro tests only assess short-term effects and consider static endpoints which hinder extrapolation to realistic human exposure scenarios where concentration in target organs is varies over time. Thanks to impedance metrics, real-time cell viability monitoring for repeated exposure has become possible. We recently constructed biokinetic/toxicodynamic models (BK/TD) to analyze such data (Teng et al., 2015) for three hepatotoxic cosmetic ingredients: coumarin, isoeugenol and benzophenone-2. In the present study, we aim to apply these models to analyze the dynamics of mixture impedance data using the concepts of concentration addition and independent action. Metabolic interactions between the mixture components were investigated, characterized and implemented in the models, as they impacted the actual cellular exposure. Indeed, cellular metabolism following mixture exposure induced a quick disappearance of the compounds from the exposure system. We showed that isoeugenol substantially decreased the metabolism of benzophenone-2, reducing the disappearance of this compound and enhancing its in vitro toxicity. Apart from this metabolic interaction, no mixtures showed any interaction, and all binary mixtures were successfully modeled by at least one model based on exposure to the individual compounds.
Collapse
Affiliation(s)
- S Teng
- Models for Toxicology and Ecotoxicology Unit, INERIS, Parc Technologique Alata, BP 2, 60550 Verneuil-en-Halatte, France
| | - C Tebby
- Models for Toxicology and Ecotoxicology Unit, INERIS, Parc Technologique Alata, BP 2, 60550 Verneuil-en-Halatte, France
| | - S Barcellini-Couget
- ODESIA Neosciences, Sophia Antipolis, 400 route des chappes, 06903 Sophia Antipolis, France
| | - G De Sousa
- INRA, ToxAlim, 400 route des Chappes, BP, 167 06903 Sophia Antipolis, Cedex, France
| | - C Brochot
- Models for Toxicology and Ecotoxicology Unit, INERIS, Parc Technologique Alata, BP 2, 60550 Verneuil-en-Halatte, France
| | - R Rahmani
- INRA, ToxAlim, 400 route des Chappes, BP, 167 06903 Sophia Antipolis, Cedex, France
| | - A R R Pery
- AgroParisTech, UMR 1402 INRA-AgroParisTech Ecosys, 78850 Thiverval Grignon, France; INRA, UMR 1402 INRA-AgroParisTech Ecosys, 78850 Thiverval Grignon, France.
| |
Collapse
|
25
|
Alteration of human hepatic drug transporter activity and expression by cigarette smoke condensate. Toxicology 2016; 363-364:58-71. [DOI: 10.1016/j.tox.2016.07.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/04/2016] [Accepted: 07/19/2016] [Indexed: 02/07/2023]
|
26
|
Riley RJ, Foley SA, Barton P, Soars MG, Williamson B. Hepatic drug transporters: the journey so far. Expert Opin Drug Metab Toxicol 2016; 12:201-16. [PMID: 26670591 DOI: 10.1517/17425255.2016.1132308] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION The key role of transporter biology in both the manifestation and treatment of disease is now firmly established. Experiences of sub-optimal drug exposure due to drug-transporter interplay have supported incorporation of studies aimed at understanding the interactions between compounds and drug transporters much earlier in drug discovery. While drug transporters can impact the most pivotal pharmacokinetic parameter with respect to human dose and exposure projections, clearance, at a renal or hepatobiliary level, the latter will form the focus of this perspective. AREAS COVERED A synopsis of guidelines on which transporters to study together with an overview of the currently available toolkit is presented. A perspective on when to conduct studies with various hepatic transporters is also provided together with structural "alerts" which should prompt early investigation. EXPERT OPINION Great progress has been made in individual laboratories and via consortia to understand the role of drug transporters in disease, drug disposition, drug-drug interactions and toxicity. A systematic analysis of the value posed by the available approaches and an inter-lab comparison now seems warranted. The emerging ability to use physico-chemical properties to guide future screening cascades promises to revolutionise the efficiency of early drug discovery.
Collapse
Affiliation(s)
| | | | - P Barton
- b School of Life Sciences , University of Nottingham , Nottingham , UK
| | - M G Soars
- c Drug Metabolism and Pharmacokinetics , Bristol-Myers Squibb , Wallingford , CT , USA
| | | |
Collapse
|
27
|
Mayati A, Bruyere A, Moreau A, Jouan E, Denizot C, Parmentier Y, Fardel O. Protein Kinase C-Independent Inhibition of Organic Cation Transporter 1 Activity by the Bisindolylmaleimide Ro 31-8220. PLoS One 2015; 10:e0144667. [PMID: 26657401 PMCID: PMC4675551 DOI: 10.1371/journal.pone.0144667] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 11/20/2015] [Indexed: 02/02/2023] Open
Abstract
Ro 31–8220 is a potent protein kinase C (PKC) inhibitor belonging to the chemical class of bisindolylmaleimides (BIMs). Various PKC-independent effects of Ro 31–8220 have however been demonstrated, including inhibition of the ATP-binding cassette drug transporter breast cancer resistance protein. In the present study, we reported that the BIM also blocks activity of the solute carrier organic cation transporter (OCT) 1, involved in uptake of marketed drugs in the liver, in a PKC-independent manner. Ro 31–8220, in contrast to other pan-PKC inhibitors such as staurosporine and chelerythrine, was thus shown to cis-inhibit uptake of the reference OCT1 substrate tetraethylammonium in OCT1-transfected HEK293 cells in a concentration-dependent manner (IC50 = 0.18 μM) and without altering membrane expression of OCT1. This blockage of OCT1 was also observed in human hepatic HepaRG cells that constitutionally express OCT1. It likely occurred through a mixed mechanism of inhibition. Ro 31–8220 additionally trans-inhibited TEA uptake in OCT1-transfected HEK293 cells, which likely discards a transport of Ro 31–8220 by OCT1. Besides Ro 31–8220, 7 additional BIMs, including the PKC inhibitor LY 333531, inhibited OCT1 activity, whereas 4 other BIMs were without effect. In silico analysis of structure-activity relationships next revealed that various molecular descriptors, especially 3D-WHIM descriptors related to total size, correspond to key physico-chemical parameters for inhibition of OCT1 activity by BIMs. In addition to activity of OCT1, Ro 31–8220 inhibited those of other organic cation transporters such as multidrug and toxin extrusion protein (MATE) 1 and MATE2-K, whereas, by contrast, it stimulated that of OCT2. Taken together, these data extend the nature of cellular off-targets of the BIM Ro 31–8220 to OCT1 and other organic cation transporters, which has likely to be kept in mind when using Ro 31–8220 and other BIMs as PKC inhibitors in experimental or clinical studies.
Collapse
Affiliation(s)
- Abdullah Mayati
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie, 2 Avenue du Pr Léon Bernard, 35043, Rennes, France
| | - Arnaud Bruyere
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie, 2 Avenue du Pr Léon Bernard, 35043, Rennes, France
| | - Amélie Moreau
- Centre de Pharmacocinétique, Technologie Servier, 25–27 rue Eugène Vignat, 45000, Orléans, France
| | - Elodie Jouan
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie, 2 Avenue du Pr Léon Bernard, 35043, Rennes, France
| | - Claire Denizot
- Centre de Pharmacocinétique, Technologie Servier, 25–27 rue Eugène Vignat, 45000, Orléans, France
| | - Yannick Parmentier
- Centre de Pharmacocinétique, Technologie Servier, 25–27 rue Eugène Vignat, 45000, Orléans, France
| | - Olivier Fardel
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie, 2 Avenue du Pr Léon Bernard, 35043, Rennes, France
- Pôle Biologie, Centre Hospitalier Universitaire, 2 rue Henri Le Guilloux, 35033, Rennes, France
- * E-mail:
| |
Collapse
|
28
|
Ambolet-Camoit A, Ottolenghi C, Leblanc A, Kim MJ, Letourneur F, Jacques S, Cagnard N, Guguen-Guillouzo C, Barouki R, Aggerbeck M. Two persistent organic pollutants which act through different xenosensors (alpha-endosulfan and 2,3,7,8 tetrachlorodibenzo-p-dioxin) interact in a mixture and downregulate multiple genes involved in human hepatocyte lipid and glucose metabolism. Biochimie 2015; 116:79-91. [PMID: 26159488 DOI: 10.1016/j.biochi.2015.07.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 07/03/2015] [Indexed: 11/16/2022]
Abstract
Individuals, typically, are exposed to mixtures of environmental xenobiotics affecting multiple organs and acting through different xenosensors and pathways in species and cell-type specific manners. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and α-endosulfan are Persistent Organic Pollutants (POPs) and endocrine disruptors which act through different xenosensors and accumulate in the liver. Our objective in this HEALS study was to investigate the effects of the mixture of these POPs on gene expression in a human-derived hepatocyte cell line, HepaRG. We found that, in spite of having largely uncorrelated effects, TCDD and α-endosulfan, when mixed, alter the expression of genes. The combined effects of the mixture of the POPs significantly altered the expression of 100 genes (42 up- and 58 down-regulated) whereas the same concentration of either POP alone did not alter significantly the expression of these genes. For 32 other genes, selective inhibitory crosstalk between TCDD and α-endosulfan was observed. One of the POPs inhibited the effect, on gene expression, of the other in the mixture although, when used alone, that POP did not affect expression. The expression of another 82 genes was significantly altered (up- or down-regulated) by a single POP. The addition of the second POP either increased, in the same direction, the effect on gene expression or had no further effect. At low concentrations (0.2 nM TCDD and 1 μM α-endosulfan), the POPs still had significant effects and the levels of expression of the corresponding proteins were found to be affected for some genes. Particularly striking was the 80-90% inhibition, by the mixture, of the expression of a number of genes of several hepatic intermediary metabolic pathways (glycerolipid metabolism, FXR/RXR activation, glycolysis/gluconeogenesis, retinoid and bile acid biosynthesis), whereas each pollutant alone had only a moderate effect.
Collapse
Affiliation(s)
- Ariane Ambolet-Camoit
- Inserm UMR-S 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Chris Ottolenghi
- Inserm UMR-S 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; APHP, Hôpital Necker Enfants Malades, Service de Biochimie Métabolique, Paris, France
| | - Alix Leblanc
- Inserm UMR-S 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Min Ji Kim
- Inserm UMR-S 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Franck Letourneur
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Plateforme de Génomique, Institut Cochin, Paris, France
| | - Sébastien Jacques
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Plateforme de Génomique, Institut Cochin, Paris, France
| | - Nicolas Cagnard
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Plateforme de Génomique, Institut Cochin, Paris, France
| | | | - Robert Barouki
- Inserm UMR-S 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; APHP, Hôpital Necker Enfants Malades, Service de Biochimie Métabolique, Paris, France
| | - Martine Aggerbeck
- Inserm UMR-S 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
29
|
Rehman Z, Fahim A, Sadia H. Deciphering the mystery of hepatitis B virus receptors: A historical perspective. Virusdisease 2015; 26:97-104. [PMID: 26396975 DOI: 10.1007/s13337-015-0260-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 06/25/2015] [Indexed: 12/22/2022] Open
Abstract
Hepatitis B virus is one of the major reasons of viral hepatitis with an estimated 350 million infected patients worldwide. Although, the virus was discovered and cloned more than three decades ago, its entry mechanism has still been in investigation. Numerous potential candidates have been proposed and investigated rigorously to reveal HBV entry mechanism and to unveil the first door of viral entry to hepatocytes. This review provides a short account of role of receptors for entry of HBV into hepatocytes. The viral preS1 region of large surface protein is involved in the attachment of HBV to hepatocytes. The putative attachment site of HBV is located at amino acids 21-47 of preS1. So far, several proteins have been proposed to interact with these different regions of the preS1 domain which includes human immunoglobulin A receptor, glyceraldehyde-3-phosphate dehydrogenase, interleukin-6, a 31-kDa protein, HBV binding factor, asialoglycoprotein receptor, nascent polypeptide-associated complex α polypeptide, lipoprotein lipase, hepatocyte-associated heparan sulfate proteoglycans, glucose-regulated protein 75. However, none of them have appeared to be generally accepted as a true receptor for the virus until recently when sodium taurocholate cotransporting polypeptide identified as HBV entry receptor. Current review provides scientific historical perspective of various candidates known to be interacting with preS1 of HBV for their possible role in viral entry.
Collapse
Affiliation(s)
- Zaira Rehman
- Healthcare Biotechnology Department, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences & Technology (NUST), Sector H-12, Islamabad, Pakistan
| | - Ammad Fahim
- Healthcare Biotechnology Department, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences & Technology (NUST), Sector H-12, Islamabad, Pakistan
| | - Hajra Sadia
- Healthcare Biotechnology Department, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences & Technology (NUST), Sector H-12, Islamabad, Pakistan
| |
Collapse
|
30
|
Yan R, Zhang Y, Cai D, Liu Y, Cuconati A, Guo H. Spinoculation Enhances HBV Infection in NTCP-Reconstituted Hepatocytes. PLoS One 2015; 10:e0129889. [PMID: 26070202 PMCID: PMC4466484 DOI: 10.1371/journal.pone.0129889] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 05/14/2015] [Indexed: 12/12/2022] Open
Abstract
Hepatitis B virus (HBV) infection and its sequelae remain a major public health burden, but both HBV basic research and the development of antiviral therapeutics have been hindered by the lack of an efficient in vitro infection system. Recently, sodium taurocholate cotransporting polypeptide (NTCP) has been identified as the HBV receptor. We herein report that we established a NTCP-complemented HepG2 cell line (HepG2-NTCP12) that supports HBV infection, albeit at a low infectivity level following the reported infection procedures. In our attempts to optimize the infection conditions, we found that the centrifugation of HepG2-NTCP12 cells during HBV inoculation (termed “spinoculation”) significantly enhanced the virus infectivity. Moreover, the infection level gradually increased with accelerated speed of spinoculation up to 1,000g tested. However, the enhancement of HBV infection was not significantly dependent upon the duration of centrifugation. Furthermore, covalently closed circular (ccc) DNA was detected in infected cells under optimized infection condition by conventional Southern blot, suggesting a successful establishment of HBV infection after spinoculation. Finally, the parental HepG2 cells remained uninfected under HBV spinoculation, and HBV entry inhibitors targeting NTCP blocked HBV infection when cells were spinoculated, suggesting the authentic virus entry mechanism is unaltered under centrifugal inoculation. Our data suggest that spinoculation could serve as a standard protocol for enhancing the efficiency of HBV infection in vitro.
Collapse
Affiliation(s)
- Ran Yan
- Department of Microbiology and Immunology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, Indiana, 46202, United States of America
| | - Yongmei Zhang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Rd, Shanghai, 200040, China
| | - Dawei Cai
- Department of Microbiology and Immunology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, Indiana, 46202, United States of America
| | - Yuanjie Liu
- Department of Microbiology and Immunology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, Indiana, 46202, United States of America
| | - Andrea Cuconati
- Baruch S. Blumberg Institute, Hepatitis B Foundation, 3805 Old Easton Rd, Doylestown, Pennsylvania, 18902, United States of America
| | - Haitao Guo
- Department of Microbiology and Immunology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, Indiana, 46202, United States of America
- * E-mail:
| |
Collapse
|
31
|
Tsukuda S, Watashi K, Iwamoto M, Suzuki R, Aizaki H, Okada M, Sugiyama M, Kojima S, Tanaka Y, Mizokami M, Li J, Tong S, Wakita T. Dysregulation of retinoic acid receptor diminishes hepatocyte permissiveness to hepatitis B virus infection through modulation of sodium taurocholate cotransporting polypeptide (NTCP) expression. J Biol Chem 2015; 290:5673-5684. [PMID: 25550158 PMCID: PMC4342479 DOI: 10.1074/jbc.m114.602540] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 12/20/2014] [Indexed: 12/15/2022] Open
Abstract
Sodium taurocholate cotransporting polypeptide (NTCP) is an entry receptor for hepatitis B virus (HBV) and is regarded as one of the determinants that confer HBV permissiveness to host cells. However, how host factors regulate the ability of NTCP to support HBV infection is largely unknown. We aimed to identify the host signaling that regulated NTCP expression and thereby permissiveness to HBV. Here, a cell-based chemical screening method identified that Ro41-5253 decreased host susceptibility to HBV infection. Pretreatment with Ro41-5253 inhibited the viral entry process without affecting HBV replication. Intriguingly, Ro41-5253 reduced expression of both NTCP mRNA and protein. We found that retinoic acid receptor (RAR) regulated the promoter activity of the human NTCP (hNTCP) gene and that Ro41-5253 repressed the hNTCP promoter by antagonizing RAR. RAR recruited to the hNTCP promoter region, and nucleotides -112 to -96 of the hNTCP was suggested to be critical for RAR-mediated transcriptional activation. HBV susceptibility was decreased in pharmacologically RAR-inactivated cells. CD2665 showed a stronger anti-HBV potential and disrupted the spread of HBV infection that was achieved by continuous reproduction of the whole HBV life cycle. In addition, this mechanism was significant for drug development, as antagonization of RAR blocked infection of multiple HBV genotypes and also a clinically relevant HBV mutant that was resistant to nucleoside analogs. Thus, RAR is crucial for regulating NTCP expression that determines permissiveness to HBV infection. This is the first demonstration showing host regulation of NTCP to support HBV infection.
Collapse
Affiliation(s)
- Senko Tsukuda
- From the Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan, the Micro-signaling Regulation Technology Unit, RIKEN Center for Life Science Technologies, Wako 351-0198, Japan
| | - Koichi Watashi
- From the Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan,
| | - Masashi Iwamoto
- From the Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Ryosuke Suzuki
- From the Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Hideki Aizaki
- From the Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Maiko Okada
- the Department of Translational Oncology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan
| | - Masaya Sugiyama
- the Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa 272-8516, Japan
| | - Soichi Kojima
- the Micro-signaling Regulation Technology Unit, RIKEN Center for Life Science Technologies, Wako 351-0198, Japan
| | - Yasuhito Tanaka
- the Department of Virology and Liver Unit, Nagoya City University Graduate School of Medicinal Sciences, Nagoya 467-8601, Japan, and
| | - Masashi Mizokami
- the Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa 272-8516, Japan
| | - Jisu Li
- the Liver Research Center Rhode Island Hospital, Warren Alpert School of Medicine, Brown University, Providence, Rhode Island 02912
| | - Shuping Tong
- the Liver Research Center Rhode Island Hospital, Warren Alpert School of Medicine, Brown University, Providence, Rhode Island 02912
| | - Takaji Wakita
- From the Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| |
Collapse
|
32
|
Bachour-El Azzi P, Sharanek A, Burban A, Li R, Guével RL, Abdel-Razzak Z, Stieger B, Guguen-Guillouzo C, Guillouzo A. Comparative Localization and Functional Activity of the Main Hepatobiliary Transporters in HepaRG Cells and Primary Human Hepatocytes. Toxicol Sci 2015; 145:157-68. [PMID: 25690737 DOI: 10.1093/toxsci/kfv041] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The role of hepatobiliary transporters in drug-induced liver injury remains poorly understood. Various in vivo and in vitro biological approaches are currently used for studying hepatic transporters; however, appropriate localization and functional activity of these transporters are essential for normal biliary flow and drug transport. Human hepatocytes (HHs) are considered as the most suitable in vitro cell model but erratic availability and inter-donor functional variations limit their use. In this work, we aimed to compare localization of influx and efflux transporters and their functional activity in differentiated human HepaRG hepatocytes with fresh HHs in conventional (CCHH) and sandwich (SCHH) cultures. All tested influx and efflux transporters were correctly localized to canalicular [bile salt export pump (BSEP), multidrug resistance-associated protein 2 (MRP2), multidrug resistance protein 1 (MDR1), and MDR3] or basolateral [Na(+)-taurocholate co-transporting polypeptide (NTCP) and MRP3] membrane domains and were functional in all models. Contrary to other transporters, NTCP and BSEP were less abundant and active in HepaRG cells, cellular uptake of taurocholate was 2.2- and 1.4-fold and bile excretion index 2.8- and 2.6-fold lower, than in SCHHs and CCHHs, respectively. However, when taurocholate canalicular efflux was evaluated in standard and divalent cation-free conditions in buffers or cell lysates, the difference between the three models did not exceed 9.3%. Interestingly, cell imaging showed higher bile canaliculi contraction/relaxation activity in HepaRG hepatocytes and larger bile canaliculi networks in SCHHs. Altogether, our results bring new insights in mechanisms involved in bile acids accumulation and excretion in HHs and suggest that HepaRG cells represent a suitable model for studying hepatobiliary transporters and drug-induced cholestasis.
Collapse
Affiliation(s)
- Pamela Bachour-El Azzi
- *Inserm UMR991, Foie, Métabolismes et Cancer, Rennes, France; Université de Rennes 1, Rennes, France, Université Libanaise, EDST-PRASE and EDST-AZM-center-LBA3B, Beirut, Lebanon, Biopredic International, Saint Grégoire, France, ImPACcell, SFR Biosit, Université de Rennes 1, Rennes, France and Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland *Inserm UMR991, Foie, Métabolismes et Cancer, Rennes, France; Université de Rennes 1, Rennes, France, Université Libanaise, EDST-PRASE and EDST-AZM-center-LBA3B, Beirut, Lebanon, Biopredic International, Saint Grégoire, France, ImPACcell, SFR Biosit, Université de Rennes 1, Rennes, France and Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland *Inserm UMR991, Foie, Métabolismes et Cancer, Rennes, France; Université de Rennes 1, Rennes, France, Université Libanaise, EDST-PRASE and EDST-AZM-center-LBA3B, Beirut, Lebanon, Biopredic International, Saint Grégoire, France, ImPACcell, SFR Biosit, Université de Rennes 1, Rennes, France and Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland
| | - Ahmad Sharanek
- *Inserm UMR991, Foie, Métabolismes et Cancer, Rennes, France; Université de Rennes 1, Rennes, France, Université Libanaise, EDST-PRASE and EDST-AZM-center-LBA3B, Beirut, Lebanon, Biopredic International, Saint Grégoire, France, ImPACcell, SFR Biosit, Université de Rennes 1, Rennes, France and Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland *Inserm UMR991, Foie, Métabolismes et Cancer, Rennes, France; Université de Rennes 1, Rennes, France, Université Libanaise, EDST-PRASE and EDST-AZM-center-LBA3B, Beirut, Lebanon, Biopredic International, Saint Grégoire, France, ImPACcell, SFR Biosit, Université de Rennes 1, Rennes, France and Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland
| | - Audrey Burban
- *Inserm UMR991, Foie, Métabolismes et Cancer, Rennes, France; Université de Rennes 1, Rennes, France, Université Libanaise, EDST-PRASE and EDST-AZM-center-LBA3B, Beirut, Lebanon, Biopredic International, Saint Grégoire, France, ImPACcell, SFR Biosit, Université de Rennes 1, Rennes, France and Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland *Inserm UMR991, Foie, Métabolismes et Cancer, Rennes, France; Université de Rennes 1, Rennes, France, Université Libanaise, EDST-PRASE and EDST-AZM-center-LBA3B, Beirut, Lebanon, Biopredic International, Saint Grégoire, France, ImPACcell, SFR Biosit, Université de Rennes 1, Rennes, France and Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland
| | - Ruoya Li
- *Inserm UMR991, Foie, Métabolismes et Cancer, Rennes, France; Université de Rennes 1, Rennes, France, Université Libanaise, EDST-PRASE and EDST-AZM-center-LBA3B, Beirut, Lebanon, Biopredic International, Saint Grégoire, France, ImPACcell, SFR Biosit, Université de Rennes 1, Rennes, France and Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland
| | - Rémy Le Guével
- *Inserm UMR991, Foie, Métabolismes et Cancer, Rennes, France; Université de Rennes 1, Rennes, France, Université Libanaise, EDST-PRASE and EDST-AZM-center-LBA3B, Beirut, Lebanon, Biopredic International, Saint Grégoire, France, ImPACcell, SFR Biosit, Université de Rennes 1, Rennes, France and Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland
| | - Ziad Abdel-Razzak
- *Inserm UMR991, Foie, Métabolismes et Cancer, Rennes, France; Université de Rennes 1, Rennes, France, Université Libanaise, EDST-PRASE and EDST-AZM-center-LBA3B, Beirut, Lebanon, Biopredic International, Saint Grégoire, France, ImPACcell, SFR Biosit, Université de Rennes 1, Rennes, France and Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland
| | - Bruno Stieger
- *Inserm UMR991, Foie, Métabolismes et Cancer, Rennes, France; Université de Rennes 1, Rennes, France, Université Libanaise, EDST-PRASE and EDST-AZM-center-LBA3B, Beirut, Lebanon, Biopredic International, Saint Grégoire, France, ImPACcell, SFR Biosit, Université de Rennes 1, Rennes, France and Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland *Inserm UMR991, Foie, Métabolismes et Cancer, Rennes, France; Université de Rennes 1, Rennes, France, Université Libanaise, EDST-PRASE and EDST-AZM-center-LBA3B, Beirut, Lebanon, Biopredic International, Saint Grégoire, France, ImPACcell, SFR Biosit, Université de Rennes 1, Rennes, France and Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland
| | - Christiane Guguen-Guillouzo
- *Inserm UMR991, Foie, Métabolismes et Cancer, Rennes, France; Université de Rennes 1, Rennes, France, Université Libanaise, EDST-PRASE and EDST-AZM-center-LBA3B, Beirut, Lebanon, Biopredic International, Saint Grégoire, France, ImPACcell, SFR Biosit, Université de Rennes 1, Rennes, France and Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland
| | - André Guillouzo
- *Inserm UMR991, Foie, Métabolismes et Cancer, Rennes, France; Université de Rennes 1, Rennes, France, Université Libanaise, EDST-PRASE and EDST-AZM-center-LBA3B, Beirut, Lebanon, Biopredic International, Saint Grégoire, France, ImPACcell, SFR Biosit, Université de Rennes 1, Rennes, France and Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland *Inserm UMR991, Foie, Métabolismes et Cancer, Rennes, France; Université de Rennes 1, Rennes, France, Université Libanaise, EDST-PRASE and EDST-AZM-center-LBA3B, Beirut, Lebanon, Biopredic International, Saint Grégoire, France, ImPACcell, SFR Biosit, Université de Rennes 1, Rennes, France and Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland
| |
Collapse
|
33
|
Urban S, Bartenschlager R, Kubitz R, Zoulim F. Strategies to inhibit entry of HBV and HDV into hepatocytes. Gastroenterology 2014; 147:48-64. [PMID: 24768844 DOI: 10.1053/j.gastro.2014.04.030] [Citation(s) in RCA: 254] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 03/11/2014] [Accepted: 04/21/2014] [Indexed: 02/07/2023]
Abstract
Although there has been much research into the pathogenesis and treatment of hepatitis B virus (HBV) and hepatitis D virus (HDV) infections, we still do not completely understand how these pathogens enter hepatocytes. This is because in vitro infection studies have only been performed in primary human hepatocytes. Development of a polarizable, HBV-susceptible human hepatoma cell line and studies of primary hepatocytes from Tupaia belangeri have provided important insights into the viral and cellular factors involved in virus binding and infection. The large envelope (L) protein on the surface of HBV and HDV particles has many different functions and is required for virus entry. The L protein mediates attachment of virions to heparan sulfate proteoglycans on the surface of hepatocytes. The myristoylated N-terminal preS1 domain of the L protein subsequently binds to the sodium taurocholate cotransporting polypeptide (NTCP, encoded by SLC10A1), the recently identified bona fide receptor for HBV and HDV. The receptor functions of NTCP and virus entry are blocked, in vitro and in vivo, by Myrcludex B, a synthetic N-acylated preS1 lipopeptide. Currently, the only agents available to treat chronic HBV infection target the viral polymerase, and no selective therapies are available for HDV infection. It is therefore important to study the therapeutic potential of virus entry inhibitors, especially when combined with strategies to induce immune-mediated killing of infected hepatocytes.
Collapse
Affiliation(s)
- Stephan Urban
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany; German Center for Infection Research, Heidelberg University, Heidelberg, Germany.
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany; German Center for Infection Research, Heidelberg University, Heidelberg, Germany
| | - Ralf Kubitz
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Fabien Zoulim
- INSERM Unité 1052, Cancer Research Center of Lyon, Lyon University, Lyon, France
| |
Collapse
|
34
|
Ni Y, Lempp FA, Mehrle S, Nkongolo S, Kaufman C, Fälth M, Stindt J, Königer C, Nassal M, Kubitz R, Sültmann H, Urban S. Hepatitis B and D viruses exploit sodium taurocholate co-transporting polypeptide for species-specific entry into hepatocytes. Gastroenterology 2014; 146:1070-83. [PMID: 24361467 DOI: 10.1053/j.gastro.2013.12.024] [Citation(s) in RCA: 610] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 12/04/2013] [Accepted: 12/07/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Hepatitis B and D viruses (HBV and HDV) are human pathogens with restricted host ranges and high selectivity for hepatocytes; the HBV L-envelope protein interacts specifically with a receptor on these cells. We aimed to identify this receptor and analyze whether it is the recently described sodium-taurocholate co-transporter polypeptide (NTCP), encoded by the SLC10A1 gene. METHODS To identify receptor candidates, we compared gene expression patterns between differentiated HepaRG cells, which express the receptor, and naïve cells, which do not. Receptor candidates were evaluated by small hairpin RNA silencing in HepaRG cells; the ability of receptor expression to confer binding and infection were tested in transduced hepatoma cell lines. We used interspecies domain swapping to identify motifs for receptor-mediated host discrimination of HBV and HDV binding and infection. RESULTS Bioinformatic analyses of comparative expression arrays confirmed that NTCP, which was previously identified through a biochemical approach is a bona fide receptor for HBV and HDV. NTCPs from rat, mouse, and human bound Myrcludex B, a peptide ligand derived from the HBV L-protein. Myrcludex B blocked NTCP transport of bile salts; small hairpin RNA-mediated knockdown of NTCP in HepaRG cells prevented their infection by HBV or HDV. Expression of human but not mouse NTCP in HepG2 and HuH7 cells conferred a limited cell-type-related and virus-dependent susceptibility to infection; these limitations were overcome when cells were cultured with dimethyl sulfoxide. We identified 2 short-sequence motifs in human NTCP that were required for species-specific binding and infection by HBV and HDV. CONCLUSIONS Human NTCP is a specific receptor for HBV and HDV. NTCP-expressing cell lines can be efficiently infected with these viruses, and might be used in basic research and high-throughput screening studies. Mapping of motifs in NTCPs have increased our understanding of the species specificities of HBV and HDV, and could lead to small animal models for studies of viral infection and replication.
Collapse
Affiliation(s)
- Yi Ni
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Florian A Lempp
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Mehrle
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Shirin Nkongolo
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Christina Kaufman
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Maria Fälth
- German Cancer Research Center and National Center for Tumor Diseases, Unit Cancer Genome Research, Heidelberg, Germany
| | - Jan Stindt
- Clinic for Gastroenterology, Hepatology and Infectiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Christian Königer
- Department of Internal Medicine II, University Hospital Freiburg, Freiburg, Germany
| | - Michael Nassal
- Department of Internal Medicine II, University Hospital Freiburg, Freiburg, Germany
| | - Ralf Kubitz
- Clinic for Gastroenterology, Hepatology and Infectiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Holger Sültmann
- German Cancer Research Center and National Center for Tumor Diseases, Unit Cancer Genome Research, Heidelberg, Germany
| | - Stephan Urban
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
35
|
Watashi K, Urban S, Li W, Wakita T. NTCP and beyond: opening the door to unveil hepatitis B virus entry. Int J Mol Sci 2014; 15:2892-905. [PMID: 24557582 PMCID: PMC3958888 DOI: 10.3390/ijms15022892] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 02/13/2014] [Accepted: 02/14/2014] [Indexed: 12/19/2022] Open
Abstract
Chronic hepatitis B virus (HBV) infection, affecting approximately 240 million people worldwide, is a major public health problem that elevates the risk of developing liver cirrhosis and hepatocellular carcinoma. Given that current anti-HBV drugs are limited to interferon-based regimens and nucleos(t)ide analogs, the development of new anti-HBV agents is urgently needed. The viral entry process is generally an attractive target implicated in antiviral strategies. Using primary cells from humans and Tupaia belangeri, as well as HepaRG cells, important determinants of viral entry have been achieved. Recently, sodium taurocholate cotransporting polypeptide (NTCP) was identified as an HBV entry receptor and enabled the establishment of a susceptible cell line that can efficiently support HBV infection. This finding will allow a deeper understanding of the requirements for efficient HBV infection, including the elucidation of the molecular entry mechanism. In addition, pharmacological studies suggest that NTCP is able to serve as a therapeutic target. This article summarizes our current knowledge on the mechanisms of HBV entry and the role of NTCP in this process.
Collapse
Affiliation(s)
- Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, 162-8640 Tokyo, Japan.
| | - Stephan Urban
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany.
| | - Wenhui Li
- National Institute of Biological Sciences, No.7 Science Park Road, ZGC Life Science Park, Changping, 102206 Beijing, China.
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, 162-8640 Tokyo, Japan.
| |
Collapse
|
36
|
Le Vee M, Noel G, Jouan E, Stieger B, Fardel O. Polarized expression of drug transporters in differentiated human hepatoma HepaRG cells. Toxicol In Vitro 2013; 27:1979-86. [PMID: 23850984 DOI: 10.1016/j.tiv.2013.07.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Revised: 07/02/2013] [Accepted: 07/03/2013] [Indexed: 11/28/2022]
Abstract
The HepaRG cell line is a well-differentiated human hepatoma cell line proposed as a surrogate for human hepatocytes, especially for hepatic detoxification studies. Polarized status of drug transporters, i.e., their coordinated location at sinusoidal or canalicular membranes, which represents a key hallmark of hepato-biliary drug transport, remains however incompletely documented in HepaRG cells. The present study was therefore designed to analyze transporter location in HepaRG cells, which exhibit mRNA expressions of most of hepatic transporters. HepaRG cells were demonstrated, through immunofluorescence staining, to express several drug transporters at their sinusoidal pole, especially the influx transporters organic anion transporting polypeptide (OATP) 1B1, OATP2B1 and organic cation transporter (OCT) 1 and the efflux transporter multidrug resistance-associated protein (MRP) 3. In addition, the efflux transporters P-glycoprotein and MRP2 were detected at the canalicular pole of HepaRG cells. Moreover, saturable uptake of reference substrates for the sinusoidal transporters sodium-taurocholate cotransporting polypeptide, OATPs and OCT1 and canalicular secretion of reference substrates for the efflux transporters bile salt export pump and MRP2 were observed. This polarized and functional expression of various sinusoidal and canalicular transporters in HepaRG cells highlights the interest of using these hepatoma cells in xenobiotic transport studies.
Collapse
Affiliation(s)
- Marc Le Vee
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie, 2 Avenue du Pr Léon Bernard, 35043 Rennes, France
| | | | | | | | | |
Collapse
|
37
|
Comparison of human hepatoma HepaRG cells with human and rat hepatocytes in uptake transport assays in order to predict a risk of drug induced hepatotoxicity. PLoS One 2013; 8:e59432. [PMID: 23516635 PMCID: PMC3597610 DOI: 10.1371/journal.pone.0059432] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 02/11/2013] [Indexed: 12/02/2022] Open
Abstract
Human hepatocytes are the gold standard for toxicological studies but they have several drawbacks, like scarce availability, high inter-individual variability, a short lifetime, which limits their applicability. The aim of our investigations was to determine, whether HepaRG cells could replace human hepatocytes in uptake experiments for toxicity studies. HepaRG is a hepatoma cell line with most hepatic functions, including a considerable expression of uptake transporters in contrast to other hepatic immortalized cell lines. We compared the effect of cholestatic drugs (bosentan, cyclosporinA, troglitazone,) and bromosulfophthalein on the uptake of taurocholate and estrone-3-sulfate in human and rat hepatocytes and HepaRG cells. The substrate uptake was significantly slower in HepaRG cells than in human hepatocytes, still, in the presence of drugs we observed a concentration dependent decrease in uptake. In all cell types, the culture time had a significant impact not only on the uptake process but on the inhibitory effect of drugs too. The most significant drug effect was measured at 4 h after seeding. Our report is among the first concerning interactions of the uptake transporters in the HepaRG, at the functional level. Results of the present study clearly show that concerning the inhibition of taurocholate uptake by cholestatic drugs, HepaRG cells are closer to human hepatocytes than rat hepatocytes. In conclusion, we demonstrated that HepaRG cells may provide a suitable tool for hepatic uptake studies.
Collapse
|
38
|
Differential regulation of drug transporter expression by all-trans retinoic acid in hepatoma HepaRG cells and human hepatocytes. Eur J Pharm Sci 2013; 48:767-74. [PMID: 23352986 DOI: 10.1016/j.ejps.2013.01.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 12/26/2012] [Accepted: 01/10/2013] [Indexed: 12/13/2022]
Abstract
All-trans retinoic acid (atRA) is the active form of vitamin A, known to activate retinoid receptors, especially the heterodimer retinoid X receptor (RXR):retinoic acid receptor (RAR) that otherwise may play a role in regulation of some drug transporters. The present study was designed to characterize the nature of human hepatic transporters that may be targeted by atRA and the heterodimer RXR:RAR. Exposure of human hepatoma HepaRG cells and primary human hepatocytes to 5 μM atRA down-regulated mRNA levels of various sinusoidal solute carrier (SLC) influx transporters, including organic anion transporting polypeptide (OATP) 2B1, OATP1B1, organic cation transporter (OCT) 1 and organic anion transporter (OAT) 2, and induced those of the canalicular breast cancer resistance protein (BCRP). The retinoid concomitantly reduced protein expression of OATP2B1 and OATP1B1 and activity of OATPs and OCT1 and induced BCRP protein expression in HepaRG cells. Some transporters such as OATP1B3 and the bile salt export pump (BSEP) were however down-regulated by atRA in primary human hepatocytes, but induced in HepaRG cells, thus pointing out discrepancies between these two liver cell models in terms of detoxifying protein regulation. atRA-mediated repressions of OATP2B1, OATP1B1, OAT2 and OCT1 mRNA expression were finally shown to be counteracted by knocking-down expression of RARα and RXRα through siRNA transfection in HepaRG cells. atRA thus differentially regulated human hepatic drug transporters, mainly in a RXR:RAR-dependent manner, therefore establishing retinoids and retinoid receptors as modulators of liver drug transporter expression.
Collapse
|
39
|
Chen PJ, Wu TC. One step closer to an experimental infection system for Hepatitis B Virus? --- the identification of sodium taurocholate cotransporting peptide as a viral receptor. Cell Biosci 2013; 3:2. [PMID: 23311606 PMCID: PMC3562259 DOI: 10.1186/2045-3701-3-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 01/07/2013] [Indexed: 01/05/2023] Open
Abstract
Following the successful cloning of receptor for SARS coronavirus a few years ago, Dr. Wenhui Li and colleagues raised attention again by publishing a possible receptor for hepatitis B virus in eLife. We will briefly review the significance of this finding and the future prospects of hepatitis B research.
Collapse
Affiliation(s)
- Pei-Jer Chen
- Hepatitis Research Center, National Taiwan University and Hospital, Hsinchu City, Taiwan.
| | | |
Collapse
|
40
|
Yoshida K, Maeda K, Sugiyama Y. Hepatic and Intestinal Drug Transporters: Prediction of Pharmacokinetic Effects Caused by Drug-Drug Interactions and Genetic Polymorphisms. Annu Rev Pharmacol Toxicol 2013; 53:581-612. [DOI: 10.1146/annurev-pharmtox-011112-140309] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Kenta Yoshida
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; ,
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; ,
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Research Cluster for Innovation, Yokohama 230-0045, Japan;
| |
Collapse
|