Elsevier

Toxicology in Vitro

Volume 28, Issue 1, February 2014, Pages 104-112
Toxicology in Vitro

3D organotypic HepaRG cultures as in vitro model for acute and repeated dose toxicity studies

https://doi.org/10.1016/j.tiv.2013.06.024Get rights and content

Highlights

  • A simple high throughput method to produce 3D microtissues.

  • 3D HepaRG cultures more sensitive to drug effects than 2D cultures.

  • Simple measurements of glucose and lactate indicate cell stress.

  • Repeated dose toxicity assessment possible.

  • An excellent in vitro alternative method of human relevance.

Abstract

Predictive in vitro models alternative to in vivo animal will have a significant impact in toxicology. Conventional 2D models do not reflect the complexity of a 3D organ resulting in discrepancies between experimental in vitro and in vivo data. Using 3D HepaRG organotypic cultures we tested four drugs (aflatoxin B1, amiodarone, valproic acid and chlorpromazine) for toxic effects and compared the results with 2D HepaRG and HepG2 cultures. We show that 3D HepaRG cultures are more sensitive than the other tested cultures to aflatoxin B1 which is only toxic upon metabolic activation in the liver. We observed that CYP3A4 activity is higher in the 3D HepaRG cultures compared to the 2D HepaRG cultures. Furthermore, we investigated repeated dose toxicity of chlorpromazine and assessed its effects on glucose and lactate metabolism. Sub-toxic concentrations of chlorpromazine induced significant metabolic changes in both 2D and 3D HepaRG cultures upon acute and repeated dose (3 doses) exposure. In summary, our data support the hypothesis that 3D cell culture models better mimic the in vivo tissue and improve cellular functionality. The 3D HepaRG organotypic cultures represent a high throughput system for drug toxicity screening. This system is therefore a promising tool in preclinical testing of human relevance which can allow reducing and/or replacing animal testing for drug adverse effects.

Introduction

Toxicity is one of the major reasons for attrition during drug development and drug withdrawal from the market (Schuster et al., 2005). The main sites of drug toxicity are the cardiovascular system and the liver (Guengerich, 2011). Suitable in vitro test systems as well as predictive test methods are urgently needed (Mandenius et al., 2011a, Mandenius et al., 2011b). An accurate in vitro model for toxicity studies should identify affected pathways and mechanisms and must reflect the in vivo situation as closely as possible. The system should also allow long term functional cultivation of the cells. For liver, primary human hepatocytes (PHH) are still considered as the best available in vitro model for toxicity and metabolism studies (Gomez-Lechon et al., 2003). After isolation, PHH show high expression levels of liver-specific genes which are important for drug metabolism (Guillouzo, 1998). However, they dedifferentiate rapidly in standard cultures (Godoy et al., 2009) and cannot be applied to long term studies. Moreover, the availability of fresh PHH is highly limited in contrast to established cell lines.

The HepaRG cell line shows very promising characteristics such as high CYP450 activities (Aninat et al., 2006, Gripon et al., 2002, Guillouzo et al., 2007). When differentiated, this cell line consists of hepatocyte-like cells and biliary cells in a ratio of about 1:1. The hepatocyte-like cells express both phase I and phase II enzymes and hepatic membrane transporters, indicating that this cell line could serve as a surrogate for primary hepatocytes (Aninat et al., 2006, Kanebratt and Andersson, 2008, Le Vee et al., 2006, Lubberstedt et al., 2011).

Drug toxicity studies show that this cell line is more sensitive to metabolism-mediated toxicity than the HepG2 cell line (Guillouzo et al., 2007). It was also shown that acetaminophen-induced toxic mechanisms in HepaRG cells are similar to animal models and humans, indicating that this cell line is a good in vitro model to study hepatotoxicity of acetaminophen and other drugs (Gunness et al., 2013, McGill et al., 2011).

HepaRG cells can be maintained for weeks with high liver-specific activities in conventional 2D cultures (Aninat et al., 2006). However, these cultures do not reflect the complex, three-dimensional architecture of the liver, where cell–cell and cell-matrix interactions as well as cellular and spatial polarity highly influence gene expression profiles and therefore functionality (Chaumontet et al., 1998, Haouzi et al., 2005, Kono and Roberts, 1996, Narayanan et al., 2002). Recently, several 3D culture systems were developed to improve performance of liver cells for clinical and research purposes (Gerlach et al., 2003, Mueller et al., 2011b; Sauer et al., 2003, van de Kerkhove et al., 2003).

The HepaRG cell line has already been applied to different 3D cultivation systems. In a 3D bioreactor, HepaRG cells maintained CYP450 activities which could be induced and inhibited similar to in vivo (Darnell et al., 2011, Darnell et al., 2012). In another 3D bioreactor system, HepaRG cells showed high liver functionality and moreover, this bioreactor (BAL; bioartificial liver) increased survival time of rats with acute liver failure (Hoekstra et al., 2011, Nibourg et al., 2012a, Nibourg et al., 2012b). 3D HepaRG aggregates produced in spinner-bioreactors maintained liver-specific functions for many weeks (Leite et al., 2012). Taken together, these 3D bioreactor systems are promising tools in terms of maintaining liver-specific functionality, not only for HepaRG cells, but also for PHH (Mueller et al., 2012, Mueller et al., 2011a). However, most 3D bioreactor types are low-throughput systems not allowing toxicity screenings. A 96 well plate high-throughput system for the production of multicellular spheroids was developed (Kelm and Fussenegger, 2004, Kelm et al., 2003). This scaffold-free, hanging drop technique which is now commercially available can be applied for production of spheroids of different cell types including liver cells. The initial cell number and therefore the size of the spheroid is adjustable in this system (Drewitz et al., 2011, Kelm and Fussenegger, 2004, Kelm et al., 2003). We have already shown that spheroids of the hepatoma cell line HepG2 show improved liver-specific functionality compared to 2D HepG2 cultures, such as the production of albumin, CYP450 induction and activity of hepatic drug transporters (Mueller et al., 2011a). Moreover, by using the same system, 3D spheroids of the HepaRG cell line were produced and characterized in terms of long-term maintenance, functionality and responses to drugs (Gunness et al., 2013). These 3D cultures show higher long-term functionality than 2D cultures (increased production of albumin, urea and glucose). They also maintain high drug transporter (MRP2) and CYP2E1 activities.

In this study, we produced 3D spheroids of HepaRG and HepG2 cells using the hanging drop system and compared them for toxicity of selected drugs. We assessed the acute toxicity (24 h exposure) of four drugs (aflatoxin B1, amiodarone, valproic acid and chlorpromazine) with different mechanisms of toxicity and compared the results to conventional 2D cultures.

We tested Aflatoxin B1, which is a naturally occurring mycotoxin produced by Aspergillus species. This compound is another example of metabolism-mediated toxicity. Aflatoxin B1 is metabolized by different Cytochrome P450 (CYP450) isoforms, mainly by CYP3A4 to reactive intermediates, e.g. a reactive epoxide (McLean and Dutton, 1995). Amiodarone is an anti-arrhythmic drug which causes abnormal liver function in 15–50% of patients. The toxicity of amiodarone does not require metabolic activation (Golli-Bennour et al., 2012, Spaniol et al., 2001).

As fatty acid analogue, valproic acid can also induce steatosis in patients by competitive inhibition of fatty acid metabolism. A wide range of metabolites, generated by CYP450 or other metabolizing enzymes (Eadie et al., 1988, Park et al., 1995) are described. The main urinary metabolite is valproate β-glucuronide which is generated by UDP-glucuronosyltransferase (Argikar and Remmel, 2009). Both the parent compound as well as the metabolites are implicated in valproic-acid induced toxicity (Baillie, 1992).

Chlorpromazine is an anti-psychotic drug which causes cholestasis in vivo (Padda et al., 2012). Again, both the parent compound as well as the metabolites are hepatotoxic, however the exact mechanism of idiosyncratic toxicity is still unknown (Wen and Zhou, 2009). In HepaRG cells, chlorpromazine induces oxidative stress as a major mechanism leading to cholestasis (Antherieu et al., 2012).

We carried out single and repeated dose experiments (3 daily doses, 72 h exposure) with chlorpromazine. The effects of subtoxic chlorpromazine concentrations on the glucose uptake/production and lactate secretion in HepaRG 2D and 3D cultures and on the 3D bile canaliculi network were analyzed as well. Moreover, we show that CYP3A4 activity was significantly higher in the 3D HepaRG spheroids compared to the 2D cultures.

In summary, our results suggest that the 3D HepaRG spheroids have better functionality than HepaRG 2D cultures and therefore are a good model for application in drug toxicity screenings especially for metabolism mediated toxicity. This case study was carried out within the framework of the NOTOX project as one of the six research projects of the SEURAT-1 (Safety Evaluation Ultimately Replacing Animal Testing) initiative jointly funded by EC and Cosmetics Europe. The major focus of SEURAT-1 is to develop in vitro models for long term toxicity assessment. The drugs used in this study were chosen in close collaboration with the SEURAT-1 cluster project namely; TOXBANK.

Section snippets

Cell culture

HepG2 cells, obtained from the German collection of microorganisms and cell cultures (DSMZ, Braunschweig, Germany), were maintained in Williams Medium E supplemented with penicillin/streptomycin (100U/100 μg/ml) and 10% FCS.

Cryopreserved, differentiated HepaRG cells (HPR 116) were obtained from Biopredic International, Saint-Grégoire, France and cultivated according to the company’s instructions. The cells were maintained in an incubator (Memmert GmbH, Schwabach, Germany) at 37 °C, 95% air

Morphology of 2D and 3D HepG2 and HepaRG cultures

The morphology of 2D and 3D HepG2 and HepaRG cultures is shown in Fig. 1. The 2D HepG2 cultures show an epithelial morphology. The 3D HepG2 spheroids increased in size over time and attain a diameter of 500–600 μm at cultivation day 7. 2D HepaRG cultures consist of clusters of hepatocyte like cells and surrounding flat biliary cells. These cells did not proliferate. Bile canaliculi like structure (at cell to cell contacts, white, see Fig. 1c) can be observed. In contrast, it is not possible to

Discussion

In this study, we tested the toxicity of four drugs on 2D and 3D cultures of HepG2 and HepaRG cells in serum free conditions. We performed the drug exposure studies either in conventional 96 well plates (for the 2D cultures) or in the GravityTrap plates, a specifically designed 96 well plate format for harvesting and maintaining the 3D spheroids. The free drug concentration is mainly influenced by evaporation and/or binding to culture plates and proteins especially to albumin. Since we used

Conflict of interest

There are no conflicts of interest whatsoever.

Acknowledgements

The research leading to these results has received funding from the European Community’s Seventh Framework Programme (FP7/2007–2013) under grant agreement No 267038 and Cosmetics Europe within the framework of the “NOTOX” Project of the SEURAT-1 initiative. We thank InSphero AG for technical support and Leon Mujis for excellent technical assistance in confocal microscopy. We would also like to thank the TOXBANK consortium for excellent support on compound selection.

References (66)

  • M. Le Vee et al.

    Functional expression of sinusoidal and canalicular hepatic drug transporters in the differentiated human hepatoma HepaRG cell line

    Eur. J. Pharm. Sci.

    (2006)
  • A.P. Li et al.

    Definition of metabolism-dependent xenobiotic toxicity with co-cultures of human hepatocytes and mouse 3T3 fibroblasts in the novel integrated discrete multiple organ co-culture (IdMOC) experimental system: results with model toxicants aflatoxin B1, cyclophosphamide and tamoxifen

    Chem. Biol. Interact.

    (2012)
  • A. Limonciel et al.

    Lactate is an ideal non-invasive marker for evaluating temporal alterations in cell stress and toxicity in repeat dose testing regimes

    Toxicol. In Vitro

    (2011)
  • M. Lubberstedt et al.

    HepaRG human hepatic cell line utility as a surrogate for primary human hepatocytes in drug metabolism assessment in vitro

    J. Pharmacol. Toxicol. Methods

    (2011)
  • M. McLean et al.

    Cellular interactions and metabolism of aflatoxin: an update

    Pharmacol. Ther.

    (1995)
  • J. Niklas et al.

    Effects of drugs in subtoxic concentrations on the metabolic fluxes in human hepatoma cell line Hep G2

    Toxicol. Appl. Pharmacol.

    (2009)
  • B.K. Park et al.

    The role of cytochrome P450 enzymes in hepatic and extrahepatic human drug toxicity

    Pharmacol. Ther.

    (1995)
  • M. Spaniol et al.

    Toxicity of amiodarone and amiodarone analogues on isolated rat liver mitochondria

    J. Hepatol.

    (2001)
  • B. Wen et al.

    Metabolic activation of the phenothiazine antipsychotics chlorpromazine and thioridazine to electrophilic iminoquinone species in human liver microsomes and recombinant P450s

    Chem. Biol. Interact.

    (2009)
  • C. Aninat et al.

    Expression of cytochromes P450, conjugating enzymes and nuclear receptors in human hepatoma HepaRG cells

    Drug Metab. Dispos.

    (2006)
  • S. Antherieu et al.

    Oxidative stress plays a major role in chlorpromazine-induced cholestasis in human HepaRG cells

    Hepatology

    (2012)
  • U.A. Argikar et al.

    Effect of aging on glucuronidation of valproic acid in human liver microsomes and the role of UDP-glucuronosyltransferase UGT1A4, UGT1A8, and UGT1A10

    Drug Metab. Dispos.

    (2009)
  • T.A. Baillie

    Metabolism of valproate to hepatotoxic intermediates

    Pharm. Weekblad Sci.

    (1992)
  • C. Chaumontet et al.

    The polarized hepatic human/rat hybrid WIF 12-1 and WIF-B cells communicate efficiently in vitro via connexin 32-constituted gap junctions

    Hepatology

    (1998)
  • L.A. Corcuera et al.

    An approach to the toxicity and toxicokinetics of aflatoxin B1 and ochratoxin A after simultaneous oral administration to fasted F344 rats

    Food Chem. Toxicol.

    (2012)
  • M. Darnell et al.

    Cytochrome P450-dependent metabolism in HepaRG cells cultured in a dynamic three-dimensional bioreactor

    Drug Metab. Dispos.

    (2011)
  • M.H. Darnell et al.

    In vitro evaluation of major in vivo drug metabolic pathways using primary human hepatocytes and HepaRG cells in suspension and in a dynamic three-dimensional bioreactor system

    J. Pharm. Exp. Ther.

    (2012)
  • M. Drewitz et al.

    Towards automated production and drug sensitivity testing using scaffold-free spherical tumor microtissues

    Biotechnol. J.

    (2011)
  • M.J. Eadie et al.

    Valproate-associated hepatotoxicity and its biochemical mechanisms

    Med. Toxicol. Adverse Drug Exp.

    (1988)
  • S.J. Fey et al.

    Determination of drug toxicity using 3D spheroids constructed from an immortal human hepatocyte cell line

    Toxicol. Sci.

    (2012)
  • J.C. Gerlach et al.

    Use of primary human liver cells originating from discarded grafts in a bioreactor for liver support therapy and the prospects of culturing adult liver stem cells in bioreactors: a morphologic study

    Transplantation

    (2003)
  • P. Godoy et al.

    Extracellular matrix modulates sensitivity of hepatocytes to fibroblastoid dedifferentiation and transforming growth factor beta-induced apoptosis

    Hepatology

    (2009)
  • M.J. Gomez-Lechon et al.

    Human hepatocytes as a tool for studying toxicity and drug metabolism

    Curr. Drug Metab.

    (2003)
  • Cited by (0)

    View full text