Skip to main content

Use of Liver-Derived Cell Lines for the Study of Drug-Induced Liver Injury

  • Protocol
  • First Online:

Part of the book series: Methods in Pharmacology and Toxicology ((MIPT))

Abstract

In vitro liver-derived cell lines have been used extensively in toxicity testing and related studies as alternatives and complements to primary hepatocytes. Multiple hepatocyte derived cellular carcinoma cell lines, such as HepG2, Huh7, and HepaRG cells, have been established over the years, and they display distinct characteristics regarding the expression and activity levels of drug-metabolizing enzymes and other hepatocyte-specific factors. These cell lines have become useful tools and the models based on cell lines showed promising value for screening risks of drug-induced liver injury (DILI) in the early stage of drug development, although they have deficiencies in metabolism-related investigations. Engineered cell lines, expressing drug-metabolizing enzymes or other hepatic genes either stably or transiently, have partially overcome these limitations. The liver-derived cell lines have contributed significantly to mechanistic studies of DILI, and various underlying signaling pathways and signatures of DILI have been identified. In this chapter, we first introduce the major hepatic lines (e.g., HepG2, Huh7, HepaRG, Hep3B, BC2, THLE, and Fa2N-4 cells), including their origins, characteristics, advantages, and disadvantages for application in toxicity studies. We next depict the development and application of various engineered cell lines. We then discuss the current understanding of major DILI mechanisms and the endpoints for in vitro tests. The chapter is closed with a brief discussion of the challenges and opportunities in the field.

This is a preview of subscription content, log in via an institution.

Buying options

Protocol
USD   49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD   79.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD   99.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD   139.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Learn about institutional subscriptions

Springer Nature is developing a new tool to find and evaluate Protocols. Learn more

References

  1. Olson H, Betton G, Robinson D, Thomas K, Monro A, Kolaja G, Lilly P, Sanders J, Sipes G, Bracken W, Dorato M, Van Deun K, Smith P, Berger B, Heller A (2000) Concordance of the toxicity of pharmaceuticals in humans and in animals. Regul Toxicol Pharmacol 32(1):56–67. https://doi.org/10.1006/rtph.2000.1399

    Article  CAS  PubMed  Google Scholar 

  2. Gebhardt R, Hengstler JG, Muller D, Glockner R, Buenning P, Laube B, Schmelzer E, Ullrich M, Utesch D, Hewitt N, Ringel M, Hilz BR, Bader A, Langsch A, Koose T, Burger HJ, Maas J, Oesch F (2003) New hepatocyte in vitro systems for drug metabolism: metabolic capacity and recommendations for application in basic research and drug development, standard operation procedures. Drug Metab Rev 35(2–3):145–213. https://doi.org/10.1081/DMR-120023684

    Article  CAS  PubMed  Google Scholar 

  3. Guo L, Dial S, Shi L, Branham W, Liu J, Fang JL, Green B, Deng H, Kaput J, Ning B (2011) Similarities and differences in the expression of drug-metabolizing enzymes between human hepatic cell lines and primary human hepatocytes. Drug Metab Dispos 39(3):528–538. https://doi.org/10.1124/dmd.110.035873

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. den Braver-Sewradj SP, den Braver MW, Vermeulen NP, Commandeur JN, Richert L, Vos JC (2016) Inter-donor variability of phase I/phase II metabolism of three reference drugs in cryopreserved primary human hepatocytes in suspension and monolayer. Toxicol In Vitro 33:71–79. https://doi.org/10.1016/j.tiv.2016.02.013

    Article  CAS  Google Scholar 

  5. Aden DP, Fogel A, Plotkin S, Damjanov I, Knowles BB (1979) Controlled synthesis of HBsAg in a differentiated human liver carcinoma-derived cell line. Nature 282(5739):615–616

    Article  CAS  PubMed  Google Scholar 

  6. Knowles BB, Howe CC, Aden DP (1980) Human hepatocellular carcinoma cell lines secrete the major plasma proteins and hepatitis B surface antigen. Science 209(4455):497–499

    Article  CAS  PubMed  Google Scholar 

  7. Berger E, Vega N, Weiss-Gayet M, Geloen A (2015) Gene network analysis of glucose linked signaling pathways and their role in human hepatocellular carcinoma cell growth and survival in HuH7 and HepG2 cell lines. Biomed Res Int 2015:821761. https://doi.org/10.1155/2015/821761

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Costantini S, Di Bernardo G, Cammarota M, Castello G, Colonna G (2013) Gene expression signature of human HepG2 cell line. Gene 518(2):335–345. https://doi.org/10.1016/j.gene.2012.12.106

    Article  CAS  PubMed  Google Scholar 

  9. Wisniewski JR, Vildhede A, Noren A, Artursson P (2016) In-depth quantitative analysis and comparison of the human hepatocyte and hepatoma cell line HepG2 proteomes. J Proteomics 136:234–247. https://doi.org/10.1016/j.jprot.2016.01.016

    Article  CAS  PubMed  Google Scholar 

  10. Marroquin LD, Hynes J, Dykens JA, Jamieson JD, Will Y (2007) Circumventing the Crabtree effect: replacing media glucose with galactose increases susceptibility of HepG2 cells to mitochondrial toxicants. Toxicol Sci 97(2):539–547. https://doi.org/10.1093/toxsci/kfm052

    Article  CAS  PubMed  Google Scholar 

  11. O'Brien PJ, Irwin W, Diaz D, Howard-Cofield E, Krejsa CM, Slaughter MR, Gao B, Kaludercic N, Angeline A, Bernardi P, Brain P, Hougham C (2006) High concordance of drug-induced human hepatotoxicity with in vitro cytotoxicity measured in a novel cell-based model using high content screening. Arch Toxicol 80(9):580–604. https://doi.org/10.1007/s00204-006-0091-3

    Article  CAS  PubMed  Google Scholar 

  12. Noor F, Niklas J, Muller-Vieira U, Heinzle E (2009) An integrated approach to improved toxicity prediction for the safety assessment during preclinical drug development using Hep G2 cells. Toxicol Appl Pharmacol 237(2):221–231. https://doi.org/10.1016/j.taap.2009.03.011

    Article  CAS  PubMed  Google Scholar 

  13. O'Leary KA, Day AJ, Needs PW, Mellon FA, O'Brien NM, Williamson G (2003) Metabolism of quercetin-7- and quercetin-3-glucuronides by an in vitro hepatic model: the role of human beta-glucuronidase, sulfotransferase, catechol-O-methyltransferase and multi-resistant protein 2 (MRP2) in flavonoid metabolism. Biochem Pharmacol 65(3):479–491

    Article  CAS  PubMed  Google Scholar 

  14. Sassa S, Sugita O, Galbraith RA, Kappas A (1987) Drug metabolism by the human hepatoma cell, Hep G2. Biochem Biophys Res Commun 143(1):52–57

    Article  CAS  PubMed  Google Scholar 

  15. Hewitt NJ, Hewitt P (2004) Phase I and II enzyme characterization of two sources of HepG2 cell lines. Xenobiotica 34(3):243–256. https://doi.org/10.1080/00498250310001657568

    Article  CAS  PubMed  Google Scholar 

  16. Brandon EF, Raap CD, Meijerman I, Beijnen JH, Schellens JH (2003) An update on in vitro test methods in human hepatic drug biotransformation research: pros and cons. Toxicol Appl Pharmacol 189(3):233–246

    Article  CAS  PubMed  Google Scholar 

  17. Wilkening S, Stahl F, Bader A (2003) Comparison of primary human hepatocytes and hepatoma cell line Hepg2 with regard to their biotransformation properties. Drug Metab Dispos 31(8):1035–1042. https://doi.org/10.1124/dmd.31.8.1035

    Article  CAS  PubMed  Google Scholar 

  18. Westerink WM, Schoonen WG (2007) Cytochrome P450 enzyme levels in HepG2 cells and cryopreserved primary human hepatocytes and their induction in HepG2 cells. Toxicol In Vitro 21(8):1581–1591. https://doi.org/10.1016/j.tiv.2007.05.014

    Article  CAS  PubMed  Google Scholar 

  19. Gerets HH, Tilmant K, Gerin B, Chanteux H, Depelchin BO, Dhalluin S, Atienzar FA (2012) Characterization of primary human hepatocytes, HepG2 cells, and HepaRG cells at the mRNA level and CYP activity in response to inducers and their predictivity for the detection of human hepatotoxins. Cell Biol Toxicol 28(2):69–87. https://doi.org/10.1007/s10565-011-9208-4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ahlin G, Hilgendorf C, Karlsson J, Szigyarto CA, Uhlen M, Artursson P (2009) Endogenous gene and protein expression of drug-transporting proteins in cell lines routinely used in drug discovery programs. Drug Metab Dispos 37(12):2275–2283. https://doi.org/10.1124/dmd.109.028654

    Article  CAS  PubMed  Google Scholar 

  21. Sison-Young RL, Mitsa D, Jenkins RE, Mottram D, Alexandre E, Richert L, Aerts H, Weaver RJ, Jones RP, Johann E, Hewitt PG, Ingelman-Sundberg M, Goldring CE, Kitteringham NR, Park BK (2015) Comparative proteomic characterization of 4 human liver-derived single cell culture models reveals significant variation in the capacity for drug disposition, bioactivation, and detoxication. Toxicol Sci 147(2):412–424. https://doi.org/10.1093/toxsci/kfv136

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lin J, Schyschka L, Muhl-Benninghaus R, Neumann J, Hao L, Nussler N, Dooley S, Liu L, Stockle U, Nussler AK, Ehnert S (2012) Comparative analysis of phase I and II enzyme activities in 5 hepatic cell lines identifies Huh-7 and HCC-T cells with the highest potential to study drug metabolism. Arch Toxicol 86(1):87–95. https://doi.org/10.1007/s00204-011-0733-y

    Article  CAS  PubMed  Google Scholar 

  23. Doostdar H, Duthie SJ, Burke MD, Melvin WT, Grant MH (1988) The influence of culture medium composition on drug metabolising enzyme activities of the human liver derived Hep G2 cell line. FEBS Lett 241(1–2):15–18

    Article  CAS  PubMed  Google Scholar 

  24. Wilkening S, Bader A (2003) Influence of culture time on the expression of drug-metabolizing enzymes in primary human hepatocytes and hepatoma cell line HepG2. J Biochem Mol Toxicol 17(4):207–213. https://doi.org/10.1002/jbt.10085

    Article  CAS  PubMed  Google Scholar 

  25. Majer BJ, Mersch-Sundermann V, Darroudi F, Laky B, de Wit K, Knasmuller S (2004) Genotoxic effects of dietary and lifestyle related carcinogens in human derived hepatoma (HepG2, Hep3B) cells. Mutat Res 551(1–2):153–166. https://doi.org/10.1016/j.mrfmmm.2004.02.022

    Article  CAS  PubMed  Google Scholar 

  26. Tyakht AV, Ilina EN, Alexeev DG, Ischenko DS, Gorbachev AY, Semashko TA, Larin AK, Selezneva OV, Kostryukova ES, Karalkin PA, Vakhrushev IV, Kurbatov LK, Archakov AI, Govorun VM (2014) RNA-Seq gene expression profiling of HepG2 cells: the influence of experimental factors and comparison with liver tissue. BMC Genomics 15:1108. https://doi.org/10.1186/1471-2164-15-1108

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Sison-Young RL, Lauschke VM, Johann E, Alexandre E, Antherieu S, Aerts H, Gerets HH, Labbe G, Hoet D, Dorau M, Schofield CA, Lovatt CA, Holder JC, Stahl SH, Richert L, Kitteringham NR, Jones RP, Elmasry M, Weaver RJ, Hewitt PG, Ingelman-Sundberg M, Goldring CE, Park BK (2016) A multicenter assessment of single-cell models aligned to standard measures of cell health for prediction of acute hepatotoxicity. Arch Toxicol. https://doi.org/10.1007/s00204-016-1745-4

  28. Choi JM, Oh SJ, Lee SY, Im JH, Oh JM, Ryu CS, Kwak HC, Lee JY, Kang KW, Kim SK (2015) HepG2 cells as an in vitro model for evaluation of cytochrome P450 induction by xenobiotics. Arch Pharm Res 38(5):691–704. https://doi.org/10.1007/s12272-014-0502-6

    Article  CAS  PubMed  Google Scholar 

  29. Nakabayashi H, Taketa K, Miyano K, Yamane T, Sato J (1982) Growth of human hepatoma cells lines with differentiated functions in chemically defined medium. Cancer Res 42(9):3858–3863

    CAS  PubMed  Google Scholar 

  30. Fang C, Yi Z, Liu F, Lan S, Wang J, Lu H, Yang P, Yuan Z (2006) Proteome analysis of human liver carcinoma Huh7 cells harboring hepatitis C virus subgenomic replicon. Proteomics 6(2):519–527. https://doi.org/10.1002/pmic.200500233

    Article  CAS  PubMed  Google Scholar 

  31. Jouan E, Le Vee M, Denizot C, Parmentier Y, Fardel O (2016) Drug transporter expression and activity in human hepatoma HuH-7 cells. Pharmaceutics 9(1). https://doi.org/10.3390/pharmaceutics9010003

  32. Louisa M, Suyatna FD, Wanandi SI, Asih PB, Syafruddin D (2016) Differential expression of several drug transporter genes in HepG2 and Huh-7 cell lines. Adv Biomed Res 5:104. https://doi.org/10.4103/2277-9175.183664

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Sainz B, Jr., Chisari FV (2006) Production of infectious hepatitis C virus by well-differentiated, growth-arrested human hepatoma-derived cells. J Virol 80 (20):10253–10257. https://doi.org/10.1128/JVI.01059-06.

  34. Choi S, Sainz B, Jr., Corcoran P, Uprichard S, Jeong H (2009) Characterization of increased drug metabolism activity in dimethyl sulfoxide (DMSO)-treated Huh7 hepatoma cells. Xenobiotica 39 (3):205–217. doi:https://doi.org/10.1080/00498250802613620.

  35. Liu Y, Flynn TJ, Xia M, Wiesenfeld PL, Ferguson MS (2015) Evaluation of CYP3A4 inhibition and hepatotoxicity using DMSO-treated human hepatoma HuH-7 cells. Cell Biol Toxicol 31(4–5):221–230. https://doi.org/10.1007/s10565-015-9306-9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Sivertsson L, Ek M, Darnell M, Edebert I, Ingelman-Sundberg M, Neve EP (2010) CYP3A4 catalytic activity is induced in confluent Huh7 hepatoma cells. Drug Metab Dispos 38(6):995–1002. https://doi.org/10.1124/dmd.110.032367

    Article  CAS  PubMed  Google Scholar 

  37. Sivertsson L, Edebert I, Palmertz MP, Ingelman-Sundberg M, Neve EP (2013) Induced CYP3A4 expression in confluent Huh7 hepatoma cells as a result of decreased cell proliferation and subsequent pregnane X receptor activation. Mol Pharmacol 83(3):659–670. https://doi.org/10.1124/mol.112.082305

    Article  CAS  PubMed  Google Scholar 

  38. Gripon P, Rumin S, Urban S, Le Seyec J, Glaise D, Cannie I, Guyomard C, Lucas J, Trepo C, Guguen-Guillouzo C (2002) Infection of a human hepatoma cell line by hepatitis B virus. Proc Natl Acad Sci U S A 99 (24):15655–15660. https://doi.org/10.1073/pnas.232137699.

  39. Hart SN, Li Y, Nakamoto K, Subileau EA, Steen D, Zhong XB (2010) A comparison of whole genome gene expression profiles of HepaRG cells and HepG2 cells to primary human hepatocytes and human liver tissues. Drug Metab Dispos 38(6):988–994. https://doi.org/10.1124/dmd.109.031831

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Antherieu S, Chesne C, Li R, Guguen-Guillouzo C, Guillouzo A (2012) Optimization of the HepaRG cell model for drug metabolism and toxicity studies. Toxicol In Vitro 26(8):1278–1285. https://doi.org/10.1016/j.tiv.2012.05.008

    Article  CAS  PubMed  Google Scholar 

  41. Parent R, Marion MJ, Furio L, Trepo C, Petit MA (2004) Origin and characterization of a human bipotent liver progenitor cell line. Gastroenterology 126(4):1147–1156

    Article  PubMed  Google Scholar 

  42. Cerec V, Glaise D, Garnier D, Morosan S, Turlin B, Drenou B, Gripon P, Kremsdorf D, Guguen-Guillouzo C, Corlu A (2007) Transdifferentiation of hepatocyte-like cells from the human hepatoma HepaRG cell line through bipotent progenitor. Hepatology 45(4):957–967. https://doi.org/10.1002/hep.21536

    Article  CAS  PubMed  Google Scholar 

  43. Jennen DG, Magkoufopoulou C, Ketelslegers HB, van Herwijnen MH, Kleinjans JC, van Delft JH (2010) Comparison of HepG2 and HepaRG by whole-genome gene expression analysis for the purpose of chemical hazard identification. Toxicol Sci 115 (1):66–79. doi:https://doi.org/10.1093/toxsci/kfq026.

  44. Aninat C, Piton A, Glaise D, Le Charpentier T, Langouet S, Morel F, Guguen-Guillouzo C, Guillouzo A (2006) Expression of cytochromes P450, conjugating enzymes and nuclear receptors in human hepatoma HepaRG cells. Drug Metab Dispos 34 (1):75–83. doi:https://doi.org/10.1124/dmd.105.006759.

  45. Josse R, Aninat C, Glaise D, Dumont J, Fessard V, Morel F, Poul JM, Guguen-Guillouzo C, Guillouzo A (2008) Long-term functional stability of human HepaRG hepatocytes and use for chronic toxicity and genotoxicity studies. Drug Metab Dispos 36(6):1111–1118. https://doi.org/10.1124/dmd.107.019901

    Article  CAS  PubMed  Google Scholar 

  46. Antherieu S, Chesne C, Li R, Camus S, Lahoz A, Picazo L, Turpeinen M, Tolonen A, Uusitalo J, Guguen-Guillouzo C, Guillouzo A (2010) Stable expression, activity, and inducibility of cytochromes P450 in differentiated HepaRG cells. Drug Metab Dispos 38(3):516–525. https://doi.org/10.1124/dmd.109.030197

    Article  CAS  PubMed  Google Scholar 

  47. Le Vee M, Jigorel E, Glaise D, Gripon P, Guguen-Guillouzo C, Fardel O (2006) Functional expression of sinusoidal and canalicular hepatic drug transporters in the differentiated human hepatoma HepaRG cell line. Eur J Pharm Sci 28(1–2):109–117. https://doi.org/10.1016/j.ejps.2006.01.004

    Article  CAS  PubMed  Google Scholar 

  48. Le Vee M, Noel G, Jouan E, Stieger B, Fardel O (2013) Polarized expression of drug transporters in differentiated human hepatoma HepaRG cells. Toxicol In Vitro 27(6):1979–1986. https://doi.org/10.1016/j.tiv.2013.07.003

    Article  CAS  PubMed  Google Scholar 

  49. Klein S, Mueller D, Schevchenko V, Noor F (2014) Long-term maintenance of HepaRG cells in serum-free conditions and application in a repeated dose study. J Appl Toxicol 34(10):1078–1086. https://doi.org/10.1002/jat.2929

    Article  CAS  PubMed  Google Scholar 

  50. Lubberstedt M, Muller-Vieira U, Mayer M, Biemel KM, Knospel F, Knobeloch D, Nussler AK, Gerlach JC, Zeilinger K (2011) HepaRG human hepatic cell line utility as a surrogate for primary human hepatocytes in drug metabolism assessment in vitro. J Pharmacol Toxicol Methods 63(1):59–68. https://doi.org/10.1016/j.vascn.2010.04.013

    Article  CAS  PubMed  Google Scholar 

  51. Lambert CB, Spire C, Renaud MP, Claude N, Guillouzo A (2009) Reproducible chemical-induced changes in gene expression profiles in human hepatoma HepaRG cells under various experimental conditions. Toxicol In Vitro 23(3):466–475. https://doi.org/10.1016/j.tiv.2008.12.018

    Article  CAS  PubMed  Google Scholar 

  52. Tomida T, Okamura H, Satsukawa M, Yokoi T, Konno Y (2015) Multiparametric assay using HepaRG cells for predicting drug-induced liver injury. Toxicol Lett 236(1):16–24. https://doi.org/10.1016/j.toxlet.2015.04.014

    Article  CAS  PubMed  Google Scholar 

  53. Saito J, Okamura A, Takeuchi K, Hanioka K, Okada A, Ohata T (2016) High content analysis assay for prediction of human hepatotoxicity in HepaRG and HepG2 cells. Toxicol In Vitro 33:63–70. https://doi.org/10.1016/j.tiv.2016.02.019

    Article  CAS  PubMed  Google Scholar 

  54. Wu Y, Geng XC, Wang JF, Miao YF, Lu YL, Li B (2016) The HepaRG cell line, a superior in vitro model to L-02, HepG2 and hiHeps cell lines for assessing drug-induced liver injury. Cell Biol Toxicol 32(1):37–59. https://doi.org/10.1007/s10565-016-9316-2

    Article  CAS  PubMed  Google Scholar 

  55. Kanebratt KP, Andersson TB (2008) HepaRG cells as an in vitro model for evaluation of cytochrome P450 induction in humans. Drug Metab Dispos 36(1):137–145. https://doi.org/10.1124/dmd.107.017418

    Article  CAS  PubMed  Google Scholar 

  56. Kaneko A, Kato M, Sekiguchi N, Mitsui T, Takeda K, Aso Y (2009) In vitro model for the prediction of clinical CYP3A4 induction using HepaRG cells. Xenobiotica 39(11):803–810. https://doi.org/10.3109/00498250903184018

    Article  CAS  PubMed  Google Scholar 

  57. Ferreira A, Rodrigues M, Silvestre S, Falcao A, Alves G (2014) HepaRG cell line as an in vitro model for screening drug-drug interactions mediated by metabolic induction: amiodarone used as a model substance. Toxicol In Vitro 28(8):1531–1535. https://doi.org/10.1016/j.tiv.2014.08.004

    Article  CAS  PubMed  Google Scholar 

  58. Szabo M, Veres Z, Baranyai Z, Jakab F, Jemnitz K (2013) 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 8(3):e59432. https://doi.org/10.1371/journal.pone.0059432

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Bachour-El Azzi P, Sharanek A, Burban A, Li R, Guevel RL, Abdel-Razzak Z, Stieger B, Guguen-Guillouzo C, Guillouzo A (2015) Comparative localization and functional activity of the main hepatobiliary transporters in HepaRG cells and primary human hepatocytes. Toxicol Sci 145(1):157–168. https://doi.org/10.1093/toxsci/kfv041

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Jackson JP, Li L, Chamberlain ED, Wang H, Ferguson SS (2016) Contextualizing hepatocyte functionality of cryopreserved HepaRG cell cultures. Drug Metab Dispos 44(9):1463–1479. https://doi.org/10.1124/dmd.116.069831

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Glaise D, Ilyin GP, Loyer P, Cariou S, Bilodeau M, Lucas J, Puisieux A, Ozturk M, Guguen-Guillouzo C (1998) Cell cycle gene regulation in reversibly differentiated new human hepatoma cell lines. Cell Growth Differ 9(2):165–176

    CAS  PubMed  Google Scholar 

  62. Gomez-Lechon MJ, Donato T, Jover R, Rodriguez C, Ponsoda X, Glaise D, Castell JV, Guguen-Guillouzo C (2001) Expression and induction of a large set of drug-metabolizing enzymes by the highly differentiated human hepatoma cell line BC2. Eur J Biochem 268(5):1448–1459

    Article  CAS  PubMed  Google Scholar 

  63. Donato MT, Lahoz A, Castell JV, Gomez-Lechon MJ (2008) Cell lines: a tool for in vitro drug metabolism studies. Curr Drug Metab 9(1):1–11

    Article  CAS  PubMed  Google Scholar 

  64. O'Connor JE, Martinez A, Castell JV, Gomez-Lechon MJ (2005) Multiparametric characterization by flow cytometry of flow-sorted subpopulations of a human hepatoma cell line useful for drug research. Cytometry A 63(1):48–58. https://doi.org/10.1002/cyto.a.20095

    Article  CAS  PubMed  Google Scholar 

  65. Fabre N, Arrivet E, Trancard J, Bichet N, Roome NO, Prenez A, Vericat JA (2003) A new hepatoma cell line for toxicity testing at repeated doses. Cell Biol Toxicol 19(2):71–82

    Article  CAS  PubMed  Google Scholar 

  66. Pfeifer AM, Cole KE, Smoot DT, Weston A, Groopman JD, Shields PG, Vignaud JM, Juillerat M, Lipsky MM, Trump BF et al (1993) Simian virus 40 large tumor antigen-immortalized normal human liver epithelial cells express hepatocyte characteristics and metabolize chemical carcinogens. Proc Natl Acad Sci U S A 90(11):5123–5127

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Mace K, Aguilar F, Wang JS, Vautravers P, Gomez-Lechon M, Gonzalez FJ, Groopman J, Harris CC, Pfeifer AM (1997) Aflatoxin B1-induced DNA adduct formation and p53 mutations in CYP450-expressing human liver cell lines. Carcinogenesis 18(7):1291–1297

    Article  CAS  PubMed  Google Scholar 

  68. Bort R, Castell JV, Pfeifer A, Gomez-Lechon MJ, Mace K (1999) High expression of human CYP2C in immortalized human liver epithelial cells. Toxicol In Vitro 13(4–5):633–638

    Article  CAS  PubMed  Google Scholar 

  69. Soltanpour Y, Hilgendorf C, Ahlstrom MM, Foster AJ, Kenna JG, Petersen A, Ungell AL (2012) Characterization of THLE-cytochrome P450 (P450) cell lines: gene expression background and relationship to P450-enzyme activity. Drug Metab Dispos 40(11):2054–2058. https://doi.org/10.1124/dmd.112.045815

    Article  CAS  PubMed  Google Scholar 

  70. Mills JB, Rose KA, Sadagopan N, Sahi J, de Morais SM (2004) Induction of drug metabolism enzymes and MDR1 using a novel human hepatocyte cell line. J Pharmacol Exp Ther 309(1):303–309. https://doi.org/10.1124/jpet.103.061713

    Article  CAS  PubMed  Google Scholar 

  71. Hariparsad N, Carr BA, Evers R, Chu X (2008) Comparison of immortalized Fa2N-4 cells and human hepatocytes as in vitro models for cytochrome P450 induction. Drug Metab Dispos 36(6):1046–1055. https://doi.org/10.1124/dmd.108.020677

    Article  CAS  PubMed  Google Scholar 

  72. Ripp SL, Mills JB, Fahmi OA, Trevena KA, Liras JL, Maurer TS, de Morais SM (2006) Use of immortalized human hepatocytes to predict the magnitude of clinical drug-drug interactions caused by CYP3A4 induction. Drug Metab Dispos 34(10):1742–1748. https://doi.org/10.1124/dmd.106.010132

    Article  CAS  PubMed  Google Scholar 

  73. Molden E, Asberg A, Christensen H (2000) CYP2D6 is involved in O-demethylation of diltiazem. An in vitro study with transfected human liver cells. Eur J Clin Pharmacol 56(8):575–579

    Article  CAS  PubMed  Google Scholar 

  74. Barcelo S, Mace K, Pfeifer AM, Chipman JK (1998) Production of DNA strand breaks by N-nitrosodimethylamine and 2-amino-3-methylimidazo[4,5-f]quinoline in THLE cells expressing human CYP isoenzymes and inhibition by sulforaphane. Mutat Res 402(1–2):111–120

    Article  CAS  PubMed  Google Scholar 

  75. Dambach DM, Andrews BA, Moulin F (2005) New technologies and screening strategies for hepatotoxicity: use of in vitro models. Toxicol Pathol 33(1):17–26. https://doi.org/10.1080/01926230590522284

    Article  CAS  PubMed  Google Scholar 

  76. Vignati L, Turlizzi E, Monaci S, Grossi P, Kanter R, Monshouwer M (2005) An in vitro approach to detect metabolite toxicity due to CYP3A4-dependent bioactivation of xenobiotics. Toxicology 216(2–3):154–167. https://doi.org/10.1016/j.tox.2005.08.003

    Article  CAS  PubMed  Google Scholar 

  77. Thompson RA, Isin EM, Li Y, Weidolf L, Page K, Wilson I, Swallow S, Middleton B, Stahl S, Foster AJ, Dolgos H, Weaver R, Kenna JG (2012) In vitro approach to assess the potential for risk of idiosyncratic adverse reactions caused by candidate drugs. Chem Res Toxicol 25(8):1616–1632. https://doi.org/10.1021/tx300091x

    Article  CAS  PubMed  Google Scholar 

  78. Gustafsson F, Foster AJ, Sarda S, Bridgland-Taylor MH, Kenna JG (2014) A correlation between the in vitro drug toxicity of drugs to cell lines that express human P450s and their propensity to cause liver injury in humans. Toxicol Sci 137(1):189–211. https://doi.org/10.1093/toxsci/kft223

    Article  CAS  PubMed  Google Scholar 

  79. Yoshitomi S, Ikemoto K, Takahashi J, Miki H, Namba M, Asahi S (2001) Establishment of the transformants expressing human cytochrome P450 subtypes in HepG2, and their applications on drug metabolism and toxicology. Toxicol In Vitro 15(3):245–256

    Article  CAS  PubMed  Google Scholar 

  80. Xuan J, Chen S, Ning B, Tolleson WH, Guo L (2015) Development of HepG2-derived cells expressing cytochrome P450s for assessing metabolism-associated drug-induced liver toxicity. Chem Biol Interact. https://doi.org/10.1016/j.cbi.2015.10.009

  81. Wu Q, Ning B, Xuan J, Ren Z, Guo L, Bryant MS (2016) The role of CYP 3A4 and 1A1 in amiodarone-induced hepatocellular toxicity. Toxicol Lett 253:55–62. https://doi.org/10.1016/j.toxlet.2016.04.016

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Gomez-Lechon MJ, Tolosa L, Donato MT (2017) Upgrading HepG2 cells with adenoviral vectors that encode drug-metabolizing enzymes: application for drug hepatotoxicity testing. Expert Opin Drug Metab Toxicol 13(2):137–148. https://doi.org/10.1080/17425255.2017.1238459

    Article  CAS  PubMed  Google Scholar 

  83. Hashizume T, Yoshitomi S, Asahi S, Uematsu R, Matsumura S, Chatani F, Oda H (2010) Advantages of human hepatocyte-derived transformants expressing a series of human cytochrome p450 isoforms for genotoxicity examination. Toxicol Sci 116(2):488–497. https://doi.org/10.1093/toxsci/kfq154

    Article  CAS  PubMed  Google Scholar 

  84. Wu Y, Chitranshi P, Loukotkova L, Gamboa da Costa G, Beland FA, Zhang J, Fang JL (2016) Cytochrome P450-mediated metabolism of triclosan attenuates its cytotoxicity in hepatic cells. Arch Toxicol. https://doi.org/10.1007/s00204-016-1893-6

  85. Kublbeck J, Reinisalo M, Mustonen R, Honkakoski P (2010) Up-regulation of CYP expression in hepatoma cells stably transfected by chimeric nuclear receptors. Eur J Pharm Sci 40(4):263–272. https://doi.org/10.1016/j.ejps.2010.03.022

    Article  CAS  PubMed  Google Scholar 

  86. Naiki T, Nagaki M, Shidoji Y, Kojima H, Moriwaki H (2004) Functional activity of human hepatoma cells transfected with adenovirus-mediated hepatocyte nuclear factor (HNF)-4 gene. Cell Transplant 13(4):393–403

    Article  PubMed  Google Scholar 

  87. Tolosa L, Donato MT, Perez-Cataldo G, Castell JV, Gomez-Lechon MJ (2012) Upgrading cytochrome P450 activity in HepG2 cells co-transfected with adenoviral vectors for drug hepatotoxicity assessment. Toxicol In Vitro 26(8):1272–1277. https://doi.org/10.1016/j.tiv.2011.11.008

    Article  CAS  PubMed  Google Scholar 

  88. Tolosa L, Gomez-Lechon MJ, Perez-Cataldo G, Castell JV, Donato MT (2013) HepG2 cells simultaneously expressing five P450 enzymes for the screening of hepatotoxicity: identification of bioactivable drugs and the potential mechanism of toxicity involved. Arch Toxicol 87(6):1115–1127. https://doi.org/10.1007/s00204-013-1012-x

    Article  CAS  PubMed  Google Scholar 

  89. Sawada M, Kamataki T (1998) Genetically engineered cells stably expressing cytochrome P450 and their application to mutagen assays. Mutat Res 411(1):19–43

    Article  CAS  PubMed  Google Scholar 

  90. Goldring CE, Kitteringham NR, Jenkins R, Lovatt CA, Randle LE, Abdullah A, Owen A, Liu X, Butler PJ, Williams DP, Metcalfe P, Berens C, Hillen W, Foster B, Simpson A, McLellan L, Park BK (2006) Development of a transactivator in hepatoma cells that allows expression of phase I, phase II, and chemical defense genes. Am J Physiol Cell Physiol 290(1):C104–C115. https://doi.org/10.1152/ajpcell.00133.2005

    Article  CAS  PubMed  Google Scholar 

  91. van der Mark VA, Rudi de Waart D, Shevchenko V, Elferink RP, Chamuleau RA, Hoekstra R (2017) 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 45 (1):56–67. doi:https://doi.org/10.1124/dmd.116.072603.

  92. Chu CC, Pan KL, Yao HT, Hsu JT (2011) Development of a whole-cell screening system for evaluation of the human CYP1A2-mediated metabolism. Biotechnol Bioeng 108(12):2932–2940. https://doi.org/10.1002/bit.23256

    Article  CAS  PubMed  Google Scholar 

  93. Sharanek A, Azzi PB, Al-Attrache H, Savary CC, Humbert L, Rainteau D, Guguen-Guillouzo C, Guillouzo A (2014) Different dose-dependent mechanisms are involved in early cyclosporine a-induced cholestatic effects in hepaRG cells. Toxicol Sci 141(1):244–253. https://doi.org/10.1093/toxsci/kfu122

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Xu JJ, Henstock PV, Dunn MC, Smith AR, Chabot JR, de Graaf D (2008) Cellular imaging predictions of clinical drug-induced liver injury. Toxicol Sci 105 (1):97–105. doi:https://doi.org/10.1093/toxsci/kfn109.

  95. Atienzar FA, Blomme EA, Chen M, Hewitt P, Kenna JG, Labbe G, Moulin F, Pognan F, Roth AB, Suter-Dick L, Ukairo O, Weaver RJ, Will Y, Dambach DM (2016) Key challenges and opportunities associated with the use of in vitro models to detect human DILI: integrated risk assessment and mitigation plans. Biomed Res Int 2016:9737920. https://doi.org/10.1155/2016/9737920

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Nikoletopoulou V, Markaki M, Palikaras K, Tavernarakis N (2013) Crosstalk between apoptosis, necrosis and autophagy. Biochim Biophys Acta 1833(12):3448–3459. https://doi.org/10.1016/j.bbamcr.2013.06.001

    Article  CAS  PubMed  Google Scholar 

  97. Senft D, Ronai ZA (2015) UPR, autophagy, and mitochondria crosstalk underlies the ER stress response. Trends Biochem Sci 40(3):141–148. https://doi.org/10.1016/j.tibs.2015.01.002

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Nagiah S, Phulukdaree A, Chuturgoon A (2015) Mitochondrial and oxidative stress response in HepG2 cells following acute and prolonged exposure to antiretroviral drugs. J Cell Biochem 116(9):1939–1946. https://doi.org/10.1002/jcb.25149

    Article  CAS  PubMed  Google Scholar 

  99. Li X, Wang Y, Wang H, Huang C, Huang Y, Li J (2015) Endoplasmic reticulum stress is the crossroads of autophagy, inflammation, and apoptosis signaling pathways and participates in liver fibrosis. Inflamm Res 64(1):1–7. https://doi.org/10.1007/s00011-014-0772-y

    Article  CAS  PubMed  Google Scholar 

  100. Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, Walczak H, Vandenabeele P (2014) Regulated necrosis: the expanding network of non-apoptotic cell death pathways. Nat Rev Mol Cell Biol 15(2):135–147. https://doi.org/10.1038/nrm3737

    Article  CAS  Google Scholar 

  101. Krysko DV, Vanden Berghe T, D'Herde K, Vandenabeele P (2008) Apoptosis and necrosis: detection, discrimination and phagocytosis. Methods 44(3):205–221. https://doi.org/10.1016/j.ymeth.2007.12.001

    Article  CAS  PubMed  Google Scholar 

  102. Hinson JA, Roberts DW, James LP (2010) Mechanisms of acetaminophen-induced liver necrosis. Handb Exp Pharmacol 196:369–405. https://doi.org/10.1007/978-3-642-00663-0_12

    Article  CAS  Google Scholar 

  103. Jaeschke H, McGill MR, Ramachandran A (2012) Oxidant stress, mitochondria, and cell death mechanisms in drug-induced liver injury: lessons learned from acetaminophen hepatotoxicity. Drug Metab Rev 44(1):88–106. https://doi.org/10.3109/03602532.2011.602688

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Lin T, Yang MS (2008) Benzo[a]pyrene-induced necrosis in the HepG(2) cells via PARP-1 activation and NAD(+) depletion. Toxicology 245(1–2):147–153. https://doi.org/10.1016/j.tox.2007.12.020

    Article  CAS  PubMed  Google Scholar 

  105. Golstein P, Kroemer G (2007) Cell death by necrosis: towards a molecular definition. Trends Biochem Sci 32(1):37–43. https://doi.org/10.1016/j.tibs.2006.11.001

    Article  CAS  PubMed  Google Scholar 

  106. Danial NN, Korsmeyer SJ (2004) Cell death: critical control points. Cell 116(2):205–219

    Article  CAS  PubMed  Google Scholar 

  107. Taylor RC, Cullen SP, Martin SJ (2008) Apoptosis: controlled demolition at the cellular level. Nat Rev Mol Cell Biol 9(3):231–241. https://doi.org/10.1038/nrm2312

    Article  CAS  PubMed  Google Scholar 

  108. Kroemer G, Galluzzi L, Brenner C (2007) Mitochondrial membrane permeabilization in cell death. Physiol Rev 87(1):99–163. https://doi.org/10.1152/physrev.00013.2006

    Article  CAS  PubMed  Google Scholar 

  109. Chao DT, Korsmeyer SJ (1998) BCL-2 family: regulators of cell death. Annu Rev Immunol 16:395–419. https://doi.org/10.1146/annurev.immunol.16.1.395

    Article  CAS  PubMed  Google Scholar 

  110. Zimmermann KC, Green DR (2001) How cells die: apoptosis pathways. J Allergy Clin Immunol 108(4 Suppl):S99–103

    Article  CAS  PubMed  Google Scholar 

  111. Degterev A, Boyce M, Yuan J (2003) A decade of caspases. Oncogene 22(53):8543–8567. https://doi.org/10.1038/sj.onc.1207107

    Article  CAS  PubMed  Google Scholar 

  112. Dykens JA, Jamieson JD, Marroquin LD, Nadanaciva S, Xu JJ, Dunn MC, Smith AR, Will Y (2008) In vitro assessment of mitochondrial dysfunction and cytotoxicity of nefazodone, trazodone, and buspirone. Toxicol Sci 103(2):335–345. https://doi.org/10.1093/toxsci/kfn056

    Article  CAS  PubMed  Google Scholar 

  113. Labbe G, Pessayre D, Fromenty B (2008) Drug-induced liver injury through mitochondrial dysfunction: mechanisms and detection during preclinical safety studies. Fundam Clin Pharmacol 22(4):335–353. https://doi.org/10.1111/j.1472-8206.2008.00608.x

    Article  CAS  PubMed  Google Scholar 

  114. Will Y, Dykens J (2014) Mitochondrial toxicity assessment in industry—a decade of technology development and insight. Expert Opin Drug Metab Toxicol 10(8):1061–1067. https://doi.org/10.1517/17425255.2014.939628

    Article  PubMed  Google Scholar 

  115. Brand MD, Nicholls DG (2011) Assessing mitochondrial dysfunction in cells. Biochem J 435(2):297–312. https://doi.org/10.1042/BJ20110162

    Article  CAS  PubMed  Google Scholar 

  116. Li Y, Couch L, Higuchi M, Fang JL, Guo L (2012) Mitochondrial dysfunction induced by sertraline, an antidepressant agent. Toxicol Sci 127(2):582–591. https://doi.org/10.1093/toxsci/kfs100

    Article  CAS  PubMed  Google Scholar 

  117. Tirmenstein MA, Hu CX, Gales TL, Maleeff BE, Narayanan PK, Kurali E, Hart TK, Thomas HC, Schwartz LW (2002) Effects of troglitazone on HepG2 viability and mitochondrial function. Toxicol Sci 69(1):131–138

    Article  CAS  PubMed  Google Scholar 

  118. Kamalian L, Chadwick AE, Bayliss M, French NS, Monshouwer M, Snoeys J, Park BK (2015) The utility of HepG2 cells to identify direct mitochondrial dysfunction in the absence of cell death. Toxicol In Vitro 29(4):732–740. https://doi.org/10.1016/j.tiv.2015.02.011

    Article  CAS  PubMed  Google Scholar 

  119. Wang C, Youle RJ (2009) The role of mitochondria in apoptosis. Annu Rev Genet 43:95–118. https://doi.org/10.1146/annurev-genet-102108-134850

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Hatok J, Racay P (2016) Bcl-2 family proteins: master regulators of cell survival. Biomol Concepts 7(4):259–270. https://doi.org/10.1515/bmc-2016-0015

    Article  CAS  PubMed  Google Scholar 

  121. Videla LA (2009) Oxidative stress signaling underlying liver disease and hepatoprotective mechanisms. World J Hepatol 1(1):72–78. https://doi.org/10.4254/wjh.v1.i1.72

    Article  PubMed  PubMed Central  Google Scholar 

  122. Pereira CV, Nadanaciva S, Oliveira PJ, Will Y (2012) The contribution of oxidative stress to drug-induced organ toxicity and its detection in vitro and in vivo. Expert Opin Drug Metab Toxicol 8(2):219–237. https://doi.org/10.1517/17425255.2012.645536

    Article  CAS  PubMed  Google Scholar 

  123. Wang X, Fang H, Huang Z, Shang W, Hou T, Cheng A, Cheng H (2013) Imaging ROS signaling in cells and animals. J Mol Med (Berl) 91(8):917–927. https://doi.org/10.1007/s00109-013-1067-4

    Article  CAS  Google Scholar 

  124. Chen S, Zhang Z, Qing T, Ren Z, Yu D, Couch L, Ning B, Mei N, Shi L, Tolleson WH, Guo L (2016) Activation of the Nrf2 signaling pathway in usnic acid-induced toxicity in HepG2 cells. Arch Toxicol. https://doi.org/10.1007/s00204-016-1775-y

  125. Raza H, John A (2012) Streptozotocin-induced cytotoxicity, oxidative stress and mitochondrial dysfunction in human hepatoma HepG2 cells. Int J Mol Sci 13(5):5751–5767. https://doi.org/10.3390/ijms13055751

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Gao Y, Chu S, Zhang Z, Zuo W, Xia C, Ai Q, Luo P, Cao P, Chen N (2016) Early stage functions of mitochondrial autophagy and oxidative stress in acetaminophen-induced liver injury. J Cell Biochem. https://doi.org/10.1002/jcb.25788

  127. Antherieu S, Bachour-El Azzi P, Dumont J, Abdel-Razzak Z, Guguen-Guillouzo C, Fromenty B, Robin MA, Guillouzo A (2013) Oxidative stress plays a major role in chlorpromazine-induced cholestasis in human HepaRG cells. Hepatology 57(4):1518–1529. https://doi.org/10.1002/hep.26160

    Article  CAS  PubMed  Google Scholar 

  128. Nguyen T, Nioi P, Pickett CB (2009) The Nrf2-antioxidant response element signaling pathway and its activation by oxidative stress. J Biol Chem 284(20):13291–13295. https://doi.org/10.1074/jbc.R900010200

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Kobayashi A, Kang MI, Okawa H, Ohtsuji M, Zenke Y, Chiba T, Igarashi K, Yamamoto M (2004) Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Mol Cell Biol 24(16):7130–7139. https://doi.org/10.1128/MCB.24.16.7130-7139.2004

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Chen S, Melchior WB, Jr., Guo L (2014) Endoplasmic reticulum stress in drug- and environmental toxicant-induced liver toxicity. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev 32 (1):83–104. doi:https://doi.org/10.1080/10590501.2014.881648.

  131. Iurlaro R, Munoz-Pinedo C (2016) Cell death induced by endoplasmic reticulum stress. FEBS J 283(14):2640–2652. https://doi.org/10.1111/febs.13598

    Article  CAS  PubMed  Google Scholar 

  132. Samali A, Fitzgerald U, Deegan S, Gupta S (2010) Methods for monitoring endoplasmic reticulum stress and the unfolded protein response. Int J Cell Biol 2010:830307. https://doi.org/10.1155/2010/830307

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Oslowski C, Urano F (2011) Measuring ER stress and the unfolded protein response using mammalian tissue culture system. Methods Enzymol 490:71–92. https://doi.org/10.1016/B978-0-12-385114-7.00004-0

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Chen S, Xuan J, Couch L, Iyer A, Wu Y, Li QZ, Guo L (2014) Sertraline induces endoplasmic reticulum stress in hepatic cells. Toxicology 322:78–88. https://doi.org/10.1016/j.tox.2014.05.007

    Article  CAS  PubMed  Google Scholar 

  135. Ren Z, Chen S, Zhang J, Doshi U, Li AP, Guo L (2016) Endoplasmic reticulum stress induction and ERK1/2 activation contribute to nefazodone-induced toxicity in hepatic cells. Toxicol Sci. https://doi.org/10.1093/toxsci/kfw173

  136. Chen S, Zhang Z, Wu Y, Shi Q, Yan H, Mei N, Tolleson WH, Guo L (2015) Endoplasmic reticulum stress and store-operated calcium entry contribute to usnic acid-induced toxicity in hepatic cells. Toxicol Sci 146(1):116–126. https://doi.org/10.1093/toxsci/kfv075

    Article  CAS  PubMed  Google Scholar 

  137. Uzi D, Barda L, Scaiewicz V, Mills M, Mueller T, Gonzalez-Rodriguez A, Valverde AM, Iwawaki T, Nahmias Y, Xavier R, Chung RT, Tirosh B, Shibolet O (2013) CHOP is a critical regulator of acetaminophen-induced hepatotoxicity. J Hepatol 59(3):495–503. https://doi.org/10.1016/j.jhep.2013.04.024

    Article  CAS  PubMed  Google Scholar 

  138. Park IJ, Kim MJ, Park OJ, Choe W, Kang I, Kim SS, Ha J (2012) Cryptotanshinone induces ER stress-mediated apoptosis in HepG2 and MCF7 cells. Apoptosis 17(3):248–257. https://doi.org/10.1007/s10495-011-0680-3

    Article  CAS  PubMed  Google Scholar 

  139. Boyce M, Bryant KF, Jousse C, Long K, Harding HP, Scheuner D, Kaufman RJ, Ma D, Coen DM, Ron D, Yuan J (2005) A selective inhibitor of eIF2alpha dephosphorylation protects cells from ER stress. Science 307(5711):935–939. https://doi.org/10.1126/science.1101902

    Article  CAS  PubMed  Google Scholar 

  140. Maiuri AR, Breier AB, Turkus JD, Ganey PE, Roth RA (2016) Calcium contributes to the cytotoxic interaction between diclofenac and cytokines. Toxicol Sci 149(2):372–384. https://doi.org/10.1093/toxsci/kfv249

    Article  CAS  PubMed  Google Scholar 

  141. Yusuf AT, Vian L, Sabatier R, Cano JP (2000) In vitro detection of indirect-acting genotoxins in the comet assay using Hep G2 cells. Mutat Res 468(2):227–234

    Article  CAS  PubMed  Google Scholar 

  142. Huang X, Halicka HD, Traganos F, Tanaka T, Kurose A, Darzynkiewicz Z (2005) Cytometric assessment of DNA damage in relation to cell cycle phase and apoptosis. Cell Prolif 38(4):223–243. https://doi.org/10.1111/j.1365-2184.2005.00344.x

    Article  PubMed  PubMed Central  Google Scholar 

  143. Darzynkiewicz Z, Halicka DH, Tanaka T (2009) Cytometric assessment of DNA damage induced by DNA topoisomerase inhibitors. Methods Mol Biol 582:145–153. https://doi.org/10.1007/978-1-60761-340-4_12

    Article  CAS  PubMed  Google Scholar 

  144. Sharma A, Singh K, Almasan A (2012) Histone H2AX phosphorylation: a marker for DNA damage. Methods Mol Biol 920:613–626. https://doi.org/10.1007/978-1-61779-998-3_40

    Article  CAS  PubMed  Google Scholar 

  145. Chen S, Wan L, Couch L, Lin H, Li Y, Dobrovolsky VN, Mei N, Guo L (2013) Mechanism study of goldenseal-associated DNA damage. Toxicol Lett 221(1):64–72. https://doi.org/10.1016/j.toxlet.2013.05.641

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Cover C, Mansouri A, Knight TR, Bajt ML, Lemasters JJ, Pessayre D, Jaeschke H (2005) Peroxynitrite-induced mitochondrial and endonuclease-mediated nuclear DNA damage in acetaminophen hepatotoxicity. J Pharmacol Exp Ther 315(2):879–887. https://doi.org/10.1124/jpet.105.088898

    Article  CAS  PubMed  Google Scholar 

  147. Poulsen KL, Olivero-Verbel J, Beggs KM, Ganey PE, Roth RA (2014) Trovafloxacin enhances lipopolysaccharide-stimulated production of tumor necrosis factor-alpha by macrophages: role of the DNA damage response. J Pharmacol Exp Ther 350(1):164–170. https://doi.org/10.1124/jpet.114.214189

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Williams AB, Schumacher B (2016) p53 in the DNA-damage-repair process. Cold Spring Harb Perspect Med 6(5). https://doi.org/10.1101/cshperspect.a026070

  149. Hsu IC, Tokiwa T, Bennett W, Metcalf RA, Welsh JA, Sun T, Harris CC (1993) p53 gene mutation and integrated hepatitis B viral DNA sequences in human liver cancer cell lines. Carcinogenesis 14(5):987–992

    Article  CAS  PubMed  Google Scholar 

  150. Lehman TA, Modali R, Boukamp P, Stanek J, Bennett WP, Welsh JA, Metcalf RA, Stampfer MR, Fusenig N, Rogan EM et al (1993) p53 mutations in human immortalized epithelial cell lines. Carcinogenesis 14(5):833–839

    Article  CAS  PubMed  Google Scholar 

  151. Stahler F, Roemer K (1998) Mutant p53 can provoke apoptosis in p53-deficient Hep3B cells with delayed kinetics relative to wild-type p53. Oncogene 17(26):3507–3512. https://doi.org/10.1038/sj.onc.1202245

    Article  CAS  PubMed  Google Scholar 

  152. Huang X, Darzynkiewicz Z (2006) Cytometric assessment of histone H2AX phosphorylation: a reporter of DNA damage. Methods Mol Biol 314:73–80. https://doi.org/10.1385/1-59259-973-7:073

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Tanaka T, Halicka D, Traganos F, Darzynkiewicz Z (2009) Cytometric analysis of DNA damage: phosphorylation of histone H2AX as a marker of DNA double-strand breaks (DSBs). Methods Mol Biol 523:161–168. https://doi.org/10.1007/978-1-59745-190-1_11

    Article  CAS  PubMed  Google Scholar 

  154. Barth S, Glick D, Macleod KF (2010) Autophagy: assays and artifacts. J Pathol 221(2):117–124. https://doi.org/10.1002/path.2694

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Mizushima N, Yoshimori T, Levine B (2010) Methods in mammalian autophagy research. Cell 140(3):313–326. https://doi.org/10.1016/j.cell.2010.01.028

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Klionsky DJ et al (2016) Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy 12(1):1–222. https://doi.org/10.1080/15548627.2015.1100356

    Article  PubMed  PubMed Central  Google Scholar 

  157. Chen S, Dobrovolsky VN, Liu F, Wu Y, Zhang Z, Mei N, Guo L (2014) The role of autophagy in usnic acid-induced toxicity in hepatic cells. Toxicol Sci 142(1):33–44. https://doi.org/10.1093/toxsci/kfu154

    Article  CAS  PubMed  Google Scholar 

  158. Burkard A, Dahn C, Heinz S, Zutavern A, Sonntag-Buck V, Maltman D, Przyborski S, Hewitt NJ, Braspenning J (2012) Generation of proliferating human hepatocytes using Upcyte(R) technology: characterisation and applications in induction and cytotoxicity assays. Xenobiotica 42(10):939–956. https://doi.org/10.3109/00498254.2012.675093

    Article  CAS  PubMed  Google Scholar 

  159. Tolosa L, Gomez-Lechon MJ, Lopez S, Guzman C, Castell JV, Donato MT, Jover R (2016) Human Upcyte hepatocytes: characterization of the hepatic phenotype and evaluation for acute and long-term hepatotoxicity routine testing. Toxicol Sci 152(1):214–229. https://doi.org/10.1093/toxsci/kfw078

    Article  CAS  PubMed  Google Scholar 

  160. Ware BR, Khetani SR (2016) Engineered liver platforms for different phases of drug development. Trends Biotechnol. https://doi.org/10.1016/j.tibtech.2016.08.001

  161. Peng CC, Liao WH, Chen YH, Wu CY, Tung YC (2013) A microfluidic cell culture array with various oxygen tensions. Lab Chip 13(16):3239–3245. https://doi.org/10.1039/c3lc50388g

    Article  CAS  PubMed  Google Scholar 

  162. Oshikata-Miyazaki A, Takezawa T (2016) Development of an oxygenation culture method for activating the liver-specific functions of HepG2 cells utilizing a collagen vitrigel membrane chamber. Cytotechnology 68(5):1801–1811. https://doi.org/10.1007/s10616-015-9934-1

    Article  CAS  PubMed  Google Scholar 

Download references

Disclaimer

This chapter is not an official guidance or policy statement of the US Food and Drug Administration (FDA). No official support or endorsement by the US FDA is intended or should be inferred.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lei Guo .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer Science+Business Media, LLC, part of Springer Nature

About this protocol

Check for updates. Verify currency and authenticity via CrossMark

Cite this protocol

Ren, Z., Chen, S., Ning, B., Guo, L. (2018). Use of Liver-Derived Cell Lines for the Study of Drug-Induced Liver Injury. In: Chen, M., Will, Y. (eds) Drug-Induced Liver Toxicity. Methods in Pharmacology and Toxicology. Humana, New York, NY. https://doi.org/10.1007/978-1-4939-7677-5_8

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-7677-5_8

  • Published:

  • Publisher Name: Humana, New York, NY

  • Print ISBN: 978-1-4939-7676-8

  • Online ISBN: 978-1-4939-7677-5

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics