Skip to main content

Advertisement

Log in

Toxicogenomics directory of chemically exposed human hepatocytes

  • In vitro systems
  • Published:
Archives of Toxicology Aims and scope Submit manuscript

A Correction to this article was published on 31 January 2022

This article has been updated

Abstract

A long-term goal of numerous research projects is to identify biomarkers for in vitro systems predicting toxicity in vivo. Often, transcriptomics data are used to identify candidates for further evaluation. However, a systematic directory summarizing key features of chemically influenced genes in human hepatocytes is not yet available. To bridge this gap, we used the Open TG-GATES database with Affymetrix files of cultivated human hepatocytes incubated with chemicals, further sets of gene array data with hepatocytes from human donors generated in this study, and publicly available genome-wide datasets of human liver tissue from patients with non-alcoholic steatohepatitis (NASH), cirrhosis, and hepatocellular cancer (HCC). After a curation procedure, expression data of 143 chemicals were included into a comprehensive biostatistical analysis. The results are summarized in the publicly available toxicotranscriptomics directory (http://wiki.toxbank.net/toxicogenomics-map/) which provides information for all genes whether they are up- or downregulated by chemicals and, if yes, by which compounds. The directory also informs about the following key features of chemically influenced genes: (1) Stereotypical stress response. When chemicals induce strong expression alterations, this usually includes a complex but highly reproducible pattern named ‘stereotypical response.’ On the other hand, more specific expression responses exist that are induced only by individual compounds or small numbers of compounds. The directory differentiates if the gene is part of the stereotypical stress response or if it represents a more specific reaction. (2) Liver disease-associated genes. Approximately 20 % of the genes influenced by chemicals are up- or downregulated, also in liver disease. Liver disease genes deregulated in cirrhosis, HCC, and NASH that overlap with genes of the aforementioned stereotypical chemical stress response include CYP3A7, normally expressed in fetal liver; the phase II metabolizing enzyme SULT1C2; ALDH8A1, known to generate the ligand of RXR, one of the master regulators of gene expression in the liver; and several genes involved in normal liver functions: CPS1, PCK1, SLC2A2, CYP8B1, CYP4A11, ABCA8, and ADH4. (3) Unstable baseline genes. The process of isolating and the cultivation of hepatocytes was sufficient to induce some stress leading to alterations in the expression of genes, the so-called unstable baseline genes. (4) Biological function. Although more than 2,000 genes are transcriptionally influenced by chemicals, they can be assigned to a relatively small group of biological functions, including energy and lipid metabolism, inflammation and immune response, protein modification, endogenous and xenobiotic metabolism, cytoskeletal organization, stress response, and DNA repair. In conclusion, the introduced toxicotranscriptomics directory offers a basis for a rationale choice of candidate genes for biomarker evaluation studies and represents an easy to use source of background information on chemically influenced genes.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10

Similar content being viewed by others

Change history

References

  • Alexa A, Rahnenführer J (2010) topGO: topGO: enrichment analysis for gene ontology. R package version 2.12.0

  • Anders S, Huber W (2010) Differential expression analysis for sequence count data. Genome Biol 11(10):R106

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Antoun J, Amet Y, Simon B, Dréano Y, Corlu A, Corcos L, Salaun JP, Plée-Gautier E (2006) CYP4A11 is repressed by retinoic acid in human liver cells. FEBS Lett 580(14):3361–3367

    Article  CAS  PubMed  Google Scholar 

  • Balmer NV, Klima S, Rempel E, Ivanova VN, Kolde R, Weng MK, Meganathan K, Henry M, Sachinidis A, Berthold MR, Hengstler JG, Rahnenführer J, Waldmann T, Leist M (2014) From transient transcriptome responses to disturbed neurodevelopment: role of histone acetylation and methylation as epigenetic switch between reversible and irreversible drug effects. Arch Toxicol 88(7):1451–1468

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Bauer A, Schumann A, Gilbert M, Wilhelm C, Hengstler JG, Schiller J, Fuchs B (2009) Evaluation of carbon tetrachloride-induced stress on rat hepatocytes by 31P NMR and MALDI-TOF mass spectrometry: lysophosphatidylcholine generation from unsaturated phosphatidylcholines. Chem Phys Lipids 159(1):21–29

    Article  CAS  PubMed  Google Scholar 

  • Benjamini Y, Hochberg Y (1995) Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Roy Stat Soc B 57:289–300

    Google Scholar 

  • Campos G, Schmidt-Heck W, Ghallab A, Rochlitz K, Pütter L, Medinas DB, Hetz C, Widera A, Cadenas C, Begher-Tibbe B, Reif R, Günther G, Sachinidis A, Hengstler JG, Godoy P (2014) The transcription factor CHOP, a central component of the transcriptional regulatory network induced upon CCl4 intoxication in mouse liver, is not a critical mediator of hepatotoxicity. Arch Toxicol 88(6):1267–1280

    Article  CAS  PubMed  Google Scholar 

  • Elkon R, Linhart C, Sharan R, Shamir R, Shiloh Y (2003) Genome-wide in silico identification of transcriptional regulators controlling the cell cycle in human cells. Genome Res 13:773–780

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Ellinger-Ziegelbauer H, Gmuender H, Bandenburg A, Ahr HJ (2008) Prediction of a carcinogenic potential of rat hepatocarcinogens using toxicogenomics analysis of short-term in vivo studies. Mutat Res 637(1–2):23–39

    Article  CAS  PubMed  Google Scholar 

  • Fijten RR, Jennen DG, van Delft JH (2013) Pathways for ligand activated nuclear receptors to unravel the genomic responses induced by hepatotoxicants. Curr Drug Metab 14(10):1022–1028

    Article  CAS  PubMed  Google Scholar 

  • Froguel P, Zouali H, Sun F, Velho G, Fukumoto H, Passa P, Cohen D (1991) CA repeat polymorphism in the glucose transporter GLUT 2 gene. Nucleic Acids Res 19(13):3754

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Gåfvels M, Olin M, Chowdhary BP, Raudsepp T, Andersson U, Persson B, Jansson M, Björkhem I, Eggertsen G (1999) Structure and chromosomal assignment of the sterol 12alpha-hydroxylase gene (CYP8B1) in human and mouse: eukaryotic cytochrome P-450 gene devoid of introns. Genomics 56(2):184–196

    Article  PubMed  Google Scholar 

  • Godoy P (2013) Recent advances in 2D and 3D in vitro systems using primary hepatocytes, alternative hepatocyte sources and non-parenchymal liver cells and their use in investigating mechanisms of hepatotoxicity, cell signaling and ADME. Arch Toxicol 87:1315–1530

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Godoy P, Hengstler JG, Ilkavets I, Meyer C, Bachmann A, Müller A, Tuschl G, Mueller SO, Dooley S (2009) Extracellular matrix modulates sensitivity of hepatocytes to fibroblastoid dedifferentiation and transforming growth factor beta-induced apoptosis. Hepatology 49(6):2031–2043

    Article  CAS  PubMed  Google Scholar 

  • Godoy P, Lakkapamu S, Schug M, Bauer A, Stewart JD, Bedawi E, Hammad S, Amin J, Marchan R, Schormann W, Maccoux L, von Recklinghausen I, Reif R, Hengstler JG (2010a) Dexamethasone-dependent versus -independent markers of epithelial to mesenchymal transition in primary hepatocytes. Biol Chem 391(1):73–83

    Article  CAS  PubMed  Google Scholar 

  • Godoy P, Schug M, Bauer A, Hengstler JG (2010b) Reversible manipulation of apoptosis sensitivity in cultured hepatocytes by matrix-mediated manipulation of signaling activities. Methods Mol Biol 640:139–155

    Article  CAS  PubMed  Google Scholar 

  • Harbron C, Chang KM, South MC (2007) RefPlus: an R package extending the RMA Algorithm. Bioinformatics 23(18):2493–2494 Epub 2007 Jul 10

    Article  CAS  PubMed  Google Scholar 

  • Heise T, Schug M, Storm D, Ellinger-Ziegelbauer H, Ahr HJ, Hellwig B, Rahnenfuhrer J, Ghallab A, Guenther G, Sisnaiske J, Reif R, Godoy P, Mielke H, Gundert-Remy U, Lampen A, Oberemm A, Hengstler JG (2012) In vitro–in vivo correlation of gene expression alterations induced by liver carcinogens. Curr Med Chem 19(11):1721–1730

    Article  CAS  PubMed  Google Scholar 

  • Hewitt NJ, Lechón MJ, Houston JB, Hallifax D, Brown HS, Maurel P, Kenna JG, Gustavsson L, Lohmann C, Skonberg C, Guillouzo A, Tuschl G, Li AP, LeCluyse E, Groothuis GM, Hengstler JG (2007) Primary hepatocytes: current understanding of the regulation of metabolic enzymes and transporter proteins, and pharmaceutical practice for the use of hepatocytes in metabolism, enzyme induction, transporter, clearance, and hepatotoxicity studies. Drug Metab Rev 39(1):159–234

    Article  CAS  PubMed  Google Scholar 

  • Hirode M, Omura K, Kiyosawa N, Uehara T, Shimuzu T, Ono A, Miyagishima T, Nagao T, Ohno Y, Urushidani T (2009) Gene expression profiling in rat liver treated with various hepatotoxic-compounds inducing coagulopathy. J Toxicol Sci 34(3):281–293

    Article  CAS  PubMed  Google Scholar 

  • Hoehme S, Hengstler JG, Brulport M, Schäfer M, Bauer A, Gebhardt R, Drasdo D (2007) Mathematical modelling of liver regeneration after intoxication with CCl(4). Chem Biol Interact 168(1):74–93

    Article  CAS  Google Scholar 

  • Hoehme S, Brulport M, Bauer A, Bedawy E, Schormann W, Hermes M, Puppe V, Gebhardt R, Zellmer S, Schwarz M, Bockamp E, Timmel T, Hengstler JG, Drasdo D (2010) Prediction and validation of cell alignment along microvessels as order principle to restore tissue architecture in liver regeneration. Proc Natl Acad Sci USA 107(23):10371–10376

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Huang WY, Li ZG, Rus H, Wang X, Jose PA, Chen SY (2009) RGC-32 mediates transforming growth factor-beta-induced epithelial-mesenchymal transition in human renal proximal tubular cells. J Biol Chem 284(14):9426–9432

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Kamiya A, Inoue Y, Gonzalez FJ (2003) Role of the hepatocyte nuclear factor 4alpha in control of the pregnane X receptor during fetal liver development. Hepatology 37(6):1375–1384

    Article  CAS  PubMed  Google Scholar 

  • Kimura Y, Nishimura FT, Abe S, Fukunaga T, Tanii H, Saijoh K (2009) Polymorphisms in the promoter region of the human class II alcohol dehydrogenase (ADH4) gene affect both transcriptional activity and ethanol metabolism in Japanese subjects. J Toxicol Sci 34(1):89–97

    Article  CAS  PubMed  Google Scholar 

  • Kiyosawa N, Ando Y, Watanabe K, Niino N, Manabe S, Yamoto T (2009) Scoring multiple toxicological endpoints using a toxicogenomic database. Toxicol Lett 188(2):91–97

    Article  CAS  PubMed  Google Scholar 

  • Krug AK, Kolde R, Gaspar JA, Rempel E, Balmer NV, Meganathan K, Vojnits K, Baquié M, Waldmann T, Ensenat-Waser R, Jagtap S, Evans RM, Julien S, Peterson H, Zagoura D, Kadereit S, Gerhard D, Sotiriadou I, Heke M, Natarajan K, Henry M, Winkler J, Marchan R, Stoppini L, Bosgra S, Westerhout J, Verwei M, Vilo J, Kortenkamp A, Hescheler J, Hothorn L, Bremer S, van Thriel C, Krause KH, Hengstler JG, Rahnenführer J, Leist M, Sachinidis A (2013) Human embryonic stem cell-derived test systems for developmental neurotoxicity: a transcriptomics approach. Arch Toxicol 87(1):123–143

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Lake AD, Novak P, Fisher CD, Jackson JP, Hardwick RN, Billheimer DD, Klimecki WT, Cherrington NJ (2011) Analysis of global and absorption, distribution, metabolism, and elimination gene expression in the progressive stages of human nonalcoholic fatty liver disease. Drug Metab Dispos 39:1954–1960

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Lin M, Napoli JL (2000) cDNA cloning and expression of a human aldehyde dehydrogenase (ALDH) active with 9-cis-retinal and identification of a rat ortholog, ALDH12. J Biol Chem 275(51):40106–40112

    Article  CAS  PubMed  Google Scholar 

  • NIBIO (2013) National Institute of Biomedical Innovation. http://toxico.nibio.go.jp. Stand: 13.08.2013

  • Pang XY, Cheng J, Kim JH, Matsubara T, Krausz KW, Gonzalez FJ (2012) Expression and regulation of human fetal-specific CYP3A7 in mice. Endocrinology 153(3):1453–1463

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Pilz AJ, Willer E, Povey S, Abbott CM (1992) The genes coding for phosphoenolpyruvate carboxykinase-1 (PCK1) and neuronal nicotinic acetylcholine receptor alpha 4 subunit (CHRNA4) map to human chromosome 20, extending the known region of homology with mouse chromosome 2. Ann Hum Genet 56(Pt 4):289–293

    Article  CAS  PubMed  Google Scholar 

  • Saigusa K, Imoto I, Tanikawa C, Aoyagi M, Ohno K, Nakamura Y, Inazawa J (2007) RGC32, a novel p53-inducible gene, is located on centrosomes during mitosis and results in G2/M arrest. Oncogene 26(8):1110–1121

    Article  CAS  PubMed  Google Scholar 

  • Schliess F, Hoehme S, Henkel SG, Ghallab A, Driesch D, Böttger J, Guthke R, Pfaff M, Hengstler JG, Gebhardt R, Häussinger D, Drasdo D, Zellmer S (2014) Integrated metabolic spatial-temporal model for the prediction of ammonia detoxification during liver damage and regeneration. Hepatology. doi:10.1002/hep.27136 [Epub ahead of print]

  • Schug M, Stöber R, Heise T, Mielke H, Gundert-Remy U, Godoy P, Reif R, Blaszkewicz M, Ellinger-Ziegelbauer H, Ahr HJ, Selinski S, Günther G, Marchan R, Blaszkewicz M, Sachinidis A, Nüssler A, Oberemm A, Hengstler JG (2013) Pharmacokinetics explain in vivo/in vitro discrepancies of carcinogen-induced gene expression alterations in rat liver and cultivated hepatocytes. Arch Toxicol 87(2):337–345

    Article  CAS  PubMed  Google Scholar 

  • Seglen PO (1976) Preparation of isolated rat liver cells. Methods Cell Biol 13:29–83

    Article  CAS  PubMed  Google Scholar 

  • Simmer JP, Kelly RE, Rinker AG Jr, Scully JL, Evans DR (1990) Mammalian carbamyl phosphate synthetase (CPS). DNA sequence and evolution of the CPS domain of the Syrian hamster multifunctional protein CAD. J Biol Chem 265(18):10395–10402

    CAS  PubMed  Google Scholar 

  • Tsuruoka S, Ishibashi K, Yamamoto H, Wakaumi M, Suzuki M, Schwartz GJ, Imai M, Fujimura A (2002) Functional analysis of ABCA8, a new drug transporter. Biochem Biophys Res Commun 298(1):41–45

    Article  CAS  PubMed  Google Scholar 

  • Tung EK, Mak CK, Fatima S, Lo RC, Zhao H, Zhang C, Dai H, Poon RT, Yuen MF, Lai CL et al (2011) Clinicopathological and prognostic significance of serum and tissue Dickkopf-1 levels in human hepatocellular carcinoma. Liver Int 31:1494–1504

    Article  CAS  PubMed  Google Scholar 

  • Uehara T, Minowa Y, Morikawa Y, Kondo C, Maruyama T, Kato I, Nakatsu N, Igarashi Y, Ono A, Hayashi H, Mitsumori K, Yamada H, Ohno Y, Urushidani T (2011) Prediction model of potential hepatocarcinogenicity of rat hepatocarcinogens using a large-scale toxicogenomics database. Toxicol Appl Pharmacol 255(3):297–306

    Article  CAS  PubMed  Google Scholar 

  • Ulitsky I, Maron-Katz A, Shavit S, Sagir D, Linhart C, Elkon R, Tanay A, Sharan R, Shiloh Y, Shamir R (2010) Expander: from expression microarrays to networks and functions. Nat Protoc 5:303–322

    Article  CAS  PubMed  Google Scholar 

  • Väremo L, Nielsen J, Nookaew I (2013) Enriching the gene set analysis of genome-wide data by incorporating directionality of gene expression and combining statistical hypotheses and methods. Nucleic Acids Res 4:4378–4391

    Article  Google Scholar 

  • Waldmann T, Rempel E, Balmer NV, König A, Kolde R, Gaspar JA, Henry M, Hescheler J, Sachinidis A, Rahnenführer J, Hengstler JG, Leist M (2014) Design principles of concentration-dependent transcriptome deviations in drug-exposed differentiating stem cells. Chem Res Toxicol 27(3):408–420

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Watt AJ, Garrison WD, Duncan SA (2003) HNF4: a central regulator of hepatocyte differentiation and function. Hepatology 37(6):1249–1253

    Article  CAS  PubMed  Google Scholar 

  • Weng MK, Natarajan K, Scholz D, Ivanova VN, Sachinidis A, Hengstler JG, Waldmann T, Leist M (2014) Lineage-specific regulation of epigenetic modifier genes in human liver and brain. PLoS ONE 9(7):e102035

    Article  PubMed Central  PubMed  Google Scholar 

  • Wu J, Wang C, Li S, Li S, Wang W, Li J, Chi Y, Yang H, Kong X, Zhou Y, Dong C, Wang F, Xu G, Yang J, Gustafsson JÅ, Guan Y (2013) Thyroid hormone-responsive SPOT 14 homolog promotes hepatic lipogenesis, and its expression is regulated by liver X receptor α through a sterol regulatory element-binding protein 1c-dependent mechanism in mice. Hepatology 58(2):617–628

    Article  CAS  PubMed  Google Scholar 

  • Xing L, Wu L, Liu Y, Ai N, Lu X, Fan X (2014) LTMap: a web server for assessing the potential liver toxicity by genome-wide transcriptional expression data. J Appl Toxicol 34(7):805–809

    Article  CAS  PubMed  Google Scholar 

  • Zellmer S, Schmidt-Heck W, Godoy P, Weng H, Meyer C, Lehmann T, Sparna T, Schormann W, Hammad S, Kreutz C, Timmer J, von Weizsäcker F, Thürmann PA, Merfort I, Guthke R, Dooley S, Hengstler JG, Gebhardt R (2010) Transcription factors ETF, E2F, and SP-1 are involved in cytokine-independent proliferation of murine hepatocytes. Hepatology 52(6):2127–2136

    Article  CAS  PubMed  Google Scholar 

  • Zhang JD, Berntenis N, Roth A, Ebeling M (2014) Data mining reveals a network of early-response genes as a consensus signature of drug-induced in vitro and in vivo toxicity. Pharmacogenomics J 14(3):208–216

    Article  CAS  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgments

This study was supported by the SEURAT-1 projects DETECTIVE, TOXBANK, and NOTOX, the BMBF projects LivSys, SysDT and Lebersimulator and Deutsche Forschungsgemeinschaft (DFG) within the Collaborative Research Center SFB 876  “Providing Information by Resource-Constrained Analysis”, project A3. The results published here are in part based upon data generated by the TCGA Research Network: http://cancergenome.nih.gov/.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jan G. Hengstler.

Additional information

Marianna Grinberg and Regina M. Stöber shared first authorship.

Jörg Rahnenführer and Jan G. Hengstler shared senior authorship.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (XLSX 11 kb)

Supplementary material 2 (XLSX 22 kb)

Supplementary material 3 (DOCX 16 kb)

Supplementary material 4 (DOCX 20 kb)

Supplementary material 5 (XLSX 16 kb)

Supplementary material 6 (XLSX 64 kb)

Supplementary material 7 (XLSX 14 kb)

Supplementary material 8 (DOCX 16 kb)

Supplementary material 9 (XLSX 1347 kb)

Supplementary material 10 (XLSX 12 kb)

Supplementary material 11 (XLSX 456 kb)

Supplementary material 12 (XLSX 278 kb)

Supplementary material 13 (XLSX 4725 kb)

Supplementary material 14 (XLSX 16 kb)

Supplementary material 15 (DOCX 59 kb)

Supplementary material 16 (XLSX 22 kb)

Supplementary material 17 (XLSX 282 kb)

Supplementary material 18 (XLSX 24 kb)

Supplementary material 19 (XLSX 16 kb)

204_2014_1400_MOESM20_ESM.pptx

Fig. S1: Corresponding data to Fig. 1 summarizing all further incubation conditions besides the high concentration and 24h exposure already shown in Fig. 1. A. Low concentration, 2h, 8h and 24h incubation; B. middle concentration, 2h, 8h and 24h incubation; C. high concentration, 2h, 8h and 24h incubation. (PPTX 1204 kb)

204_2014_1400_MOESM21_ESM.pptx

Fig. S2: Numbers of significantly downregulated genes. The x-axis lists all compounds that were tested at the indicated concentration for the corresponding period. The y-axis gives the number of - downregulated genes with at least 1.5-, 2.0- and 3.0-fold change. The result shows that the number of deregulated genes differs strongly between the chemicals. The figure corresponds to Fig. 2 where the upregulated genes are shown. Dark green: more than 1.5-fold downregulated; light green: more than 2-fold downregulated; black: more than 3-fold downregulated. (PPTX 714 kb)

204_2014_1400_MOESM22_ESM.pptx

Fig. S3: ‘Exclusivity analysis’ of the downregulated genes. This analysis first determines the 100 strongest downregulated genes across all compounds. Next, these genes are assigned to the compound with the most extreme fold change. The analysis corresponds to Fig. 4 where the upregulated genes are shown. (PPTX 150 kb)

204_2014_1400_MOESM23_ESM.pptx

Fig. S4: Selection values for the downregulated genes. A selection value of e.g. five means that at least five compounds downregulate (3-fold) the indicated number of genes. The figure corresponds to Fig. 8A where the selection values for the upregulated genes are shown. (PPTX 116 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Grinberg, M., Stöber, R.M., Edlund, K. et al. Toxicogenomics directory of chemically exposed human hepatocytes. Arch Toxicol 88, 2261–2287 (2014). https://doi.org/10.1007/s00204-014-1400-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00204-014-1400-x

Keywords

Navigation