Stable Chinese Hamster Ovary Suspension Cell Lines Harboring Recombinant Human Cytochrome P450 Oxidoreductase and Human Cytochrome P450 Monooxygenases as Platform for In Vitro Biotransformation Studies

In the liver, phase-1 biotransformation of drugs and other xenobiotics is largely facilitated by enzyme complexes consisting of cytochrome P450 oxidoreductase (CPR) and cytochrome P450 monooxygenases (CYPs). Generated from human liver-derived cell lines, recombinant in vitro cell systems with overexpression of defined phase-1 enzymes are widely used for pharmacological and toxicological drug assessment and laboratory-scale production of drug-specific reference metabolites. Most, if not all, of these cell lines, however, display some background activity of several CYPs, making it difficult to attribute effects to defined CYPs. The aim of this study was to generate cell lines with stable overexpression of human phase-1 enzymes based on Chinese hamster ovary (CHO) suspension cells. Cells were sequentially modified with cDNAs for human CPR in combination with CYP1A2, CYP2B6, or CYP3A4, using lentiviral gene transfer. In parallel, CYP-overexpressing cell lines without recombinant CPR were generated. Successful recombinant expression was demonstrated by mRNA and protein analyses. Using prototypical CYP-substrates, generated cell lines proved to display specific enzyme activities of each overexpressed CYP while we did not find any endogenous activity of those CYPs in parental CHO cells. Interestingly, cell lines revealed some evidence that the dependence of CYP activity on CPR could vary between CYPs. This needs to be confirmed in further studies. Recombinant expression of CPR was also shown to enhance CYP3A4-independent metabolisation of testosterone to androstenedione in CHO cells. We propose the novel serum-free CHO suspension cell lines with enhanced CPR and/or defined CYP activity as a promising "humanised" in vitro model to study the specific effects of those human CYPs. This could be relevant for toxicology and/or pharmacology studies in the pharmaceutical industry or medicine.

[1]  A. Jamin,et al.  Liposome-Mediated Gene Transfer in Differentiated HepaRG™ Cells: Expression of Liver Specific Functions and Application to the Cytochrome P450 2D6 Expression , 2022, Cells.

[2]  O. Kepp,et al.  Metabolic Profiling of CHO Cells during the Production of Biotherapeutics , 2022, Cells.

[3]  T. Fujita,et al.  Generation of HepG2 Cells with High Expression of Multiple Drug-Metabolizing Enzymes for Drug Discovery Research Using a PITCh System , 2022, Cells.

[4]  V. Zoumpourlis,et al.  Suitability of Human Mesenchymal Stem Cells Derived from Fetal Umbilical Cord (Wharton’s Jelly) as an Alternative In Vitro Model for Acute Drug Toxicity Screening , 2022, Cells.

[5]  A. Sadowska,et al.  Transcriptional profiling of Chinese hamster ovary (CHO) cells exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). , 2021, Reproductive toxicology.

[6]  D. Fanni,et al.  Anatomical distribution and expression of CYP in humans: Neuropharmacological implications , 2021, Drug development research.

[7]  M. Hiratsuka,et al.  Heterologous expression of high-activity cytochrome P450 in mammalian cells , 2020, Scientific Reports.

[8]  D. Schrenk,et al.  Estragole: DNA adduct formation in primary rat hepatocytes and genotoxic potential in HepG2-CYP1A2 cells. , 2020, Toxicology.

[9]  A. Zemella,et al.  Cell-Free Protein Synthesis: A Promising Option for Future Drug Development , 2020, BioDrugs.

[10]  J. Küpper,et al.  HepG2-1A2 C2 and C7: Lentivirus vector-mediated stable and functional overexpression of cytochrome P450 1A2 in human hepatoblastoma cells. , 2019, Toxicology letters.

[11]  J. Küpper,et al.  NADPH-cytochrome P450 reductase expression and enzymatic activity in primary-like human hepatocytes and HepG2 cells for in vitro biotransformation studies , 2019, Clinical hemorheology and microcirculation.

[12]  M. Henry,et al.  Improvements in single‐use bioreactor film material composition leads to robust and reliable Chinese hamster ovary cell performance , 2019, Biotechnology progress.

[13]  M. Henry,et al.  A proteomic profiling dataset of recombinant Chinese hamster ovary cells showing enhanced cellular growth following miR-378 depletion , 2018, Data in brief.

[14]  J. Küpper,et al.  Human hepatocyte systems for in vitro toxicology analysis , 2018, Journal of Cellular Biotechnology.

[15]  Edward A McKenzie,et al.  Expression of recombinant proteins in insect and mammalian cells. , 2018, Methods.

[16]  Na Gao,et al.  Effect of P450 Oxidoreductase Polymorphisms on the Metabolic Activities of Ten Cytochrome P450s Varied by Polymorphic CYP Genotypes in Human Liver Microsomes , 2018, Cellular Physiology and Biochemistry.

[17]  D. Satoh,et al.  Establishment of a novel hepatocyte model that expresses four cytochrome P450 genes stably via mammalian-derived artificial chromosome for pharmacokinetics and toxicity studies , 2017, PloS one.

[18]  P. Beaune,et al.  Engineered mesenchymal stem cells as vectors in a suicide gene therapy against preclinical murine models for solid tumors. , 2016, Journal of controlled release : official journal of the Controlled Release Society.

[19]  M. Ingelman-Sundberg,et al.  Characterization of primary human hepatocyte spheroids as a model system for drug-induced liver injury, liver function and disease , 2016, Scientific Reports.

[20]  D. Laskin,et al.  Selective Targeting of Heme Protein in Cytochrome P450 and Nitric Oxide Synthase by Diphenyleneiodonium. , 2016, Toxicological sciences : an official journal of the Society of Toxicology.

[21]  D. Ryu,et al.  Characterization of the Gly45Asp variant of human cytochrome P450 1A1 using recombinant expression. , 2015, Toxicology letters.

[22]  M. Pawłowska,et al.  CYP3A4 overexpression enhances apoptosis induced by anticancer agent imidazoacridinone C-1311, but does not change the metabolism of C-1311 in CHO cells , 2013, Acta Pharmacologica Sinica.

[23]  Gillian Smith,et al.  Cytochrome P450 CYP1B1 Interacts with 8-Methoxypsoralen (8-MOP) and Influences Psoralen-Ultraviolet A (PUVA) Sensitivity , 2013, PloS one.

[24]  Y. Daali,et al.  Applications of CYP450 Testing in the Clinical Setting , 2013, Molecular Diagnosis & Therapy.

[25]  M. Schwab,et al.  Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. , 2013, Pharmacology & therapeutics.

[26]  W. Miller P450 Oxidoreductase Deficiency: A Disorder of Steroidogenesis with Multiple Clinical Manifestations , 2012, Science Signaling.

[27]  W. Miller,et al.  Transcriptional regulation of the human P450 oxidoreductase gene: hormonal regulation and influence of promoter polymorphisms. , 2011, Molecular endocrinology.

[28]  S. Bahr,et al.  Using microarray technology to select housekeeping genes in Chinese hamster ovary cells. , 2009, Biotechnology and bioengineering.

[29]  M. Chiu,et al.  Expression and purification of human TRPV1 in baculovirus-infected insect cells for structural studies. , 2009, Protein expression and purification.

[30]  M. Schwab,et al.  Functional pharmacogenetics/genomics of human cytochromes P450 involved in drug biotransformation , 2008, Analytical and bioanalytical chemistry.

[31]  T. Lynch,et al.  The effect of cytochrome P450 metabolism on drug response, interactions, and adverse effects. , 2007, American family physician.

[32]  F. Katzen Gateway® recombinational cloning: a biological operating system , 2007, Expert opinion on drug discovery.

[33]  J. Lengler,et al.  Cytochrome P450 reductase dependent inhibition of cytochrome P450 2B1 activity: Implications for gene directed enzyme prodrug therapy. , 2006, Biochemical pharmacology.

[34]  Rieko Arimoto,et al.  Computational models for predicting interactions with cytochrome p450 enzyme. , 2006, Current topics in medicinal chemistry.

[35]  J. Doehmer,et al.  Heterologous Co-Expression of Human Cytochrome P450 1A2 and Polymorphic Forms of N-Acetyltransferase 2 for Studies on Aromatic Amines in V79 Chinese Hamster Cells , 2005, Alternatives to laboratory animals : ATLA.

[36]  T. Poulos,et al.  Intermediates in P450 catalysis , 2005, Philosophical Transactions of the Royal Society A: Mathematical, Physical and Engineering Sciences.

[37]  F. Schmidt,et al.  Recombinant expression systems in the pharmaceutical industry , 2004, Applied Microbiology and Biotechnology.

[38]  K. Usmani,et al.  Human Cytochrome P450: Metabolism of Testosterone by CYP3A4 and Inhibition by Ketoconazole , 2004, Current protocols in toxicology.

[39]  N. J. Hewitt,et al.  Phase I and II enzyme characterization of two sources of HepG2 cell lines , 2004, Xenobiotica; the fate of foreign compounds in biological systems.

[40]  C. Wolf,et al.  Human NADPH-P450 oxidoreductase modulates the level of cytochrome P450 CYP2D6 holoprotein via haem oxygenase-dependent and -independent pathways. , 2001, The Biochemical journal.

[41]  L. Naldini,et al.  Lentiviral vectors: excellent tools for experimental gene transfer and promising candidates for gene therapy , 2000, The journal of gene medicine.

[42]  A. Luch,et al.  Stable expression of human cytochrome P450 1B1 in V79 Chinese hamster cells and metabolically catalyzed DNA adduct formation of dibenzo[a,l]pyrene. , 1998, Chemical research in toxicology.

[43]  C. Wolf,et al.  High levels of recombinant CYP3A4 expression in Chinese hamster ovary cells are modulated by coexpressed human P450 reductase and hemin supplementation. , 1997, Archives of biochemistry and biophysics.

[44]  F. Gonzalez,et al.  Coexpression of cytochrome P4502A6 and human NADPH-P450 oxidoreductase in the baculovirus system. , 1997, Drug metabolism and disposition: the biological fate of chemicals.

[45]  A. Seidel,et al.  Cytochrome P450-mediated activation of phenanthrene in genetically engineered V79 Chinese hamster cells. , 1996, Environmental toxicology and pharmacology.

[46]  H. Yamazaki,et al.  Interindividual variations in human liver cytochrome P-450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicals: studies with liver microsomes of 30 Japanese and 30 Caucasians. , 1994, The Journal of pharmacology and experimental therapeutics.

[47]  F. Hanaoka,et al.  Blasticidin S-resistance gene (bsr): a novel selectable marker for mammalian cells. , 1991, Experimental cell research.

[48]  W. Anderson,et al.  Gonadotropin stimulation of pregnenolone metabolism in Chinese Hamster Ovary cells in culture , 1981, Journal of cellular physiology.

[49]  U. K. Laemmli,et al.  Cleavage of Structural Proteins during the Assembly of the Head of Bacteriophage T4 , 1970, Nature.

[50]  J. Küpper,et al.  HepG2 cells with recombinant cytochrome P450 enzyme overexpression: Their use and limitation as in vitro liver model , 2019, Journal of Cellular Biotechnology.

[51]  Jan G. Hengstler,et al.  Comparative analysis of 3D culture methods on human HepG2 cells , 2016, Archives of Toxicology.

[52]  Kai‐Uwe Schmidtke,et al.  Generation of cytochrome P450 3A4-overexpressing HepG2 cell clones for standardization of hepatocellular testosterone 6β-hydroxylation activity , 2015 .

[53]  C. Liu,et al.  Scalable transient protein expression. , 2014, Methods in molecular biology.

[54]  F. Guengerich Cytochrome p450 and chemical toxicology. , 2008, Chemical research in toxicology.

[55]  M. Eichelbaum,et al.  V79 Chinese hamster cells genetically engineered for polymorphic cytochrome P450 2D6 and their predictive value for humans. , 2003, ALTEX.