1
|
Vlach M, Coppens-Exandier H, Jamin A, Berchel M, Scaviner J, Chesné C, Montier T, Jaffrès PA, Corlu A, Loyer P. Liposome-Mediated Gene Transfer in Differentiated HepaRG™ Cells: Expression of Liver Specific Functions and Application to the Cytochrome P450 2D6 Expression. Cells 2022; 11:cells11233904. [PMID: 36497165 PMCID: PMC9737581 DOI: 10.3390/cells11233904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/19/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
The goal of this study was to establish a procedure for gene delivery mediated by cationic liposomes in quiescent differentiated HepaRG™ human hepatoma cells. We first identified several cationic lipids promoting efficient gene transfer with low toxicity in actively dividing HepG2, HuH7, BC2 and progenitor HepaRG™ human hepatoma cells. The lipophosphoramidate Syn1-based nanovector, which allowed the highest transfection efficiencies of progenitor HepaRG™ cells, was next used to transfect differentiated HepaRG™ cells. Lipofection of these cells using Syn1-based liposome was poorly efficient most likely because the differentiated HepaRG™ cells are highly quiescent. Thus, we engineered the differentiated HepaRG™ Mitogenic medium supplement (ADD1001) that triggered robust proliferation of differentiated cells. Importantly, we characterized the phenotypical changes occurring during proliferation of differentiated HepaRG™ cells and demonstrated that mitogenic stimulation induced a partial and transient decrease in the expression levels of some liver specific functions followed by a fast recovery of the full differentiation status upon removal of the mitogens. Taking advantage of the proliferation of HepaRG™ cells, we defined lipofection conditions using Syn1-based liposomes allowing transient expression of the cytochrome P450 2D6, a phase I enzyme poorly expressed in HepaRG cells, which opens new means for drug metabolism studies in HepaRG™ cells.
Collapse
Affiliation(s)
- Manuel Vlach
- Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
- Institut AGRO Rennes-Angers, F-35042 Rennes, France
| | - Hugo Coppens-Exandier
- Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
- Biopredic International, F-35760 Saint Grégoire, France
| | - Agnès Jamin
- Biopredic International, F-35760 Saint Grégoire, France
| | - Mathieu Berchel
- Univ. Brest, CNRS, CEMCA, UMR 6521, F-29238 Brest, France
- Plateforme BiogenOuest SynNanoVect, F-44035 Nantes, France
| | - Julien Scaviner
- Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
- Biopredic International, F-35760 Saint Grégoire, France
| | | | - Tristan Montier
- Plateforme BiogenOuest SynNanoVect, F-44035 Nantes, France
- Univ. Brest, INSERM, EFS, UMR 1078, GGB-GTCA, F-29200 Brest, France
| | - Paul-Alain Jaffrès
- Univ. Brest, CNRS, CEMCA, UMR 6521, F-29238 Brest, France
- Plateforme BiogenOuest SynNanoVect, F-44035 Nantes, France
| | - Anne Corlu
- Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
- Correspondence: (A.C.); (P.L.); Tel.: +33-(02)-23233873 (P.L.)
| | - Pascal Loyer
- Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
- Plateforme BiogenOuest SynNanoVect, F-44035 Nantes, France
- Correspondence: (A.C.); (P.L.); Tel.: +33-(02)-23233873 (P.L.)
| |
Collapse
|
2
|
Coltman NJ, Coke BA, Chatzi K, Shepherd EL, Lalor PF, Schulz-Utermoehl T, Hodges NJ. Application of HepG2/C3A liver spheroids as a model system for genotoxicity studies. Toxicol Lett 2021; 345:34-45. [PMID: 33865918 DOI: 10.1016/j.toxlet.2021.04.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/19/2021] [Accepted: 04/11/2021] [Indexed: 12/14/2022]
Abstract
HepG2 cells continue to be a valuable tool in early drug discovery and pharmaceutical development. In the current study we develop a 3D in vitro liver model, using HepG2/C3A cells that is predictive of human genotoxic exposure. HepG2/C3A cells cultured for 7-days in agarose-coated microplates formed spheroids which were uniform in shape and had well defined outer perimeters and no evidence of a hypoxic core. Quantitative real-time-PCR analysis showed statistically significant transcriptional upregulation of xenobiotic metabolising genes (CYP1A1, CYP1A2, UG1A1, UGT1A3, UGT1A6, EPHX, NAT2) and genes linked to liver function (ALB, CAR) in 3D cultures. In response to three model pro-genotoxicants: benzo[a]pyrene, amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) and 2-aminoanthracene (2-AA), we observed further transcriptional upregulation of xenobiotic metabolising genes (CYP1A1, CYP1A2, NAT1/2, SULT1A2, UGT1A1, UGT1A3) compared to untreated spheroids. Consistent with this, spheroids were more sensitive than 2D monolayers to compound induced single- and double- stranded DNA-damage as assessed by the comet assay and γH2AX phosphorylation respectively. In contrast, levels of DNA-damage induced by the direct acting mutagen 4-nitroquinoline N-oxide (4NQO) was the same in spheroids and monolayers. In support of the enhanced genotoxic response in spheroids we also observed transcriptional upregulation of genes relating to DNA-damage and cellular stress response (e.g. GADD45A and CDKN1A) in spheroids. In conclusion, HepG2/C3A 3D spheroids are a sensitive model for in vitro genotoxicity assessment with potential applications in early stage drug development.
Collapse
Affiliation(s)
- Nicholas J Coltman
- School of Biosciences, University of Birmingham, Birmingham, B15 2TT, United Kingdom.
| | - Brandon A Coke
- School of Biosciences, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Kyriaki Chatzi
- School of Biosciences, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Emma L Shepherd
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Inflammation, and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, The Medical School, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Patricia F Lalor
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Inflammation, and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, The Medical School, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Timothy Schulz-Utermoehl
- Sygnature Discovery, The Discovery Building, BioCity, Pennyfoot Street, Nottingham, United Kingdom
| | - Nikolas J Hodges
- School of Biosciences, University of Birmingham, Birmingham, B15 2TT, United Kingdom.
| |
Collapse
|
3
|
Chen S, Wu Q, Li X, Li D, Mei N, Ning B, Puig M, Ren Z, Tolleson WH, Guo L. Characterization of cytochrome P450s (CYP)-overexpressing HepG2 cells for assessing drug and chemical-induced liver toxicity. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2021; 39:68-86. [PMID: 33576714 PMCID: PMC7931144 DOI: 10.1080/26896583.2021.1880242] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Hepatic metabolism catalyzed by the cytochrome P450 (CYP) superfamily affects liver toxicity associated with exposures to natural compounds and xenobiotic agents. Previously we generated a battery of HepG2-derived stable cell lines that individually express 14 CYPs (1A1, 1A2, 1B1, 2A6, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6, 2E1, 3A4, 3A5, and 3A7). In this study, we comprehensively characterized each cell line for its CYP expression and enzyme activity. Specifically, we measured the mRNA expression, protein expression, and metabolite formation. Using CYP3A4, 2D6, and 2C9-overexpressing cells as representatives, we examined the stability of these cells in long-term cultures for up to 10 passages. The results showed that CYPs can be stably overexpressed for up to 10 cell culture passages without losing their activities. The robustness of responses to stimuli among the cells at different passages was also investigated in CYP3A4-overexpressing cells and the response to amiodarone and dronedarone showed no difference between the cells at the passage 2 and 10. Moreover, the mRNA expression level of most CYPs was higher in CYP-overexpressing HepG2 cells than that in HepaRG cells and primary human hepatocytes. This study confirmed the stability of CYP-overexpressing HepG2 cell lines and provided useful information for a broader use of these cells in pharmacologic and toxicologic research.
Collapse
Affiliation(s)
- Si Chen
- Division of Biochemical Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| | - Qiangen Wu
- Division of Biochemical Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| | - Xilin Li
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| | - Dongying Li
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| | - Nan Mei
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| | - Baitang Ning
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| | - Montserrat Puig
- Division of Biotechnology Review and Research III, Office of Biotechnology Products, Center for Drug Evaluation and Research/U.S. FDA, Silver Spring, Maryland, USA
| | - Zhen Ren
- Division of Biochemical Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| | - William H. Tolleson
- Division of Biochemical Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| | - Lei Guo
- Division of Biochemical Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, Arkansas, USA
| |
Collapse
|
4
|
Schütz R, Müller M, Geisslinger F, Vollmar A, Bartel K, Bracher F. Synthesis, biological evaluation and toxicity of novel tetrandrine analogues. Eur J Med Chem 2020; 207:112810. [PMID: 32942071 PMCID: PMC7473156 DOI: 10.1016/j.ejmech.2020.112810] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/22/2020] [Accepted: 08/31/2020] [Indexed: 11/16/2022]
Abstract
In this work, we present the design and synthesis of novel fully synthetic analogues of the bisbenzylisoquinoline tetrandrine, a molecule with numerous pharmacological properties and the potential to treat life-threatening diseases, such as viral infections and cancer. Its toxicity to liver and lungs and the underlying mechanisms, however, are controversially discussed. Along this line, novel tetrandrine analogues were synthesized and biologically evaluated for their hepatotoxicity, as well as their antiproliferative and chemoresistance reversing activity on cancer cells. Previous studies suggesting CYP-mediated toxification of tetrandrine prompted us to amend/replace the suspected metabolically instable 12-methoxy group. Of note, employing several in vitro models showed that the proposed CYP3A4-driven metabolism of tetrandrine and analogues is not the major cause of hepatotoxicity. Biological characterization revealed that some of the novel tetrandrine analogues sensitized drug-resistant leukemia cells by inhibition of the P-glycoprotein. Interestingly, direct anticancer effects improved in comparison to tetrandrine, as several compounds displayed a markedly enhanced ability to reduce proliferation of drug-resistant leukemia cells and to induce cell death of liver cancer cells. Those enhanced anticancer properties were linked to influences on activation of the kinase Akt and mitochondrial events. In sum, our study clarifies the role of CYP3A4-mediated toxicity of the bisbenzylisoquinoline alkaloid tetrandrine and provides the basis for the exploitation of novel synthetic analogues for their antitumoral potential.
Collapse
Affiliation(s)
- Ramona Schütz
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Martin Müller
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Franz Geisslinger
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Angelika Vollmar
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Karin Bartel
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Franz Bracher
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany.
| |
Collapse
|
5
|
Li X, Chen S, Guo X, Wu Q, Seo JE, Guo L, Manjanatha MG, Zhou T, Witt KL, Mei N. Development and Application of TK6-derived Cells Expressing Human Cytochrome P450s for Genotoxicity Testing. Toxicol Sci 2020; 175:251-265. [PMID: 32159784 PMCID: PMC7334878 DOI: 10.1093/toxsci/kfaa035] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Metabolism plays a key role in chemical genotoxicity; however, most mammalian cells used for in vitro genotoxicity testing lack effective metabolizing enzymes. We recently developed a battery of TK6-derived cell lines that individually overexpress 1 of 8 cytochrome P450s (CYP1A1, 1A2, 1B1, 2A6, 2B6, 2C9, 2C19, and 3A4) using a lentiviral expression system. The increased expression and metabolic function of each individual CYP in each established cell line were confirmed using real-time PCR, Western blotting, and mass spectrometry analysis; the parental TK6 cells and empty vector (EV) transduced cells had negligible CYP levels. Subsequently, we evaluated these cell lines using 2 prototypical polyaromatic hydrocarbon mutagens, 7,12-dimethylbenz[a]anthracene (DMBA) and benzo[a]pyrene (B[a]P), that require metabolic activation to exert their genotoxicity. DMBA-induced cytotoxicity, phosphorylation of histone H2A.X, and micronucleus formation were significantly increased in TK6 cells with CYP1A1, 1B1, 2B6, and 2C19 expression as compared with EV controls. B[a]P significantly increased cytotoxicity, DNA damage, and chromosomal damage in TK6 cells overexpressing CYP1A1 and 1B1 when compared with EV controls. B[a]P also induced micronucleus formation in TK6 cells expressing CYP1A2. These results suggest that our CYP-expressing TK6 cell system can be used to detect the genotoxicity of compounds requiring metabolic transformation.
Collapse
Affiliation(s)
- Xilin Li
- Division of Genetic and Molecular Toxicology
| | - Si Chen
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas 72079
| | | | - Qiangen Wu
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas 72079
| | - Ji-Eun Seo
- Division of Genetic and Molecular Toxicology
| | - Lei Guo
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas 72079
| | | | - Tong Zhou
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Rockville, Maryland 20855
| | - Kristine L Witt
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Nan Mei
- Division of Genetic and Molecular Toxicology
| |
Collapse
|
6
|
Guo X, Seo JE, Li X, Mei N. Genetic toxicity assessment using liver cell models: past, present, and future. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2019; 23:27-50. [PMID: 31746269 DOI: 10.1080/10937404.2019.1692744] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Genotoxic compounds may be detoxified to non-genotoxic metabolites while many pro-carcinogens require metabolic activation to exert their genotoxicity in vivo. Standard genotoxicity assays were developed and utilized for risk assessment for over 40 years. Most of these assays are conducted in metabolically incompetent rodent or human cell lines. Deficient in normal metabolism and relying on exogenous metabolic activation systems, the current in vitro genotoxicity assays often have yielded high false positive rates, which trigger unnecessary and costly in vivo studies. Metabolically active cells such as hepatocytes have been recognized as a promising cell model in predicting genotoxicity of carcinogens in vivo. In recent years, significant advances in tissue culture and biological technologies provided new opportunities for using hepatocytes in genetic toxicology. This review encompasses published studies (both in vitro and in vivo) using hepatocytes for genotoxicity assessment. Findings from both standard and newly developed genotoxicity assays are summarized. Various liver cell models used for genotoxicity assessment are described, including the potential application of advanced liver cell models such as 3D spheroids, organoids, and engineered hepatocytes. An integrated strategy, that includes the use of human-based cells with enhanced biological relevance and throughput, and applying the quantitative analysis of data, may provide an approach for future genotoxicity risk assessment.
Collapse
Affiliation(s)
- Xiaoqing Guo
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Jefferson, AR, USA
| | - Ji-Eun Seo
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Jefferson, AR, USA
| | - Xilin Li
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Jefferson, AR, USA
| | - Nan Mei
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Jefferson, AR, USA
| |
Collapse
|
7
|
Vlach M, Quesnot N, Dubois-Pot-Schneider H, Ribault C, Verres Y, Petitjean K, Rauch C, Morel F, Robin MA, Corlu A, Loyer P. Cytochrome P450 1A1/2, 2B6 and 3A4 HepaRG Cell-Based Biosensors to Monitor Hepatocyte Differentiation, Drug Metabolism and Toxicity. SENSORS 2019; 19:s19102245. [PMID: 31096615 PMCID: PMC6567340 DOI: 10.3390/s19102245] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 01/20/2023]
Abstract
Human hepatoma HepaRG cells express most drug metabolizing enzymes and constitute a pertinent in vitro alternative cell system to primary cultures of human hepatocytes in order to determine drug metabolism and evaluate the toxicity of xenobiotics. In this work, we established novel transgenic HepaRG cells transduced with lentiviruses encoding the reporter green fluorescent protein (GFP) transcriptionally regulated by promoter sequences of cytochromes P450 (CYP) 1A1/2, 2B6 and 3A4 genes. Here, we demonstrated that GFP-biosensor transgenes shared similar expression patterns with the corresponding endogenous CYP genes during proliferation and differentiation in HepaRG cells. Interestingly, differentiated hepatocyte-like HepaRG cells expressed GFP at higher levels than cholangiocyte-like cells. Despite weaker inductions of GFP expression compared to the strong increases in mRNA levels of endogenous genes, we also demonstrated that the biosensor transgenes were induced by prototypical drug inducers benzo(a)pyrene and phenobarbital. In addition, we used the differentiated biosensor HepaRG cells to evidence that pesticide mancozeb triggered selective cytotoxicity of hepatocyte-like cells. Our data demonstrate that these new biosensor HepaRG cells have potential applications in the field of chemicals safety evaluation and the assessment of drug hepatotoxicity.
Collapse
Affiliation(s)
- Manuel Vlach
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Nicolas Quesnot
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | | | - Catherine Ribault
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Yann Verres
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Kilian Petitjean
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Claudine Rauch
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Fabrice Morel
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Marie-Anne Robin
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Anne Corlu
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
| | - Pascal Loyer
- Inserm, INRA, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR-A 1341, UMR-S 1241, Plateforme BiogenOuest SynNanoVect, F-35000 Rennes, France; (M.V.); (N.Q.); (C.R.); (Y.V.); (K.P.); (C.R.); (A.C.)
- Correspondence: ; Tel.: +33-(0)223233873; Fax: +33-(0)299540137
| |
Collapse
|
8
|
Weyers C, Dingle LMK, Wilhelmi BS, Edkins AL, Veale CGL. Use of a non-hepatic cell line highlights limitations associated with cell-based assessment of metabolically induced toxicity. Drug Chem Toxicol 2019; 43:656-662. [PMID: 30880486 DOI: 10.1080/01480545.2019.1585869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Metabolically induced drug-toxicity is a major cause of drug failure late in drug optimization phases. Accordingly, in vitro metabolic profiling of compounds is being introduced at earlier stages of the drug discovery pipeline. An increasingly common method to obtain these profiles is through overexpression of key CYP450 metabolic enzymes in immortalized liver cells, to generate competent hepatocyte surrogates. Enhanced cytotoxicity is presumed to be due to toxic metabolite production via the overexpressed enzyme. However, metabolically induced toxicity is a complex multi-parameter phenomenon and the potential background contribution to metabolism arising from the use of liver cells which endogenously express CYP450 isoforms is consistently overlooked. In this study, we sought to reduce the potential background interference by applying this methodology in kidney-derived HEK293 cells which lack endogenous CYP450 expression. Overexpression of CYP3A4 resulted in increased HEK293 proliferation, while exposure to four compounds with reported metabolically induced cytotoxicity in liver-derived cells overexpressing CYP3A4 resulted in no increase in cytotoxicity. Our results indicate that overexpression of a single CYP450 isoform in hepatic cell lines may not be a reliable method to discriminate which enzymes are responsible for metabolic induced cytotoxicity.
Collapse
Affiliation(s)
- Carli Weyers
- Faculty of Pharmacy, Rhodes University, Grahamstown, South Africa.,Biomedical Biotechnology Research Unit, Rhodes University, Grahamstown, South Africa
| | - Laura M K Dingle
- Biomedical Biotechnology Research Unit, Rhodes University, Grahamstown, South Africa.,Department of Biochemistry and Microbiology, Rhodes University, Grahamstown, South Africa
| | - Brendan S Wilhelmi
- Department of Biochemistry and Microbiology, Rhodes University, Grahamstown, South Africa
| | - Adrienne L Edkins
- Biomedical Biotechnology Research Unit, Rhodes University, Grahamstown, South Africa.,Department of Biochemistry and Microbiology, Rhodes University, Grahamstown, South Africa
| | - Clinton G L Veale
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Scottsville, South Africa
| |
Collapse
|
9
|
Xu D, Liang D, Guo Y, Sun Y. Endosulfan causes the alterations of DNA damage response through ATM-p53 signaling pathway in human leukemia cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 238:1048-1055. [PMID: 29705383 DOI: 10.1016/j.envpol.2018.03.044] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 03/09/2018] [Accepted: 03/13/2018] [Indexed: 06/08/2023]
Abstract
Exposure to pesticides results in DNA damage and genomic instability. We previously predicted that endosulfan might be associated with leukemia, but the role of endosulfan in leukemia cells has been unexplored. The aim of this study is to elucidate molecular mechanism of endosulfan-induced DNA damage response in human leukemia cells. We performed endosulfan exposure experiments in K562 cells with varying concentrations of endosulfan for 48 h and found that endosulfan lowered cell viability in a dose-dependent manner. We observed the dramatic DNA damage using comet assay and the increase of micronucleus in 75 μM endosulfan-exposed cells. Endosulfan at 75 μM caused the expression alterations of ATM and DNA repair genes such as FANCD2, and BRCA1/2 at different exposure time points (12, 24, 48 h), which was reversed by ATM inhibitor KU-55933. Endosulfan significantly increased the mRNA expression levels of p53 and GADD45A, and decreased PCNA and XRCC2 at 48 h after exposure. Flow cytometric analysis showed that endosulfan at 50 and 75 μM induced cell cycle G1 arrest, a response attributed to down-regulation of CDK6 and up-regulation of p21. We also observed that endosulfan at 50 and 75 μM induced a considerable percentage of cells to undergo apoptosis, as detected by Annexin-V binding assays. Endosulfan resulted in the activation of caspase-3, and elevated the expression levels of PUMA and the ratio of BAX/Bcl-2. These findings suggest that endosulfan caused DNA damage response throughATM-p53 signaling pathway, implicating the potential correlation between endosulfan and leukemia.
Collapse
Affiliation(s)
- Dan Xu
- Institute of Environmental Systems Biology, Dalian Maritime University, Linghai Road 1, Dalian, 116026, PR China.
| | - Dong Liang
- Institute of Environmental Systems Biology, Dalian Maritime University, Linghai Road 1, Dalian, 116026, PR China.
| | - Yubing Guo
- Institute of Environmental Systems Biology, Dalian Maritime University, Linghai Road 1, Dalian, 116026, PR China.
| | - Yeqing Sun
- Institute of Environmental Systems Biology, Dalian Maritime University, Linghai Road 1, Dalian, 116026, PR China.
| |
Collapse
|
10
|
Dias Viegas FP, de Freitas Silva M, Divino da Rocha M, Castelli MR, Riquiel MM, Machado RP, Vaz SM, Simões de Lima LM, Mancini KC, Marques de Oliveira PC, Morais ÉP, Gontijo VS, da Silva FMR, D'Alincourt da Fonseca Peçanha D, Castro NG, Neves GA, Giusti-Paiva A, Vilela FC, Orlandi L, Camps I, Veloso MP, Leomil Coelho LF, Ionta M, Ferreira-Silva GÁ, Pereira RM, Dardenne LE, Guedes IA, de Oliveira Carneiro Junior W, Quaglio Bellozi PM, Pinheiro de Oliveira AC, Ferreira FF, Pruccoli L, Tarozzi A, Viegas C. Design, synthesis and pharmacological evaluation of N-benzyl-piperidinyl-aryl-acylhydrazone derivatives as donepezil hybrids: Discovery of novel multi-target anti-alzheimer prototype drug candidates. Eur J Med Chem 2018; 147:48-65. [PMID: 29421570 DOI: 10.1016/j.ejmech.2018.01.066] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/18/2018] [Accepted: 01/19/2018] [Indexed: 12/23/2022]
Abstract
A new series of sixteen multifunctional N-benzyl-piperidine-aryl-acylhydrazones hybrid derivatives was synthesized and evaluated for multi-target activities related to Alzheimer's disease (AD). The molecular hybridization approach was based on the combination, in a single molecule, of the pharmacophoric N-benzyl-piperidine subunit of donepezil, the substituted hydroxy-piperidine fragment of the AChE inhibitor LASSBio-767, and an acylhydrazone linker, a privileged structure present in a number of synthetic aryl- and aryl-acylhydrazone derivatives with significant AChE and anti-inflammatory activities. Among them, compounds 4c, 4d, 4g and 4j presented the best AChE inhibitory activities, but only compounds 4c and 4g exhibited concurrent anti-inflammatory activity in vitro and in vivo, against amyloid beta oligomer (AβO) induced neuroinflammation. Compound 4c also showed the best in vitro and in vivo neuroprotective effects against AβO-induced neurodegeneration. In addition, compound 4c showed a similar binding mode to donepezil in both acetylated and free forms of AChE enzyme in molecular docking studies and did not show relevant toxic effects on in vitro and in vivo assays, with good predicted ADME parameters in silico. Overall, all these results highlighted compound 4c as a promising and innovative multi-target drug prototype candidate for AD treatment.
Collapse
Affiliation(s)
- Flávia Pereira Dias Viegas
- Institute of Chemistry, Laboratory of Research on Medicinal Chemistry, Federal University of Alfenas, MG 37133-840, Brazil
| | - Matheus de Freitas Silva
- Institute of Chemistry, Laboratory of Research on Medicinal Chemistry, Federal University of Alfenas, MG 37133-840, Brazil
| | - Miguel Divino da Rocha
- Institute of Chemistry, Laboratory of Research on Medicinal Chemistry, Federal University of Alfenas, MG 37133-840, Brazil
| | - Maísa Rosa Castelli
- Institute of Chemistry, Laboratory of Research on Medicinal Chemistry, Federal University of Alfenas, MG 37133-840, Brazil
| | - Mariana Máximo Riquiel
- Institute of Chemistry, Laboratory of Research on Medicinal Chemistry, Federal University of Alfenas, MG 37133-840, Brazil
| | - Rafael Pereira Machado
- Institute of Chemistry, Laboratory of Research on Medicinal Chemistry, Federal University of Alfenas, MG 37133-840, Brazil
| | - Sarah Macedo Vaz
- Institute of Chemistry, Laboratory of Research on Medicinal Chemistry, Federal University of Alfenas, MG 37133-840, Brazil
| | - Laís Medeiros Simões de Lima
- Institute of Chemistry, Laboratory of Research on Medicinal Chemistry, Federal University of Alfenas, MG 37133-840, Brazil
| | - Karla Cristine Mancini
- Institute of Chemistry, Laboratory of Research on Medicinal Chemistry, Federal University of Alfenas, MG 37133-840, Brazil
| | | | - Élida Parreira Morais
- Institute of Chemistry, Laboratory of Research on Medicinal Chemistry, Federal University of Alfenas, MG 37133-840, Brazil
| | - Vanessa Silva Gontijo
- Institute of Chemistry, Laboratory of Research on Medicinal Chemistry, Federal University of Alfenas, MG 37133-840, Brazil
| | - Fernanda Motta R da Silva
- Laboratory of Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, RJ 21941-902, Brazil
| | | | - Newton Gonçalves Castro
- Laboratory of Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, RJ 21941-902, Brazil
| | - Gilda A Neves
- Laboratory of Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, RJ 21941-902, Brazil
| | - Alexandre Giusti-Paiva
- Institute of Biomedical Sciences, Federal University of Minas Gerais, MG 37130-000, Brazil
| | - Fabiana Cardoso Vilela
- Institute of Biomedical Sciences, Federal University of Minas Gerais, MG 37130-000, Brazil
| | - Lidiane Orlandi
- Institute of Biomedical Sciences, Federal University of Minas Gerais, MG 37130-000, Brazil
| | - Ihosvany Camps
- Institute of Exact Sciences, Federal University of Alfenas, MG 37130-000, Brazil
| | | | - Luis Felipe Leomil Coelho
- Laboratory of Vaccines, Institute of Biomedical Sciences, Federal University of Alfenas, MG 37130-000, Brazil
| | - Marisa Ionta
- Institute of Biomedical Sciences, Federal University of Minas Gerais, MG 37130-000, Brazil
| | | | | | - Laurent E Dardenne
- National Laboratory of Computational Sciences, Petrópolis, RJ 25651-075, Brazil
| | | | | | | | | | - Fábio Furlan Ferreira
- Centre of Natural and Human Sciences, Federal University of ABC, Santo André, SP 09210-580, Brazil
| | - Letizia Pruccoli
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Rimini 47921, Italy
| | - Andrea Tarozzi
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Rimini 47921, Italy
| | - Claudio Viegas
- Institute of Chemistry, Laboratory of Research on Medicinal Chemistry, Federal University of Alfenas, MG 37133-840, Brazil.
| |
Collapse
|
11
|
Ren Z, Chen S, Ning B, Guo L. Use of Liver-Derived Cell Lines for the Study of Drug-Induced Liver Injury. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2018. [DOI: 10.1007/978-1-4939-7677-5_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
12
|
Tolosa L, Jiménez N, Pérez G, Castell JV, Gómez-Lechón MJ, Donato MT. Customised in vitro model to detect human metabolism-dependent idiosyncratic drug-induced liver injury. Arch Toxicol 2017; 92:383-399. [PMID: 28762043 PMCID: PMC5773651 DOI: 10.1007/s00204-017-2036-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/12/2017] [Indexed: 12/17/2022]
Abstract
Drug-induced liver injury (DILI) has a considerable impact on human health and is a major challenge in drug safety assessments. DILI is a frequent cause of liver injury and a leading reason for post-approval drug regulatory actions. Considerable variations in the expression levels of both cytochrome P450 (CYP) and conjugating enzymes have been described in humans, which could be responsible for increased susceptibility to DILI in some individuals. We herein explored the feasibility of the combined use of HepG2 cells co-transduced with multiple adenoviruses that encode drug-metabolising enzymes, and a high-content screening assay to evaluate metabolism-dependent drug toxicity and to identify metabolic phenotypes with increased susceptibility to DILI. To this end, HepG2 cells with different expression levels of specific drug-metabolism enzymes (CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4, GSTM1 and UGT2B7) were exposed to nine drugs with reported hepatotoxicity. A panel of pre-lethal mechanistic parameters (mitochondrial superoxide production, mitochondrial membrane potential, ROS production, intracellular calcium concentration, apoptotic nuclei) was used. Significant differences were observed according to the level of expression and/or the combination of several drug-metabolism enzymes in the cells created ad hoc according to the enzymes implicated in drug toxicity. Additionally, the main mechanisms implicated in the toxicity of the compounds were also determined showing also differences between the different types of cells employed. This screening tool allowed to mimic the variability in drug metabolism in the population and showed a highly efficient system for predicting human DILI, identifying the metabolic phenotypes associated with increased DILI risk, and indicating the mechanisms implicated in their toxicity.
Collapse
Affiliation(s)
- Laia Tolosa
- Unidad de Hepatología Experimental, Torre A, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, 46026, Valencia, Spain.
| | - Nuria Jiménez
- Unidad de Hepatología Experimental, Torre A, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Gabriela Pérez
- Unidad de Hepatología Experimental, Torre A, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - José V Castell
- Unidad de Hepatología Experimental, Torre A, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, 46026, Valencia, Spain.,Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, 46010, Valencia, Spain
| | - M José Gómez-Lechón
- Unidad de Hepatología Experimental, Torre A, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - M Teresa Donato
- Unidad de Hepatología Experimental, Torre A, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, 46026, Valencia, Spain. .,Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, 46010, Valencia, Spain.
| |
Collapse
|
13
|
Demazeau M, Quesnot N, Ripoche N, Rauch C, Jeftić J, Morel F, Gauffre F, Benvegnu T, Loyer P. Efficient transfection of Xenobiotic Responsive Element-biosensor plasmid using diether lipid and phosphatidylcholine liposomes in differentiated HepaRG cells. Int J Pharm 2017; 524:268-278. [PMID: 28365389 DOI: 10.1016/j.ijpharm.2017.03.080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 10/19/2022]
Abstract
In this study, we evaluated cationic liposomes prepared from diether-NH2 and egg phosphatidylcholine (EPC) for in vitro gene delivery. The impact of the lipid composition, i.e. the EPC and Diether-NH2 molar ratio, on in vitro transfection efficiency and cytotoxicity was investigated using the human HEK293T and hepatoma HepaRG cells known to be permissive and poorly permissive cells for liposome-mediated gene transfer, respectively. Here, we report that EPC/Diether-NH2-based liposomes enabled a very efficient transfection with low cytotoxicity compared to commercial transfection reagents in both HEK293T and proliferating progenitor HepaRG cells. Taking advantage of these non-toxic EPC/Diether-NH2-based liposomes, we developed a method to efficiently transfect differentiated hepatocyte-like HepaRG cells and a biosensor plasmid containing a Xenobiotic Responsive Element and a minimal promoter driving the transcription of the luciferase reporter gene. We demonstrated that the luciferase activity was induced by a canonical inducer of cytochrome P450 genes, the benzo[a]pyrene, and two environmental contaminants, the fluoranthene, a polycyclic aromatic hydrocarbon, and the endosulfan, an organochlorine insecticide, known to induce toxicity and genotoxicity in differentiated HepaRG cells. In conclusion, we established a new efficient lipofection-mediated gene transfer in hepatocyte-like HepaRG cells opening new perspectives in drug evaluation relying on xenobiotic inducible biosensor plasmids.
Collapse
Affiliation(s)
- Maxime Demazeau
- Ecole Nationale Supérieure de Chimie de Rennes, Institut des Sciences Chimiques de Rennes, UMR 6226 CNRS, Plateforme SynNanoVect, Biogenouest, 11 allée de Beaulieu, CS 50837, 35708 Rennes Cedex 7, France
| | - Nicolas Quesnot
- INSERM, INRA, Univ Rennes 1, Univ Bretagne Loire, Nutrition Metabolisms and Cancer (NuMeCan), Plateforme SynNanoVect, Biogenouest, Rennes, France
| | - Nicolas Ripoche
- Ecole Nationale Supérieure de Chimie de Rennes, Institut des Sciences Chimiques de Rennes, UMR 6226 CNRS, Plateforme SynNanoVect, Biogenouest, 11 allée de Beaulieu, CS 50837, 35708 Rennes Cedex 7, France
| | - Claudine Rauch
- INSERM, INRA, Univ Rennes 1, Univ Bretagne Loire, Nutrition Metabolisms and Cancer (NuMeCan), Plateforme SynNanoVect, Biogenouest, Rennes, France
| | - Jelena Jeftić
- Ecole Nationale Supérieure de Chimie de Rennes, Institut des Sciences Chimiques de Rennes, UMR 6226 CNRS, Plateforme SynNanoVect, Biogenouest, 11 allée de Beaulieu, CS 50837, 35708 Rennes Cedex 7, France
| | - Fabrice Morel
- INSERM, INRA, Univ Rennes 1, Univ Bretagne Loire, Nutrition Metabolisms and Cancer (NuMeCan), Plateforme SynNanoVect, Biogenouest, Rennes, France
| | - Fabienne Gauffre
- Institut des Sciences Chimiques de Rennes, UMR 6226 CNRS, Université de Rennes 1, Campus de Beaulieu, 263 Avenue du Général Leclerc, F-35042 Rennes Cedex, France.
| | - Thierry Benvegnu
- Ecole Nationale Supérieure de Chimie de Rennes, Institut des Sciences Chimiques de Rennes, UMR 6226 CNRS, Plateforme SynNanoVect, Biogenouest, 11 allée de Beaulieu, CS 50837, 35708 Rennes Cedex 7, France.
| | - Pascal Loyer
- INSERM, INRA, Univ Rennes 1, Univ Bretagne Loire, Nutrition Metabolisms and Cancer (NuMeCan), Plateforme SynNanoVect, Biogenouest, Rennes, France.
| |
Collapse
|
14
|
Gómez-Lechón MJ, Tolosa L, Donato MT. Upgrading HepG2 cells with adenoviral vectors that encode drug-metabolizing enzymes: application for drug hepatotoxicity testing. Expert Opin Drug Metab Toxicol 2016; 13:137-148. [PMID: 27671376 DOI: 10.1080/17425255.2017.1238459] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Drug attrition rates due to hepatotoxicity are an important safety issue considered in drug development. The HepG2 hepatoma cell line is currently being used for drug-induced hepatotoxicity evaluations, but its expression of drug-metabolizing enzymes is poor compared with hepatocytes. Different approaches have been proposed to upgrade HepG2 cells for more reliable drug-induced liver injury predictions. Areas covered: We describe the advantages and limitations of HepG2 cells transduced with adenoviral vectors that encode drug-metabolizing enzymes for safety risk assessments of bioactivable compounds. Adenoviral transduction facilitates efficient and controlled delivery of multiple drug-metabolizing activities to HepG2 cells at comparable levels to primary human hepatocytes by generating an 'artificial hepatocyte'. Furthermore, adenoviral transduction enables the design of tailored cells expressing particular metabolic capacities. Expert opinion: Upgraded HepG2 cells that recreate known inter-individual variations in hepatic CYP and conjugating activities due to both genetic (e.g., polymorphisms) or environmental (e.g., induction, inhibition) factors seems a suitable model to identify bioactivable drug and conduct hepatotoxicity risk assessments. This strategy should enable the generation of customized cells by reproducing human pheno- and genotypic CYP variability to represent a valuable human hepatic cell model to develop new safer drugs and to improve existing predictive toxicity assays.
Collapse
Affiliation(s)
- M José Gómez-Lechón
- a Unidad de Hepatología Experimental , Instituto de Investigación Sanitaria La Fe (IIS La Fe) , Valencia , Spain.,b CIBEREHD, FIS , Spain
| | - Laia Tolosa
- a Unidad de Hepatología Experimental , Instituto de Investigación Sanitaria La Fe (IIS La Fe) , Valencia , Spain
| | - M Teresa Donato
- a Unidad de Hepatología Experimental , Instituto de Investigación Sanitaria La Fe (IIS La Fe) , Valencia , Spain.,b CIBEREHD, FIS , Spain.,c Departamento de Bioquímica y Biología Molecular, Facultad de Medicina , Universidad de Valencia , Valencia , Spain
| |
Collapse
|
15
|
Huang L, Zou S, Deng J, Dai T, Jiang J, Jia Y, Dai R, Xie S. Development of an optimized cytotoxicity assay system for CYP3A4-mediated metabolic activation via modified piggyBac transposition. Toxicol In Vitro 2016; 32:132-7. [DOI: 10.1016/j.tiv.2015.12.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/12/2015] [Accepted: 12/13/2015] [Indexed: 12/25/2022]
|
16
|
Dayakar MM, Shivprasad D, Dayakar A, Deepthi CA. Assessment of oral health status among endosulfan victims in endosulfan relief and remediation cell - A cross-sectional survey. J Indian Soc Periodontol 2016; 19:709-11. [PMID: 26941528 PMCID: PMC4753722 DOI: 10.4103/0972-124x.156869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Endosulfan is a highly toxic agrichemical used in the cashew plantations. The Stockholm Convention held in April 2011 recommended a global ban on the manufacture and use of endosulfan because of its adverse effects on human health and the environment. Its impact on the quality of food, water, and beverages; and its ability to cause neurobehavioral disorders, congenital malformations in female subjects, and abnormalities related to the male reproductive system are studied, but however information regarding the oral health of endosulfan victims is scant. OBJECTIVES To assess the oral health status of the endosulfan victim in rehabilitation center. METHOD AND METHODOLOGY A cross sectional study on 18 subjects of 4-50 years of age were interviewed and examined using modified WHO oral health assessment proforma (1997) in Endosulfan Relief and Remediation Cell in Kokkada, Belthangady Taluk, Dakshina Kannada district, Karnataka, India. RESULTS Among the subjects, 10 (>50%) were found to be in age group <20 years. The overall oral health status of the endosulfan victim's in rehabilitation center considered to be poor, as many of the subjects suffered from major medical problems like mental retardation, physical disabilities etc. CONCLUSION This study emphasizes the need for special attention from government and voluntary organization to improve overall health status of the victims.
Collapse
Affiliation(s)
- Mundoor Manjunath Dayakar
- Department of Periodontics and Implantology, K V G Dental College and Hospital, Kurunjibagh, Sullia, Dakshina Kannada, Karnataka, India
| | - Dasappa Shivprasad
- Department of Periodontics and Implantology, K V G Dental College and Hospital, Kurunjibagh, Sullia, Dakshina Kannada, Karnataka, India
| | - Anitha Dayakar
- Department of Oral Pathology and Microbiology, K V G Dental College and Hospital, Kurunjibagh, Sullia, Dakshina Kannada, Karnataka, India
| | - Cherian Anna Deepthi
- Department of Periodontics and Implantology, K V G Dental College and Hospital, Kurunjibagh, Sullia, Dakshina Kannada, Karnataka, India
| |
Collapse
|
17
|
Ginkgo biloba leaf extract induces DNA damage by inhibiting topoisomerase II activity in human hepatic cells. Sci Rep 2015; 5:14633. [PMID: 26419945 PMCID: PMC4588569 DOI: 10.1038/srep14633] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 09/02/2015] [Indexed: 11/09/2022] Open
Abstract
Ginkgo biloba leaf extract has been shown to increase the incidence in liver tumors in mice in a 2-year bioassay conducted by the National Toxicology Program. In this study, the DNA damaging effects of Ginkgo biloba leaf extract and many of its constituents were evaluated in human hepatic HepG2 cells and the underlying mechanism was determined. A molecular docking study revealed that quercetin, a flavonoid constituent of Ginkgo biloba, showed a higher potential to interact with topoisomerase II (Topo II) than did the other Ginkgo biloba constituents; this in silico prediction was confirmed by using a biochemical assay to study Topo II enzyme inhibition. Moreover, as measured by the Comet assay and the induction of γ-H2A.X, quercetin, followed by keampferol and isorhamnetin, appeared to be the most potent DNA damage inducer in HepG2 cells. In Topo II knockdown cells, DNA damage triggered by Ginkgo biloba leaf extract or quercetin was dramatically decreased, indicating that DNA damage is directly associated with Topo II. DNA damage was also observed when cells were treated with commercially available Ginkgo biloba extract product. Our findings suggest that Ginkgo biloba leaf extract- and quercetin-induced in vitro genotoxicity may be the result of Topo II inhibition.
Collapse
|
18
|
Quesnot N, Rondel K, Audebert M, Martinais S, Glaise D, Morel F, Loyer P, Robin MA. Evaluation of genotoxicity using automated detection of γH2AX in metabolically competent HepaRG cells. Mutagenesis 2015; 31:43-50. [PMID: 26282955 DOI: 10.1093/mutage/gev059] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The in situ detection of γH2AX was recently reported to be a promising biomarker of genotoxicity. In addition, the human HepaRG hepatoma cells appear to be relevant for investigating hepatic genotoxicity since they express most of drug metabolizing enzymes and a wild type p53. The aim of this study was to determine whether the automated in situ detection of γH2AX positive HepaRG cells could be relevant for evaluation of genotoxicity after single or long-term repeated in vitro exposure compared to micronucleus assay. Metabolically competent HepaRG cells were treated daily with environmental contaminants and genotoxicity was evaluated after 1, 7 and 14 days. Using these cells, we confirmed the genotoxicity of aflatoxin B1 and benzo(a)pyrene and demonstrated that dimethylbenzanthracene, fipronil and endosulfan previously found genotoxic with comet or micronucleus assays also induced γH2AX phosphorylation. Furthermore, we showed that fluoranthene and bisphenol A induced γH2AX while no effect had been previously reported in HepG2 cells. In addition, induction of γH2AX was observed with some compounds only after 7 days, highlighting the importance of studying long-term effects of low doses of contaminants. Together, our data demonstrate that automated γH2AX detection in metabolically competent HepaRG cells is a suitable high-through put genotoxicity screening assay.
Collapse
Affiliation(s)
- Nicolas Quesnot
- Liver, Metabolisms and Cancer, INSERM, UMR991, CHU Pontchaillou, F-35033 Rennes, France, Université de Rennes 1, F-35043 Rennes, France and
| | - Karine Rondel
- Liver, Metabolisms and Cancer, INSERM, UMR991, CHU Pontchaillou, F-35033 Rennes, France, Université de Rennes 1, F-35043 Rennes, France and
| | - Marc Audebert
- Research Centre in Food Toxicology, INRA, UMR1331, Toxalim, F-31027 Toulouse, France
| | - Sophie Martinais
- Liver, Metabolisms and Cancer, INSERM, UMR991, CHU Pontchaillou, F-35033 Rennes, France, Université de Rennes 1, F-35043 Rennes, France and
| | - Denise Glaise
- Liver, Metabolisms and Cancer, INSERM, UMR991, CHU Pontchaillou, F-35033 Rennes, France, Université de Rennes 1, F-35043 Rennes, France and
| | - Fabrice Morel
- Liver, Metabolisms and Cancer, INSERM, UMR991, CHU Pontchaillou, F-35033 Rennes, France, Université de Rennes 1, F-35043 Rennes, France and
| | - Pascal Loyer
- Liver, Metabolisms and Cancer, INSERM, UMR991, CHU Pontchaillou, F-35033 Rennes, France, Université de Rennes 1, F-35043 Rennes, France and
| | - Marie-Anne Robin
- Liver, Metabolisms and Cancer, INSERM, UMR991, CHU Pontchaillou, F-35033 Rennes, France, Université de Rennes 1, F-35043 Rennes, France and
| |
Collapse
|
19
|
Gómez-Lechón MJ, Tolosa L, Conde I, Donato MT. Competency of different cell models to predict human hepatotoxic drugs. Expert Opin Drug Metab Toxicol 2014; 10:1553-68. [PMID: 25297626 DOI: 10.1517/17425255.2014.967680] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The liver is the most important target for drug-induced toxicity. This vulnerability results from functional liver features and its role in the metabolic elimination of most drugs. Drug-induced liver injury is a significant leading cause of acute, chronic liver disease and an important safety issue when developing new drugs. AREAS COVERED This review describes the advantages and limitations of hepatic cell-based models for early safety risk assessment during drug development. These models include hepatocytes cultured as monolayer, collagen-sandwich; emerging complex 3D configuration; liver-derived cell lines; stem cell-derived hepatocytes. EXPERT OPINION In vitro toxicity assays performed in hepatocytes or hepatoma cell lines can potentially provide rapid and cost-effective early feedback to identify toxic candidates for compound prioritization. However, their capacity to predict hepatotoxicity depends critically on cells' functional performance. In an attempt to improve and prolong functional properties of cultured cells, different strategies to recreate the in vivo hepatocyte environment have been explored. 3D cultures, co-cultures of hepatocytes with other cell types and microfluidic devices seem highly promising for toxicological studies. Moreover, hepatocytes derived from human pluripotent stem cells are emerging cell-based systems that may provide a stable source of hepatocytes to reliably screen metabolism and toxicity of candidate compounds.
Collapse
Affiliation(s)
- M José Gómez-Lechón
- Unidad de Hepatología Experimental Instituto de Investigación Sanitaria La Fe (IIS LA Fe) , Torre A Avda. Fernando Abril Martorell 106, 46026 Valencia , Spain +34 961246619 ;
| | | | | | | |
Collapse
|
20
|
Tu M, Li L, Lei H, Ma Z, Chen Z, Sun S, Xu S, Zhou H, Zeng S, Jiang H. Involvement of organic cation transporter 1 and CYP3A4 in retrorsine-induced toxicity. Toxicology 2014; 322:34-42. [PMID: 24799337 DOI: 10.1016/j.tox.2014.04.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 04/20/2014] [Accepted: 04/23/2014] [Indexed: 01/11/2023]
Abstract
Retrorsine (RTS) is a hepatotoxic pyrrolizidine alkaloid present in plants of the Senecio genus. The present study is aimed at clarifying the role of organic cation transporters (OCTs) in the liver disposition of RTS, and the coupling of OCT1 and cytochrome P450 (CYP) 3A4 in the hepatotoxicity of RTS. MDCK or LLC-PK1 cells stably expressing liver uptake or efflux transporters were used to investigate the interaction of RTS with these transporters. Primary cultured rat hepatocytes (PCRH) and double-transfected MDCK-hOCT1-CYP3A4 cells were used to determine the contribution of OCT1 and CYP3A4 to the toxicity of RTS. The results showed that RTS inhibited the OCT1-mediated 1-methyl-4-phenylpyridinium (MPP(+)) uptake in MDCK-hOCT1 cells with the IC50 of 2.25±0.30μM. The uptake of RTS in MDCK-hOCT1 cells and PCRH was significantly inhibited by OCT1 inhibitors, while hOCT3, human multidrug and toxin extrusion (hMATE) transporter 1, multidrug resistance 1 (MDR1), and breast cancer resistance protein (BCRP) showed weak or no obvious interaction with RTS. The toxic effect of RTS on the PCRH was attenuated by OCT1 inhibitors, quinidine and (+)-tetrahydropalmatine ((+)-THP). Compared to mock cells, MDCK-CYP3A4 cells showed a decrease in viability after being treated with RTS. Furthermore, RTS showed a more severe toxicity in the OCT1/CYP3A4 double-transfected cells compared to all other cells. Our data suggests that OCT1 mediates the liver-specific uptake of RTS, and plays an important role in RTS-induced hepatotoxicity together with CYP3A4. Consequently, the OCT1 inhibitors could be applied to protect the liver from the toxicity of RTS.
Collapse
Affiliation(s)
- Meijuan Tu
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Liping Li
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hongmei Lei
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zhiyuan Ma
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zhongjian Chen
- Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, Hangzhou, China
| | - Siyuan Sun
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Siyun Xu
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hui Zhou
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Su Zeng
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Huidi Jiang
- Laboratory of Pharmaceutical Analysis and Drug Metabolism, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
21
|
Li L, Tu M, Yang X, Sun S, Wu X, Zhou H, Zeng S, Jiang H. The Contribution of Human OCT1, OCT3, and CYP3A4 to Nitidine Chloride–Induced Hepatocellular Toxicity. Drug Metab Dispos 2014; 42:1227-34. [DOI: 10.1124/dmd.113.056689] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
22
|
Gonçalves TS, Menezes LMD, Trindade C, Machado MDS, Thomas P, Fenech M, Henriques JAP. Cytotoxicity and genotoxicity of orthodontic bands with or without silver soldered joints. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2014; 762:1-8. [DOI: 10.1016/j.mrgentox.2014.01.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 01/22/2014] [Accepted: 01/24/2014] [Indexed: 12/26/2022]
|
23
|
Vázquez-Boucard C, Anguiano-Vega G, Mercier L, Rojas del Castillo E. Pesticide residues, heavy metals, and DNA damage in sentinel oysters Crassostrea gigas from Sinaloa and Sonora, Mexico. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2014; 77:169-176. [PMID: 24555676 DOI: 10.1080/15287394.2013.853223] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Pesticides and heavy metals were analyzed in sentinel Crassostrea gigas oysters placed in six aquaculture sites close to a contaminated agricultural region. Each site was sampled twice. Tests revealed the presence of organochlorine (OC) pesticides in the oysters at concentrations varying from 31.8 to 72.5 μg/kg for gamma-hexachlorocyclohexane (γ-HCH); from 1.2 to 3.1 μg/kg for dichlorodiphenyldichloroethylene (4,4-DDE); from 1.6 to 2.3 μg/kg for endosulfan I; and from 1.4 to 41.2 μg/kg for endosulfan II, as well as heavy metals in concentrations that exceeded Mexican tolerance levels (405.5 to 987.8 μg/g for zinc; 4.2 to 7.3 μg/g for cadmium; and 7.2 to 9.9 μg/g for lead). Significant levels of DNA damage in oyster hemocytes were also detected. There was a significant, positive correlation between genotoxic damage and concentration of nickel or the presence of endosulfan II. Cellular viability evaluated by cytotoxic analyses was found to be high at 80%. Marked inhibition in activity of acetylcholinesterase (AChE ) and induction of glutathione S-transferase (GST) activity was noted. Data demonstrated a significant relation between AChE activity inhibition and presence of endosulfan II, γ-HCH, copper, lead, and 4,4-DDE, as well as between AChE and GST activity at different sites.
Collapse
Affiliation(s)
- Celia Vázquez-Boucard
- a Biochemistry and Molecular Laboratory, Department of Environmental Management and Conservation , Centro de Investigaciones Biológicas del Noroeste (CIBNOR), Instituto Politécnico Nacional #195 , La Paz , BCS , Mexico
| | | | | | | |
Collapse
|
24
|
Peyre L, Zucchini-Pascal N, de Sousa G, Rahmani R. Effects of endosulfan on hepatoma cell adhesion: Epithelial–mesenchymal transition and anoikis resistance. Toxicology 2012; 300:19-30. [DOI: 10.1016/j.tox.2012.05.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 05/10/2012] [Accepted: 05/12/2012] [Indexed: 12/12/2022]
|
25
|
Abstract
Considerable support exists for the roles of metabolism in modulating the carcinogenic properties of chemicals. In particular, many of these compounds are pro-carcinogens that require activation to electrophilic forms to exert genotoxic effects. We systematically analyzed the existing literature on the metabolism of carcinogens by human enzymes, which has been developed largely in the past 25 years. The metabolism and especially bioactivation of carcinogens are dominated by cytochrome P450 enzymes (66% of bioactivations). Within this group, six P450s--1A1, 1A2, 1B1, 2A6, 2E1, and 3A4--accounted for 77% of the P450 activation reactions. The roles of these P450s can be compared with those estimated for drug metabolism and should be considered in issues involving enzyme induction, chemoprevention, molecular epidemiology, interindividual variations, and risk assessment.
Collapse
|
26
|
Avlasevich S, Bryce S, De Boeck M, Elhajouji A, Van Goethem F, Lynch A, Nicolette J, Shi J, Dertinger S. Flow cytometric analysis of micronuclei in mammalian cell cultures: past, present and future. Mutagenesis 2010; 26:147-52. [DOI: 10.1093/mutage/geq058] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|