1
|
Gabr A, Mohamed AM, Abou Khalil NS, Sayed AEDH. The protective effect of Chlorella vulgaris against diclofenac toxicity in Clarias gariepinus: haemato-immunological parameters and spleen histological features as outcome markers. Front Immunol 2025; 16:1566496. [PMID: 40230852 PMCID: PMC11994428 DOI: 10.3389/fimmu.2025.1566496] [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: 01/24/2025] [Accepted: 03/10/2025] [Indexed: 04/16/2025] Open
Abstract
Introduction Diclofenac (DCF) is a commonly utilized medication in the non-steroidal anti-inflammatory drug category that is released into aquatic systems in significant amounts. Chlorella vulgaris (C. vulgaris) is rich in active phytochemicals known for their haemato-immunological boosting properties. Methods Our objective was to investigate the haemato-immunological protective properties of Chlorella in mitigating the toxic effects of DCF. Five groups of Clarias gariepinus, each comprising 36 fish, were assigned over a two-week period. The groups were assigned as follows: control group, which received a basal diet only; DCF1 group, which received a basal diet and was exposed to 20 μg/L of DCF; DCF2 group, which received a basal diet and was exposed to 10 mg/L of DCF; and Chlorella +DCF1 and Chlorella+DCF2 groups, which were exposed to the same DCF doses as Groups 2 and 3, respectively, while also being fed a diet containing 25% Chlorella. Results Exposure to both doses of DCF significantly decreased erythrocyte count, hemoglobin content, white blood cell count, phagocytic index, and lysozyme activity, while increased eosinophil and neutrophil % in an equipotent manner. The low dose caused a more pronounced reduction in packed cell volume (PCV)% and large lymphocyte% compared to the high dose. A significant decline in platelet count was observed only with the low DCF dose, while the high dose led to a decrease in monocyte%. DCF intoxication led to a dose-related decrease in small lymphocyte% and an increase in erythrocyte morphological alterations and interleukin (IL)-6 levels. The DCF2 group exhibited a higher increase in apoptotic RBCs than the DCF1 group. Intervention with Chlorella alongside the two DCF doses significantly normalized RBC count and eosinophil %, increased PCV% and small lymphocyte%, and decreased erythrocyte abnormalities to an equal extent. Large lymphocyte% in the Chlorella+DCF1 group was successfully restored to normal levels. Phagocytic index and lysozyme activity in the supplemented groups were lower, while IL-6 levels were higher than in the DCF groups. The percentage of apoptotic cells decreased with Chlorella administration, with the Chlorella+DCF1 group showing fewer apoptotic cells than the Chlorella+DCF2 group. Histopathological deterioration and excessive collagen deposition were observed in the spleen of DCF groups, while notable improvements were seen following C. vulgaris supplementation. Conclusion These findings suggest that dietary inclusion of C. vulgaris may antagonize the haemato-cytological abnormalities induced by DCF intoxication.
Collapse
Affiliation(s)
- Ahmed Gabr
- Molecular Biology Research & Studies Institute, Assiut University, Assiut, Egypt
| | - Amr M. Mohamed
- Molecular Biology Research & Studies Institute, Assiut University, Assiut, Egypt
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Nasser S. Abou Khalil
- Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt
- Department of Animal Physiology and Biochemistry, Faculty of Veterinary Medicine, Badr University, Assiut, Egypt
| | - Alaa El-Din H. Sayed
- Molecular Biology Research & Studies Institute, Assiut University, Assiut, Egypt
- Department of Zoology, Faculty of Science, Assiut University, Assiut, Egypt
| |
Collapse
|
2
|
Pu J, Feng W. Metabolic Stability and Metabolite Identification of CYP450 Probe Substrates in Ferret Hepatocytes. Curr Drug Metab 2025; 25:586-604. [PMID: 39252619 DOI: 10.2174/0113892002302675240903075500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 08/14/2024] [Accepted: 08/23/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Ferrets exhibit similar lung physiology to humans and display similar clinical signs following influenza infection, making them a valuable model for studying high susceptibility and infection patterns. However, the metabolic fate of several common human CYP450 probe substrates in ferrets is still unknown and has not been studied. OBJECTIVE The purpose of this study was to investigate the metabolism of nine human CYP450 probe substrates in ferret hepatocytes and explore their metabolic rate differences between ferrets and other species. METHOD Nine substrates were individually incubated in ferret hepatocytes for up to 120 min. At each time point, 30 μL mixtures were extracted for stability analysis using LC-MS/MS methods. After a 120-minute incubation period, 400 μL of the mixtures were extracted for metabolite identification using UHPLC-QExactive Plus. RESULTS The metabolic clearance was determined as follows: testosterone > phenacetin > bupropion > omeprazole > midazolam > dextromethorphan > chlorzoxazone > taxol > diclofenac. Seven metabolites were identified from phenacetin. Deethylation was found to be the major pathway, and the major metabolite was matched with acetaminophen as probed with the CYP1A2 enzyme. Six metabolites were identified from diclofenac. Glucuronidation was the primary pathway, and a metabolite was found to match 4-OH-diclofenac as probed with the CYP2C9 enzyme. Twenty-two metabolites were identified from omeprazole. The major metabolic pathways included mono-oxygenation and sulfoxide to thioether conversion. No metabolite was found to match with 5-OH-omeprazole as probed with the CYP2C19 enzyme. Twenty-two metabolites were identified from dextromethorphan. Demethylation was found to be the major metabolic pathway, and one demethylation metabolite was matched with dextrorphan as probed with the CYP2D6 enzyme. Fourteen metabolites were identified from midazolam. Mono-oxygenation was found to be the primary metabolic pathway, and one of the mono-oxygenation metabolites was matched with 1-OH-midazolam as probed with the CYP3A4 enzyme. Eight metabolites were identified from testosterone. Mono-oxygenation and glucuronidation were identified as the major metabolic pathways. One mono-oxygenation was matched with 6-.-testosterone as probed with the CYP3A4 enzyme. Six metabolites were identified from taxol. Hydrolysis and mono-oxygenation were the top two metabolic pathways. No metabolite was matched with 6-.-OH-taxol as probed with the CYP2C8 enzyme. Ten metabolites were identified from bupropion. Mono-oxygenation and hydrogenation were identified as the top two metabolic pathways. No mono-oxygenation metabolite was matched with hydroxy-bupropion as probed with the CYP2B6 enzyme. Nine metabolites were identified from chlorzoxazone. Monooxygenation and sulfation were the top two metabolic pathways. One mono-oxygenation metabolite was matched with 6-OH-chlorzoxazone as probed with the CYP2E1 enzyme. CONCLUSION Nine human CYP probe substrates were clearly metabolized in ferret hepatocytes, demonstrating substrate-dependent metabolic rates in ferret hepatocytes and species-dependent metabolic rates in mouse, rat, dog, monkey, and human hepatocytes. Except for 6-a-5-OH-omeprazole, 6-.-OH-taxol, and hydroxy-bupropion, specific metabolites of other six probe substrates in ferret hepatocytes were detected and identified as probed with six human CYP enzymes, respectively.
Collapse
Affiliation(s)
- Jiang Pu
- Shanghai Bioduro Biologics Co., Ltd, Shanghai, China
| | - Wanyong Feng
- Shanghai Bioduro Biologics Co., Ltd, Shanghai, China
| |
Collapse
|
3
|
Araújo AM, Carvalho F, Guedes de Pinho P, Carvalho M. Toxicometabolomics: Small Molecules to Answer Big Toxicological Questions. Metabolites 2021; 11:692. [PMID: 34677407 PMCID: PMC8539642 DOI: 10.3390/metabo11100692] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/17/2022] Open
Abstract
Given the high biological impact of classical and emerging toxicants, a sensitive and comprehensive assessment of the hazards and risks of these substances to organisms is urgently needed. In this sense, toxicometabolomics emerged as a new and growing field in life sciences, which use metabolomics to provide new sets of susceptibility, exposure, and/or effects biomarkers; and to characterize in detail the metabolic responses and altered biological pathways that various stressful stimuli cause in many organisms. The present review focuses on the analytical platforms and the typical workflow employed in toxicometabolomic studies, and gives an overview of recent exploratory research that applied metabolomics in various areas of toxicology.
Collapse
Affiliation(s)
- Ana Margarida Araújo
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (F.C.); (P.G.d.P.)
- UCIBIO—Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira nº228, 4050-313 Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (F.C.); (P.G.d.P.)
- UCIBIO—Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira nº228, 4050-313 Porto, Portugal
| | - Paula Guedes de Pinho
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (F.C.); (P.G.d.P.)
- UCIBIO—Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira nº228, 4050-313 Porto, Portugal
| | - Márcia Carvalho
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (F.C.); (P.G.d.P.)
- UCIBIO—Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira nº228, 4050-313 Porto, Portugal
- FP-I3ID, FP-ENAS, University Fernando Pessoa, Praça 9 de Abril, 349, 4249-004 Porto, Portugal
- Faculty of Health Sciences, University Fernando Pessoa, Rua Carlos da Maia, 296, 4200-150 Porto, Portugal
| |
Collapse
|
4
|
A serum-free medium suitable for maintaining cell morphology and liver-specific function in induced human hepatocytes. Cytotechnology 2019; 71:329-344. [PMID: 30603919 DOI: 10.1007/s10616-018-0289-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 12/05/2018] [Indexed: 12/15/2022] Open
Abstract
hiHep is a new type of hepatocyte-like cell that is predicted to be a potential unlimited source of hepatocytes for a bioartificial liver. However, hiHep cannot currently be used in clinical settings because serum must be added during the culture process. Thus, a defined medium is required. Because serum is complex, an efficient statistical approach based on the Plackett-Burman design was used. In this manner, an original medium and several significant cell growth factors were identified. These factors include insulin, VH, and VE, and the original medium was optimized based on these significant factors. Additionally, hiHep liver-specific functions and metabolism in the optimized serum-free medium were measured. Results showed that hiHep functions, such as glycogen storage, albumin secretion, and urea production, were well maintained in our optimized serum-free medium. In summary, we created a chemically defined, serum-free medium in which cell growth, proliferation, metabolism, and function were well maintained. This medium has the potential to support the clinical use of hiHep.
Collapse
|
5
|
Han W, Wu Q, Zhang X, Duan Z. Innovation for hepatotoxicity in vitro research models: A review. J Appl Toxicol 2018; 39:146-162. [PMID: 30182494 DOI: 10.1002/jat.3711] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/11/2018] [Accepted: 07/13/2018] [Indexed: 12/18/2022]
Abstract
Many categories of drugs can induce hepatotoxicity, so improving the prediction of toxic drugs is important. In vitro models using human hepatocytes are more accurate than in vivo animal models. Good in vitro models require an abundance of metabolic enzyme activities and normal cellular polarity. However, none of the in vitro models can completely simulate hepatocytes in the human body. There are two ways to overcome this limitation: enhancing the metabolic function of hepatocytes and changing the cultural environment. In this review, we summarize the current state of research, including the main characteristics of in vitro models and their limitations, as well as improved technology and developmental prospects. We hope that this review provides some new ideas for hepatotoxicity research.
Collapse
Affiliation(s)
- Weijia Han
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| | - Qiao Wu
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| | - Xiaohui Zhang
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| | - Zhongping Duan
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| |
Collapse
|
6
|
Knöspel F, Jacobs F, Freyer N, Damm G, De Bondt A, van den Wyngaert I, Snoeys J, Monshouwer M, Richter M, Strahl N, Seehofer D, Zeilinger K. In Vitro Model for Hepatotoxicity Studies Based on Primary Human Hepatocyte Cultivation in a Perfused 3D Bioreactor System. Int J Mol Sci 2016; 17:584. [PMID: 27092500 PMCID: PMC4849040 DOI: 10.3390/ijms17040584] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 03/24/2016] [Accepted: 04/12/2016] [Indexed: 01/02/2023] Open
Abstract
Accurate prediction of the potential hepatotoxic nature of new pharmaceuticals remains highly challenging. Therefore, novel in vitro models with improved external validity are needed to investigate hepatic metabolism and timely identify any toxicity of drugs in humans. In this study, we examined the effects of diclofenac, as a model substance with a known risk of hepatotoxicity in vivo, in a dynamic multi-compartment bioreactor using primary human liver cells. Biotransformation pathways of the drug and possible effects on metabolic activities, morphology and cell transcriptome were evaluated. Formation rates of diclofenac metabolites were relatively stable over the application period of seven days in bioreactors exposed to 300 µM diclofenac (300 µM bioreactors (300 µM BR)), while in bioreactors exposed to 1000 µM diclofenac (1000 µM BR) metabolite concentrations declined drastically. The biochemical data showed a significant decrease in lactate production and for the higher dose a significant increase in ammonia secretion, indicating a dose-dependent effect of diclofenac application. The microarray analyses performed revealed a stable hepatic phenotype of the cells over time and the observed transcriptional changes were in line with functional readouts of the system. In conclusion, the data highlight the suitability of the bioreactor technology for studying the hepatotoxicity of drugs in vitro.
Collapse
Affiliation(s)
- Fanny Knöspel
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany.
| | - Frank Jacobs
- Janssen Research & Development, Beerse 2340, Belgium.
| | - Nora Freyer
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany.
| | - Georg Damm
- Department for General, Visceral and Transplantation Surgery, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany.
| | - An De Bondt
- Janssen Research & Development, Beerse 2340, Belgium.
| | | | - Jan Snoeys
- Janssen Research & Development, Beerse 2340, Belgium.
| | | | - Marco Richter
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany.
| | - Nadja Strahl
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany.
| | - Daniel Seehofer
- Department for General, Visceral and Transplantation Surgery, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany.
| | - Katrin Zeilinger
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany.
| |
Collapse
|
7
|
Mitra P, Yang RM, Sutton J, Ramsay RG, Gonda TJ. CDK9 inhibitors selectively target estrogen receptor-positive breast cancer cells through combined inhibition of MYB and MCL-1 expression. Oncotarget 2016; 7:9069-83. [PMID: 26812885 PMCID: PMC4891027 DOI: 10.18632/oncotarget.6997] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/17/2016] [Indexed: 11/25/2022] Open
Abstract
Our previous studies showed that MYB is required for proliferation of, and confers protection against apoptosis on, estrogen receptor-positive (ER(+ve)) breast cancer cells, which are almost invariably also MYB(+ve). We have also shown that MYB expression in ER(+ve) breast cancer cells is regulated at the level of transcriptional elongation and as such, is suppressed by CDK9i. Here we examined the effects of CDK9i on breast cancer cells and the involvement of MYB in these effects. ER(+ve) breast cancer cell lines including MCF-7 were much more sensitive (> 10 times) to killing by CDK9i than ER(-ve)/MYB(-ve) cells. Moreover, surviving cells showed a block at the G2/M phase of the cell cycle. Importantly, ectopic MYB expression conferred resistance to apoptosis induction, cell killing and G2/M accumulation. Expression of relevant MYB target genes including BCL2 and CCNB1 was suppressed by CDK9 inhibition, and this too was reversed by ectopic MYB expression. Nevertheless, inhibition of BCL2 alone either by MYB knockdown or by ABT-199 treatment was insufficient for significant induction of apoptosis. Further studies implied that suppression of MCL-1, a well-documented target of CDK9 inhibition, was additionally required for apoptosis induction, while maximal levels of apoptosis induced by CDK9i are likely to also involve inhibition of BCL2L1 expression. Taken together these data suggest that MYB regulation of BCL2 underlies the heightened sensitivity of ER(+ve) compared to ER(-ve) breast cancer cells to CDK9 inhibition, and that these compounds represent a potential therapeutic for ER(+ve) breast cancers and possibly other MYB-dependent cancers.
Collapse
Affiliation(s)
- Partha Mitra
- School of Pharmacy, University of Queensland, Brisbane, QLD, Australia
| | - Ren-Ming Yang
- School of Pharmacy, University of Queensland, Brisbane, QLD, Australia
| | - James Sutton
- Novartis Institute for Biomedical Research, Emeryville, CA, USA
| | - Robert G. Ramsay
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Thomas J. Gonda
- School of Pharmacy, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
8
|
Van den Eede N, Cuykx M, Rodrigues RM, Laukens K, Neels H, Covaci A, Vanhaecke T. Metabolomics analysis of the toxicity pathways of triphenyl phosphate in HepaRG cells and comparison to oxidative stress mechanisms caused by acetaminophen. Toxicol In Vitro 2015; 29:2045-54. [DOI: 10.1016/j.tiv.2015.08.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 05/15/2015] [Accepted: 08/14/2015] [Indexed: 12/21/2022]
|
9
|
Broeders JJ, Parmentier C, Truisi GL, Jossé R, Alexandre E, Savary CC, Hewitt PG, Mueller SO, Guillouzo A, Richert L, van Eijkeren JC, Hermens JL, Blaauboer BJ. Biokinetics of chlorpromazine in primary rat and human hepatocytes and human HepaRG cells after repeated exposure. Toxicol In Vitro 2015; 30:52-61. [DOI: 10.1016/j.tiv.2014.08.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Revised: 08/03/2014] [Accepted: 08/21/2014] [Indexed: 01/01/2023]
|
10
|
Noor F. A shift in paradigm towards human biology-based systems for cholestatic-liver diseases. J Physiol 2015; 593:5043-55. [PMID: 26417843 DOI: 10.1113/jp271124] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 09/16/2015] [Indexed: 12/15/2022] Open
Abstract
Cholestatic-liver diseases (CLDs) arise from diverse causes ranging from genetic factors to drug-induced cholestasis. The so-called diseases of civilization (obesity, diabetes, metabolic disorders, non-alcoholic liver disease, cardiovascular diseases, etc.) are intricately implicated in liver and gall bladder diseases. Although CLDs have been extensively studied, there seem to be important gaps in the understanding of human disease. Despite the fact that many animal models exist and substantial clinical data are available, translation of this knowledge towards therapy has been disappointingly limited. Recent advances in liver cell culture such as in vivo-like 3D cultivation of human primary hepatic cells, human induced pluripotent stem cell-derived hepatocytes; and cutting-edge analytical techniques such as 'omics' technologies and high-content screenings could play a decisive role in deeper mechanistic understanding of CLDs. This Topical Review proposes a roadmap to human biology-based research using omics technologies providing quantitative information on mechanisms in an adverse outcome/disease pathway framework. With modern sensitive tools, a shift in paradigm in human disease research seems timely and even inevitable to overcome species barriers in translation.
Collapse
Affiliation(s)
- Fozia Noor
- Biochemical Engineering Institute, Saarland University, Saarbrücken, Germany
| |
Collapse
|
11
|
Klein S, Maggioni S, Bucher J, Mueller D, Niklas J, Shevchenko V, Mauch K, Heinzle E, Noor F. In Silico Modeling for the Prediction of Dose and Pathway-Related Adverse Effects in Humans From In Vitro Repeated-Dose Studies. Toxicol Sci 2015; 149:55-66. [PMID: 26420750 DOI: 10.1093/toxsci/kfv218] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Long-term repeated-dose toxicity is mainly assessed in animals despite poor concordance of animal data with human toxicity. Nowadays advanced human in vitro systems, eg, metabolically competent HepaRG cells, are used for toxicity screening. Extrapolation of in vitro toxicity to in vivo effects is possible by reverse dosimetry using pharmacokinetic modeling. We assessed long-term repeated-dose toxicity of bosentan and valproic acid (VPA) in HepaRG cells under serum-free conditions. Upon 28-day exposure, the EC50 values for bosentan and VPA decreased by 21- and 33-fold, respectively. Using EC(10) as lowest threshold of toxicity in vitro, we estimated the oral equivalent doses for both test compounds using a simplified pharmacokinetic model for the extrapolation of in vitro toxicity to in vivo effect. The model predicts that bosentan is safe at the considered dose under the assumed conditions upon 4 weeks exposure. For VPA, hepatotoxicity is predicted for 4% and 47% of the virtual population at the maximum recommended daily dose after 3 and 4 weeks of exposure, respectively. We also investigated the changes in the central carbon metabolism of HepaRG cells exposed to orally bioavailable concentrations of both drugs. These concentrations are below the 28-day EC(10) and induce significant changes especially in glucose metabolism and urea production. These metabolic changes may have a pronounced impact in susceptible patients such as those with compromised liver function and urea cycle deficiency leading to idiosyncratic toxicity. We show that the combination of modeling based on in vitro repeated-dose data and metabolic changes allows the prediction of human relevant in vivo toxicity with mechanistic insights.
Collapse
Affiliation(s)
- Sebastian Klein
- *Biochemical Engineering, Saarland University, 66123 Saarbruecken, Germany
| | - Silvia Maggioni
- IRCCS - Instituto di Ricerche Farmacologiche "Mario Negri," 20156 Milan, Italy
| | - Joachim Bucher
- Insilico Biotechnology AG, 70563 Stuttgart, Germany, and
| | - Daniel Mueller
- *Biochemical Engineering, Saarland University, 66123 Saarbruecken, Germany
| | - Jens Niklas
- Insilico Biotechnology AG, 70563 Stuttgart, Germany, and
| | | | - Klaus Mauch
- Insilico Biotechnology AG, 70563 Stuttgart, Germany, and
| | - Elmar Heinzle
- *Biochemical Engineering, Saarland University, 66123 Saarbruecken, Germany
| | - Fozia Noor
- *Biochemical Engineering, Saarland University, 66123 Saarbruecken, Germany,
| |
Collapse
|
12
|
Kegel V, Pfeiffer E, Burkhardt B, Liu JL, Zeilinger K, Nüssler AK, Seehofer D, Damm G. Subtoxic Concentrations of Hepatotoxic Drugs Lead to Kupffer Cell Activation in a Human In Vitro Liver Model: An Approach to Study DILI. Mediators Inflamm 2015; 2015:640631. [PMID: 26491234 PMCID: PMC4600928 DOI: 10.1155/2015/640631] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 01/14/2015] [Accepted: 01/15/2015] [Indexed: 12/15/2022] Open
Abstract
Drug induced liver injury (DILI) is an idiosyncratic adverse drug reaction leading to severe liver damage. Kupffer cells (KC) sense hepatic tissue stress/damage and therefore could be a tool for the estimation of consequent effects associated with DILI. Aim of the present study was to establish a human in vitro liver model for the investigation of immune-mediated signaling in the pathogenesis of DILI. Hepatocytes and KC were isolated from human liver specimens. The isolated KC yield was 1.2 ± 0.9 × 10(6) cells/g liver tissue with a purity of >80%. KC activation was investigated by the measurement of reactive oxygen intermediates (ROI, DCF assay) and cell activity (XTT assay). The initial KC activation levels showed broad donor variability. Additional activation of KC using supernatants of hepatocytes treated with hepatotoxic drugs increased KC activity and led to donor-dependent changes in the formation of ROI compared to KC incubated with supernatants from untreated hepatocytes. Additionally, a compound- and donor-dependent increase in proinflammatory cytokines or in anti-inflammatory cytokines was detected. In conclusion, KC related immune signaling in hepatotoxicity was successfully determined in a newly established in vitro liver model. KC were able to detect hepatocyte stress/damage and to transmit a donor- and compound-dependent immune response via cytokine production.
Collapse
Affiliation(s)
- Victoria Kegel
- Department of General, Visceral and Transplantation Surgery, Charité-University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Elisa Pfeiffer
- Department of General, Visceral and Transplantation Surgery, Charité-University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Britta Burkhardt
- BG Trauma Center, Siegfried Weller Institute, Eberhard Karls University Tübingen, Schnarrenbergstrasse 95, 72076 Tübingen, Germany
| | - Jia L. Liu
- Department of General, Visceral and Transplantation Surgery, Charité-University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Katrin Zeilinger
- Bioreactor Group, Berlin-Brandenburg Centre for Regenerative Therapies (BCRT), Charité-University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Andreas K. Nüssler
- BG Trauma Center, Siegfried Weller Institute, Eberhard Karls University Tübingen, Schnarrenbergstrasse 95, 72076 Tübingen, Germany
| | - Daniel Seehofer
- Department of General, Visceral and Transplantation Surgery, Charité-University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Georg Damm
- Department of General, Visceral and Transplantation Surgery, Charité-University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
13
|
Alépée N, Bahinski A, Daneshian M, De Wever B, Fritsche E, Goldberg A, Hansmann J, Hartung T, Haycock J, Hogberg H, Hoelting L, Kelm JM, Kadereit S, McVey E, Landsiedel R, Leist M, Lübberstedt M, Noor F, Pellevoisin C, Petersohn D, Pfannenbecker U, Reisinger K, Ramirez T, Rothen-Rutishauser B, Schäfer-Korting M, Zeilinger K, Zurich MG. State-of-the-art of 3D cultures (organs-on-a-chip) in safety testing and pathophysiology. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2014. [PMID: 25027500 DOI: 10.14573/altex1406111] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Integrated approaches using different in vitro methods in combination with bioinformatics can (i) increase the success rate and speed of drug development; (ii) improve the accuracy of toxicological risk assessment; and (iii) increase our understanding of disease. Three-dimensional (3D) cell culture models are important building blocks of this strategy which has emerged during the last years. The majority of these models are organotypic, i.e., they aim to reproduce major functions of an organ or organ system. This implies in many cases that more than one cell type forms the 3D structure, and often matrix elements play an important role. This review summarizes the state of the art concerning commonalities of the different models. For instance, the theory of mass transport/metabolite exchange in 3D systems and the special analytical requirements for test endpoints in organotypic cultures are discussed in detail. In the next part, 3D model systems for selected organs--liver, lung, skin, brain--are presented and characterized in dedicated chapters. Also, 3D approaches to the modeling of tumors are presented and discussed. All chapters give a historical background, illustrate the large variety of approaches, and highlight up- and downsides as well as specific requirements. Moreover, they refer to the application in disease modeling, drug discovery and safety assessment. Finally, consensus recommendations indicate a roadmap for the successful implementation of 3D models in routine screening. It is expected that the use of such models will accelerate progress by reducing error rates and wrong predictions from compound testing.
Collapse
|
14
|
Alépée N, Bahinski A, Daneshian M, De Wever B, Fritsche E, Goldberg A, Hansmann J, Hartung T, Haycock J, Hogberg HT, Hoelting L, Kelm JM, Kadereit S, McVey E, Landsiedel R, Leist M, Lübberstedt M, Noor F, Pellevoisin C, Petersohn D, Pfannenbecker U, Reisinger K, Ramirez T, Rothen-Rutishauser B, Schäfer-Korting M, Zeilinger K, Zurich MG. State-of-the-art of 3D cultures (organs-on-a-chip) in safety testing and pathophysiology. ALTEX 2014; 31:441-77. [PMID: 25027500 PMCID: PMC4783151 DOI: 10.14573/altex.1406111] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 06/30/2014] [Indexed: 02/02/2023]
Abstract
Integrated approaches using different in vitro methods in combination with bioinformatics can (i) increase the success rate and speed of drug development; (ii) improve the accuracy of toxicological risk assessment; and (iii) increase our understanding of disease. Three-dimensional (3D) cell culture models are important building blocks of this strategy which has emerged during the last years. The majority of these models are organotypic, i.e., they aim to reproduce major functions of an organ or organ system. This implies in many cases that more than one cell type forms the 3D structure, and often matrix elements play an important role. This review summarizes the state of the art concerning commonalities of the different models. For instance, the theory of mass transport/metabolite exchange in 3D systems and the special analytical requirements for test endpoints in organotypic cultures are discussed in detail. In the next part, 3D model systems for selected organs--liver, lung, skin, brain--are presented and characterized in dedicated chapters. Also, 3D approaches to the modeling of tumors are presented and discussed. All chapters give a historical background, illustrate the large variety of approaches, and highlight up- and downsides as well as specific requirements. Moreover, they refer to the application in disease modeling, drug discovery and safety assessment. Finally, consensus recommendations indicate a roadmap for the successful implementation of 3D models in routine screening. It is expected that the use of such models will accelerate progress by reducing error rates and wrong predictions from compound testing.
Collapse
Affiliation(s)
| | - Anthony Bahinski
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, USA
| | - Mardas Daneshian
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany
| | | | - Ellen Fritsche
- Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Alan Goldberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - Jan Hansmann
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Thomas Hartung
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany,Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - John Haycock
- Department of Materials Science of Engineering, University of Sheffield, Sheffield, UK
| | - Helena T. Hogberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - Lisa Hoelting
- Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | | | - Suzanne Kadereit
- Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Emily McVey
- Board for the Authorization of Plant Protection Products and Biocides, Wageningen, The Netherlands
| | | | - Marcel Leist
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany,Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Marc Lübberstedt
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Fozia Noor
- Biochemical Engineering, Saarland University, Saarbruecken, Germany
| | | | | | | | | | - Tzutzuy Ramirez
- BASF SE, Experimental Toxicology and Ecology, Ludwigshafen, Germany
| | | | - Monika Schäfer-Korting
- Institute for Pharmacy (Pharmacology and Toxicology), Freie Universität Berlin, Berlin, Germany
| | - Katrin Zeilinger
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Marie-Gabriele Zurich
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland,Swiss Center for Applied Human Toxicology (SCAHT), University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
15
|
Parmentier C, Truisi GL, Moenks K, Stanzel S, Lukas A, Kopp-Schneider A, Alexandre E, Hewitt PG, Mueller SO, Richert L. Transcriptomic hepatotoxicity signature of chlorpromazine after short- and long-term exposure in primary human sandwich cultures. Drug Metab Dispos 2013; 41:1835-42. [PMID: 23913027 DOI: 10.1124/dmd.113.052415] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Drug-induced liver injury is the most frequent reason for market withdrawal of approved drugs, and is difficult to predict in animal models. Here, we analyzed transcriptomic data derived from short- and long-term cultured primary human hepatocytes (PHH) exposed to the well known human hepatotoxin chlorpromazine (CPZ). Samples were collected from five PHH cultures after short-term (1 and 3 days) and long-term (14 days) repeat daily treatment with 0.1 or 0.2 µM CPZ, corresponding to C(max). Two PHH cultures were additionally treated with 1 µM CPZ, and the three others with 0.02 µM CPZ. Differences in the total number of gene changes were seen between donors and throughout treatment. Specific transcriptomic hepatotoxicity signatures were created for CPZ and consisted of inflammation/hepatitis, cholestasis, and liver proliferation in all five donors, as well as fibrosis and steatosis, which were observed in four of five donors. Necrosis was present in three of five donors, and an indicative signature of cirrhosis was observed after long-term 14-day repeat treatment, also in three of five donors. The inter-donor variability in the inflammatory response to CPZ treatment was associated with variability in the strength of the response of the transcriptomic hepatotoxicity signatures, suggesting that features of inflammation could be related to the idiosyncratic hepatotoxic effects of CPZ in humans.
Collapse
Affiliation(s)
- Céline Parmentier
- KaLy-Cell, Plobsheim, France (C.P., E.A., L.R.); Non-Clinical Safety, Merck Serono, Merck KGaA, Darmstadt, Germany (G.L.T., P.G.H., S.O.M.); Emergentec Biodevelopment GmbH, Vienna, Austria (K.M., A.L.); Department of Biostatistics, German Cancer Research Center, Heidelberg, Germany (S.S., A.K.-S.); Institut für Angewandte Biowissenschaften, Karlsruhe Institute of Technology, Karlsruhe, Germany (G.L.T., S.O.M.); and Laboratoire de Toxicologie Cellulaire, EA4267 Université de Franche-Comté, Besançon, France (L.R.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Klein S, Mueller D, Schevchenko V, Noor F. Long-term maintenance of HepaRG cells in serum-free conditions and application in a repeated dose study. J Appl Toxicol 2013; 34:1078-86. [DOI: 10.1002/jat.2929] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 07/28/2013] [Accepted: 08/08/2013] [Indexed: 11/10/2022]
Affiliation(s)
- Sebastian Klein
- Biochemical Engineering; Saarland University; 66123 Saarbruecken Germany
| | - Daniel Mueller
- Biochemical Engineering; Saarland University; 66123 Saarbruecken Germany
| | | | - Fozia Noor
- Biochemical Engineering; Saarland University; 66123 Saarbruecken Germany
| |
Collapse
|
17
|
Mueller D, Tascher G, Damm G, Nüssler AK, Heinzle E, Noor F. Real-time in situ viability assessment in a 3D bioreactor with liver cells using resazurin assay. Cytotechnology 2013; 65:297-305. [PMID: 22828753 PMCID: PMC3560875 DOI: 10.1007/s10616-012-9486-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 07/10/2012] [Indexed: 01/18/2023] Open
Abstract
Three-dimensional cultivation of human cells is promising especially for long-term maintenance of specific functions and mimicking the in vivo tissue environment. However, direct viability assessment is very difficult in such systems. Commonly applied indirect methods such as glucose consumption, albumin or urea production are greatly affected by culture conditions, stress and time of cultivation and do not reflect the real time viability of the cells. In this study we established a real-time in situ viability assay namely; resazurin assay, in a 3D hollow-fiber bioreactor using human liver cells. Resazurin assay is based on the conversion of resazurin to a fluorescent dye by cytoplasmatic and mitochondrial enzymes. We show that the resazurin reagent in concentrations used in this study is non-toxic and could be rapidly removed out of the system. Moreover, we observed that dead cells do not affect the results of the assay. We optimized the assay on HepG2 cells and tested it with primary human hepatocytes. Moreover, we maintained primary human hepatocytes in the 3D bioreactor system in serum-free conditions and also assessed viability before and after the exposure to amiodarone using the resazurin assay. We show that this approach is applicable during long-term cultivation of cells in bioreactors under different conditions and can moreover be applied to pharmacological studies, e.g. investigation of chronic drug effects in such 3D bioreactors.
Collapse
Affiliation(s)
- Daniel Mueller
- />Biochemical Engineering Institute, Saarland University, Geb. A1 5, 66123 Saarbruecken, Germany
| | - Georg Tascher
- />Biochemical Engineering Institute, Saarland University, Geb. A1 5, 66123 Saarbruecken, Germany
| | - Georg Damm
- />Department of General-, Visceral- and Transplantation Surgery, Charité Medical University Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Andreas K. Nüssler
- />Department of Trauma Surgery, Eberhard-Karls University Tubingen, Schnarrenbergstr. 95, 72076 Tubingen, Germany
| | - Elmar Heinzle
- />Biochemical Engineering Institute, Saarland University, Geb. A1 5, 66123 Saarbruecken, Germany
| | - Fozia Noor
- />Biochemical Engineering Institute, Saarland University, Geb. A1 5, 66123 Saarbruecken, Germany
| |
Collapse
|