1
|
Laghezza A, Falbo E, Gilardi F, Thomas A, Brunetti L, Leuci R, Piemontese L, Tortorella P, Biswas A, Singh RP, Pattnaik AK, Jayaprakash V, Tambe S, Ca S, Wackerlig-Damle J, Paoli P, Loiodice F, Lavecchia A. A new potent and selective peroxisome proliferator-activated receptor alpha partial agonist displays anti-steatotic effects In vitro and behaves as a safe hypolipidemic and hypoglycemic agent in a diabetic mouse model. Eur J Med Chem 2025; 289:117494. [PMID: 40088662 DOI: 10.1016/j.ejmech.2025.117494] [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: 02/26/2025] [Accepted: 03/08/2025] [Indexed: 03/17/2025]
Abstract
A rational drug design approach led to the synthesis of three pairs of enantiomers derived from the peroxisome proliferator-activated receptor (PPAR) pan agonist AL29-26, identifying (S)-2 as a potent and selective PPARα partial agonist. Molecular docking and molecular dynamics simulations elucidated the binding modes of (S)-2 within the ligand-binding domains of PPARα and PPARγ. In vitro, (S)-2 demonstrated significant anti-steatotic effects, upregulating key PPARα target genes involved in lipid metabolism. In vivo, (S)-2 exhibited hypolipidemic and antihyperglycemic activity in a diabetic mouse model, outperforming fenofibrate in lowering blood glucose and lipid levels, while showing no toxicity in major organs (artery, kidney, liver, pancreas). The therapeutic effects of ((S)-2 were attributed to its PPARα selectivity, reduced activation of PPARγ, and mild protein tyrosine phosphatase 1B (PTP1B) inhibition. These findings highlight (S)-2 as a promising lead compound for the development of safer and more effective treatments for dyslipidemic type 2 diabetes.
Collapse
Affiliation(s)
- Antonio Laghezza
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro, 70125, Bari, Italy
| | - Emanuele Falbo
- Department of Pharmacy, "Drug Discovery" Laboratory, University of Napoli Federico II, 80131, Napoli, Italy
| | - Federica Gilardi
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland; Unit of Forensic Toxicology and Chemistry, CURML, Lausanne and Geneva University Hospitals, Lausanne-Geneva, Switzerland
| | - Aurélien Thomas
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland; Unit of Forensic Toxicology and Chemistry, CURML, Lausanne and Geneva University Hospitals, Lausanne-Geneva, Switzerland
| | - Leonardo Brunetti
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro, 70125, Bari, Italy
| | - Rosalba Leuci
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro, 70125, Bari, Italy
| | - Luca Piemontese
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro, 70125, Bari, Italy
| | - Paolo Tortorella
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro, 70125, Bari, Italy
| | - Abanish Biswas
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Ravi Pratap Singh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Ashok Kumar Pattnaik
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Venkatesan Jayaprakash
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Suhas Tambe
- Adgyl Lifesciences Private Ltd, Bengaluru, Karnataka, 560058, India
| | - Sudeep Ca
- Bioanalytical Section, Eurofins Advinus Biopharma Services India Pvt Ltd., Bengaluru, Karnataka, 560058, India
| | - Judith Wackerlig-Damle
- Department of Pharmaceutical Sciences, Faculty of Life Sciences, University of Vienna, 1090, Vienna, Austria
| | - Paolo Paoli
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Sezione di Scienze Biochimiche, Università degli Studi di Firenze, viale Morgagni 50, 50134, Firenze, Italy
| | - Fulvio Loiodice
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro, 70125, Bari, Italy.
| | - Antonio Lavecchia
- Department of Pharmacy, "Drug Discovery" Laboratory, University of Napoli Federico II, 80131, Napoli, Italy.
| |
Collapse
|
2
|
Seo JE, He X, Bryant M, Atrakchi AH, McGovern TJ, Davis Bruno KL, Heflich RH, Guo X. Comparative DNA damage induced by eight nitrosamines in primary human and macaque hepatocytes. Chem Biol Interact 2025; 416:111538. [PMID: 40319998 DOI: 10.1016/j.cbi.2025.111538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/15/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
N-nitrosamines have been increasingly detected in human drugs, raising serious safety concerns due to their potential mutagenicity and carcinogenicity. In order to expand upon the human data available on these drug impurities, we previously used metabolically competent HepaRG human hepatoma cells to evaluate the genotoxicity of eight small-molecule nitrosamines [N-cyclopentyl-4-nitrosopiperazine (CPNP), N-nitrosodibutylamine (NDBA), N-nitrosodiethylamine (NDEA), N-nitrosodimethylamine (NDMA), N-nitrosodiisopropylamine (NDIPA), N-nitrosoethylisopropylamine (NEIPA), N-nitroso-N-methyl-4-aminobutyric acid (NMBA), and N-nitrosomethylphenylamine (NMPA)]. In this study, we used the comet assay to further investigate the DNA damage induced by the eight nitrosamines in primary human hepatocytes (PHHs) from three individual donors and primary macaque hepatocytes (PMHs) from freshly isolated livers of six rhesus macaques. In addition, expression of genes encoding Phase I and II metabolic enzymes and the activities of the enzymes were compared in PHHs and PMHs, and Western blot was used to analyze protein biomarkers of DNA damage and apoptosis in PMHs. All eight nitrosamines induced significant DNA damage in both PHHs and PMHs; with the exception of NDMA, higher fold increases in % tail DNA were detected in PMHs compared to PHHs. Greater interindividual variability in CYP gene expression, enzyme activities, and DNA damage responses was observed in PHHs compared to PMHs. Benchmark concentration (BMC) modeling analysis showed that PHHs had more conservative BMC50 values than PMHs for most nitrosamines tested. Nonetheless, correlation analysis demonstrated that DNA damage data generated by PMHs and 3D HepaRG spheroids were comparable to those of PHHs. Western blot analysis suggested a potential role for the ethyl group in regulating protein expression in the DNA damage and apoptosis pathways for nitrosamines. Overall, this study provides human-relevant DNA damage responses for the eight nitrosamines and indicates that differences in genotoxic potency between PHHs and PMHs are likely related to CYP enzyme activity.
Collapse
Affiliation(s)
- Ji-Eun Seo
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA; Office of Scientific Coordination, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA.
| | - Xiaobo He
- Office of Scientific Coordination, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Matthew Bryant
- Office of Scientific Coordination, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Aisar H Atrakchi
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Timothy J McGovern
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Karen L Davis Bruno
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Robert H Heflich
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Xiaoqing Guo
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA.
| |
Collapse
|
3
|
Buloyan S, Harutyunyan A, Gasparyan H, Sakeyan A, Shahkhatuni A, Zakirova NF, Yusubalieva G, Kirillov IM, Fedyakina IT, Solyev PN, Lipatova AV, Bogomolov MA, Prassolov VS, Lebedev TD, Ivanov AV. Piperazine-Substituted Pyranopyridines Exhibit Antiproliferative Activity and Act as Inhibitors of HBV Virion Production. Int J Mol Sci 2025; 26:3991. [PMID: 40362232 PMCID: PMC12071598 DOI: 10.3390/ijms26093991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 05/15/2025] Open
Abstract
Advances in medicinal chemistry have led to the development of anticancer and anti-infectious drugs. However, many types of cancer and viral infections such as hepatitis B virus or SARS-CoV-2 are still treated ineffectively. Therefore, further development of effective and selective lead compounds as potential drugs is still highly demanded. In this study, we synthesized a novel series of piperazine-substituted pyranopyridines and evaluated their anticancer and antiviral properties. Antiproliferative activity was determined in a panel of various tumor cell lines as well as non-tumor hepatic HepaRG cells. Mechanisms of cytotoxicity were assessed by fluorescent microscopy techniques. Antiviral activity was analyzed towards DNA and RNA viruses in infectious cell culture systems. Several compounds showed antiproliferative activity towards various cancer cell lines at micromolar and submicromolar concentrations. Mechanisms of cytotoxicity involve the induction of apoptosis and are not mediated via ERK1/2 pathway or oxidative stress. Several compounds exhibit selective activity against hepatitis B virus by preventing the formation of virion particles. This study led to the identification of a novel class of piperazine-substituted pyranopyridines with antiproliferative activity towards a wide range of tumor cell lines as well as the non-toxic inhibitor of HBV virion production.
Collapse
Affiliation(s)
- Sona Buloyan
- Scientific Technological Center of Organic and Pharmaceutical Chemistry, National Academy of Sciences of the Republic of Armenia, 0019 Yerevan, Armenia; (S.B.)
| | - Arpine Harutyunyan
- Scientific Technological Center of Organic and Pharmaceutical Chemistry, National Academy of Sciences of the Republic of Armenia, 0019 Yerevan, Armenia; (S.B.)
| | - Hrachik Gasparyan
- Scientific Technological Center of Organic and Pharmaceutical Chemistry, National Academy of Sciences of the Republic of Armenia, 0019 Yerevan, Armenia; (S.B.)
| | - Anahit Sakeyan
- Scientific Technological Center of Organic and Pharmaceutical Chemistry, National Academy of Sciences of the Republic of Armenia, 0019 Yerevan, Armenia; (S.B.)
| | - Astghik Shahkhatuni
- Scientific Technological Center of Organic and Pharmaceutical Chemistry, National Academy of Sciences of the Republic of Armenia, 0019 Yerevan, Armenia; (S.B.)
| | - Natalia F. Zakirova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Gaukhar Yusubalieva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, 115682 Moscow, Russia
| | - Ilya M. Kirillov
- Gamaleya National Research Centre for Epidemiology and Microbiology of the Ministry of Russia, 123098 Moscow, Russia
| | - Irina T. Fedyakina
- Gamaleya National Research Centre for Epidemiology and Microbiology of the Ministry of Russia, 123098 Moscow, Russia
| | - Pavel N. Solyev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasia V. Lipatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Mikhail A. Bogomolov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Vladimir S. Prassolov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Timofey D. Lebedev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alexander V. Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
4
|
Graham RE, Zheng R, Wagner J, Unciti-Broceta A, Hay DC, Forbes SJ, Gadd VL, Carragher NO. Single-cell morphological tracking of cell states to identify small-molecule modulators of liver differentiation. iScience 2025; 28:111871. [PMID: 39995868 PMCID: PMC11848441 DOI: 10.1016/j.isci.2025.111871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/24/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
We have developed a single-cell assay that combines Cell Painting-a morphological profiling assay-with trajectory inference analysis. We have applied this morphological trajectory inference to the bi-potent HepaRG liver progenitor cell line allowing us to track liver cell fate and map small-molecule-induced changes using a morphological atlas of liver cell differentiation. Our overarching goal is to demonstrate the potential of Cell Painting to study biological processes as continuous trajectories at the single-cell level, enhancing resolution and biological understanding. This work has identified small-molecule Src family kinase inhibitors that promote the differentiation of HepaRG cells toward a hepatocyte-like lineage as well as primary human hepatic progenitor cells toward a hepatocyte-like phenotype in vitro. These findings could significantly advance research on liver cell regeneration mechanisms and facilitate the development of cell-based and small-molecule therapies.
Collapse
Affiliation(s)
- Rebecca E. Graham
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Runshi Zheng
- Centre for Regenerative Medicine, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jesko Wagner
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Asier Unciti-Broceta
- Edinburgh Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
- Cancer Research UK Scotland Centre, Edinburgh EH4 2XU, UK
| | - David C. Hay
- Centre for Regenerative Medicine, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Stuart J. Forbes
- Centre for Regenerative Medicine, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Victoria L. Gadd
- Centre for Regenerative Medicine, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Neil O. Carragher
- Edinburgh Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
- Cancer Research UK Scotland Centre, Edinburgh EH4 2XU, UK
| |
Collapse
|
5
|
Graf K, Murrieta-Coxca JM, Vogt T, Besser S, Geilen D, Kaden T, Bothe AK, Morales-Prieto DM, Amiri B, Schaller S, Kaufmann L, Raasch M, Ammar RM, Maass C. Digital twin-enhanced three-organ microphysiological system for studying drug pharmacokinetics in pregnant women. Front Pharmacol 2025; 16:1528748. [PMID: 40034823 PMCID: PMC11873563 DOI: 10.3389/fphar.2025.1528748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Background Pregnant women represent a vulnerable group in pharmaceutical research due to limited knowledge about drug metabolism and safety of commonly used corticosteroids like prednisone due to ethical and practical constraints. Current preclinical models, including animal studies, fail to accurately replicate human pregnancy conditions, resulting in gaps in drug safety and pharmacokinetics predictions. To address this issue, we used a three-organ microphysiological system (MPS) combined with a digital twin framework, to predict pharmacokinetics and fetal drug exposure. Methods The here shown human MPS integrated gut, liver, and placenta models, interconnected via the corresponding vasculature. Using prednisone as a model compound, we simulate oral drug administration and track its metabolism and transplacental transfer. To translate the generated data from MPS to human physiology, computational modelling techniques were developed. Results Our results demonstrate that the system maintains cellular integrity and accurately mimics in vivo drug dynamics, with predictions closely matching clinical data from pregnant women. Digital twinning closely aligned with the generated experimental data. Long-term exposure simulations confirmed the value of this integrated system for predicting the non-toxic metabolization of prednisone. Conclusion This approach may provide a potential non-animal alternative that could contribute to our understanding of drug behavior during pregnancy and may support early-stage drug safety assessment for vulnerable populations.
Collapse
Affiliation(s)
| | | | | | | | | | - Tim Kaden
- Dynamic42 GmbH, Jena, Germany
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | | | | | - Behnam Amiri
- MPSlabs, ESQlabs GmbH, Saterland, Germany
- ESQlabs GmbH, Saterland, Germany
| | | | - Ligaya Kaufmann
- Global Medical Affairs, Bayer Consumer Care AG, Basel, Switzerland
| | | | - Ramy M. Ammar
- Global R&D, Bayer Consumer Health, Steigerwald Arzneimittelwerk GmbH, Darmstadt, Germany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafr-El Sheikh, Egypt
| | - Christian Maass
- MPSlabs, ESQlabs GmbH, Saterland, Germany
- ESQlabs GmbH, Saterland, Germany
| |
Collapse
|
6
|
Shanka NY, Pavlov CS, Mekonnen NL. Non-invasive methods for diagnosing portal hypertension and variceal bleeding due to liver cirrhosis secondary to NAFLD/MASLD: systematic review. Front Med (Lausanne) 2025; 11:1459569. [PMID: 39911662 PMCID: PMC11794003 DOI: 10.3389/fmed.2024.1459569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/15/2024] [Indexed: 02/07/2025] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD), recently re-termed as metabolic dysfunction-associated steatotic liver disease (MASLD), is a global health concern affecting approximately 25% of adults. Complications such as portal hypertension and variceal bleeding are critical to diagnose but challenging with traditional invasive methods like hepatic venous pressure gradient (HVPG) measurement and esophagogastroduodenoscopy (EGD), which are not always feasible and carry risks. Objectives This systematic review aim to evaluate the diagnostic accuracy of non-invasive methods for diagnosing portal hypertension and variceal bleeding in patients with NAFLD/MASLD cirrhosis, comparing these methods to invasive standards. Methods A comprehensive literature search was conducted across PubMed, Cochrane Library, Google Scholar, and ScienceDirect from January 2000 to May 2024. Studies included evaluated non-invasive diagnostic techniques for portal hypertension and variceal bleeding, compared with HVPG and EGD, focusing on adult patients with confirmed NAFLD/MASLD cirrhosis. Data extraction covered study characteristics and diagnostic accuracy metrics. The quality of studies was assessed using the QUADAS-2 tool. Meta-analyses were performed using R and Python. Results Eleven studies involving 2,707 patients met the inclusion criteria. Liver stiffness measurement (LSM) via transient elastography demonstrated high sensitivity (85%) and specificity (79%) for diagnosing clinically significant portal hypertension (CSPH) at a 20 kPa cutoff. For severe portal hypertension (SPH), LSM had a sensitivity of 81% and specificity of 85% at 25 kPa. Combining LSM with platelet count resulted in a sensitivity of 97% but lower specificity (41%) for CSPH. Spleen stiffness measurement (SSM) also showed good diagnostic performance with a sensitivity of 89% and specificity of 75% for CSPH. Conclusion Non-invasive tests, particularly LSM and SSM, show promise in diagnosing portal hypertension and variceal bleeding in NAFLD/MASLD cirrhosis. These methods offer high sensitivity, especially in combination, supporting their use in clinical settings to potentially reduce the need for invasive procedures. Future research should aim to standardize protocols and explore additional biomarkers to further enhance diagnostic accuracy. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?, identifier CRD42024567024.
Collapse
Affiliation(s)
- Nebyu Yonas Shanka
- Department of Postgraduate and Doctoral Studies, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Comprehensive Specialized Hospital, Wolaita Sodo University, Soddo, Ethiopia
| | - Chavdar S. Pavlov
- Department of Gastroenterology, Botkin Hospital, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Nigatu Leul Mekonnen
- Department of Public Health, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
7
|
Shanka NY, Pavlov CS, Mekonnen NL. Non-invasive methods for diagnosing portal hypertension and variceal bleeding due to liver cirrhosis secondary to NAFLD/MASLD: systematic review. Front Med (Lausanne) 2025; 11. [DOI: pmid: 39911662 pmcid: pmc11794003 doi: 10.3389/fmed.2024.1459569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
Abstract
BackgroundNon-alcoholic fatty liver disease (NAFLD), recently re-termed as metabolic dysfunction-associated steatotic liver disease (MASLD), is a global health concern affecting approximately 25% of adults. Complications such as portal hypertension and variceal bleeding are critical to diagnose but challenging with traditional invasive methods like hepatic venous pressure gradient (HVPG) measurement and esophagogastroduodenoscopy (EGD), which are not always feasible and carry risks.ObjectivesThis systematic review aim to evaluate the diagnostic accuracy of non-invasive methods for diagnosing portal hypertension and variceal bleeding in patients with NAFLD/MASLD cirrhosis, comparing these methods to invasive standards.MethodsA comprehensive literature search was conducted across PubMed, Cochrane Library, Google Scholar, and ScienceDirect from January 2000 to May 2024. Studies included evaluated non-invasive diagnostic techniques for portal hypertension and variceal bleeding, compared with HVPG and EGD, focusing on adult patients with confirmed NAFLD/MASLD cirrhosis. Data extraction covered study characteristics and diagnostic accuracy metrics. The quality of studies was assessed using the QUADAS-2 tool. Meta-analyses were performed using R and Python.ResultsEleven studies involving 2,707 patients met the inclusion criteria. Liver stiffness measurement (LSM) via transient elastography demonstrated high sensitivity (85%) and specificity (79%) for diagnosing clinically significant portal hypertension (CSPH) at a 20 kPa cutoff. For severe portal hypertension (SPH), LSM had a sensitivity of 81% and specificity of 85% at 25 kPa. Combining LSM with platelet count resulted in a sensitivity of 97% but lower specificity (41%) for CSPH. Spleen stiffness measurement (SSM) also showed good diagnostic performance with a sensitivity of 89% and specificity of 75% for CSPH.ConclusionNon-invasive tests, particularly LSM and SSM, show promise in diagnosing portal hypertension and variceal bleeding in NAFLD/MASLD cirrhosis. These methods offer high sensitivity, especially in combination, supporting their use in clinical settings to potentially reduce the need for invasive procedures. Future research should aim to standardize protocols and explore additional biomarkers to further enhance diagnostic accuracy.Systematic review registrationhttps://www.crd.york.ac.uk/prospero/display_record.php?, identifier CRD42024567024.
Collapse
|
8
|
Delaporte F, Roger E, Bejaud J, Loyer P, Lagarce F, Savary CC. Internalization and mechanisms of toxicity of lipid nanocapsules in HepG2 and HepaRG hepatoma cells upon acute and chronic exposures. Int J Pharm 2024; 667:124815. [PMID: 39424085 DOI: 10.1016/j.ijpharm.2024.124815] [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: 04/03/2024] [Revised: 10/06/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
Lipid nanocapsules (LNCs) used as nanomedicine have been developed to enhance pharmacokinetics and decrease side effects of drugs, particularly for cancer therapies. After intravenous administration, LNCs possess an important hepatic tropism however, few data exist about their toxicity and even less after repeated exposure. This study aimed to assess the in vitro toxicity and internalization of unloaded LNCs, of 50 and 100 nm size, on HepG2 and HepaRG liver cell lines. Internalization of the 50 nm LNCs was slower compared to the 100 nm LNCs and both LNCs exhibited a higher toxicity on cancerous HepG2 cells compared to differentiated HepaRG cells. LNCs were mainly internalized via caveolin-mediated endocytosis in both cell lines. Upon chronic exposure, the toxicity of LNCs on HepaRG cells increased, although the pathways of internalization remained unchanged. Cell death studies have demonstrated an involvement of ferroptosis, but not of apoptosis. After acute and repeated exposures on HepaRG cells, the 100 nm LNCs showed a good safety profile. Finally, LNCs induced a more significant toxicity associated with faster internalization in the HepG2 cancerous model than in the differentiated HepaRG cells. This provides good evidence for LNCs to potentialize the cytotoxic effects of an active drug on liver cancer cells.
Collapse
Affiliation(s)
- Flavien Delaporte
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France; CHU Angers, 4 rue Larrey, 49033 Angers, France.
| | - Emilie Roger
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Jérome Bejaud
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Pascal Loyer
- Inserm, University of Rennes, INRAE, NuMeCan Institute (Nutrition, Metabolisms and Cancer), Rennes, France
| | - Frédéric Lagarce
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France; CHU Angers, 4 rue Larrey, 49033 Angers, France
| | - Camille C Savary
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| |
Collapse
|
9
|
Zhong S, Zheng L, Wu Y, Sun S, Luo Q, Song G, Lü D, Long M. Rotating culture regulates the formation of HepaRG-derived liver organoids via YAP translocation. BMC Biol 2024; 22:262. [PMID: 39548509 PMCID: PMC11568593 DOI: 10.1186/s12915-024-02062-1] [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: 06/06/2024] [Accepted: 11/05/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Liver organoid serves as an alternative model for liver pathophysiology in carbohydrate or lipid metabolism and xenobiotic metabolism transformation. Biomechanical cues including spaceflight mission can affect liver organoid construction and their related functions, but their underlying mechanisms are not fully understood yet. Here, a rotating cell culture device, namely Rotating Flat Chamber (RFC), was specifically designed for adhering cells or cell aggregated to elucidate the effects of altered gravity vector on HepaRG-derived liver organoids construction. RESULTS The organoids so formed under RFC presented the fast growth rate and large projection area. Meanwhile, the expressions of two pluripotency markers of SOX9 and CD44 were enhanced. This finding was positively correlated with the increased YAP expression and nuclear translocation as well as the elevated α4β6-integrin expression. Inhibition of YAP expression and nuclear translocation decreased the expression of SOX9 and CD44 under RFC, thereby attenuating the pluripotency of HepaRG-derived liver organoids. CONCLUSIONS In conclusion, we proposed a novel liver organoid construction method using rotating culture, by which the pluripotency of liver organoids so constructed is mediated by α4β6-integrin and YAP translocation. This work furthered the understanding in how the gravity vector orientation affects the construction of liver organoids and the related mechanotransductive pathways.
Collapse
Affiliation(s)
- Shaoyu Zhong
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Lu Zheng
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi Wu
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shujin Sun
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Dongyuan Lü
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Mian Long
- Center for Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology and Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
10
|
Asano R, Iizaka Y, Kashima M, Anzai Y, Yamaguchi S, Tada M. Unveiling dynamic hepatocyte plasticity in HepaRG cells with a dual CYP reporter system. PLoS One 2024; 19:e0308694. [PMID: 39527612 PMCID: PMC11554142 DOI: 10.1371/journal.pone.0308694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/16/2024] [Indexed: 11/16/2024] Open
Abstract
Primary hepatocytes are widely utilized for investigating drug efficacy and toxicity, yet variations between batches and limited proliferation capacity present significant challenges. HepaRG cells are versatile cells, capable of maintaining an undifferentiated state and differentiating through dimethyl sulfoxide treatment, allowing for molecular analysis of hepatocyte plasticity. To elucidate the underlying molecular mechanisms of HepaRG cell plasticity, we used CYP3A4G/7R HepaRG cells engineered to express DsRed under the control of the fetus-specific CYP3A7 gene and EGFP under the adult-specific CYP3A4 gene promoter. In time-lapse imaging of CYP3A4G/7R HepaRG cells, we observed CYP3A7-DsRed expression transitioning from negative to positive during proliferation period and CYP3A4-GFP expression activating during differentiation. The de-differentiation potency of differentiated CYP3A4G/7R HepaRG cells was assessed using inhibitors and cytokines. It was found that Y-27632 (Y), A-83-01 (A), and CHIR99021 (C) (collectively referred to as YAC), which are known to promote liver regeneration in mice, did not induce CYP3A7-DsRed expression. Instead, these inhibitors increased CYP3A4-GFP expressing population. Furthermore, CHIR99021 alone increased CYP3A4-GFP-positive cells, while Wnt3a treatment increased CYP3A7-DsRed-positive cells, suggesting that Wnt signaling plays distinct roles in HepaRG cells. It was apparent that de-differentiated cells had increased CYP3A4 activity after a second round of differentiation, compared to differentiated cells after the first round. Transcriptomic analysis of HepaRG cells revealed distinct profiles between proliferative, differentiated, and de-differentiated states, highlighting their robust plasticity. Notably, hepatoblastic cells de-differentiated by YAC or C displayed transcriptome patterns similar to undifferentiated cells, whereas CYP3A7-DsRed and CYP3A4-GFP exhibited expression patterns different from those of undifferentiated cells. These findings underscore the dynamic nature of HepaRG cells while cautioning against solely relying on CYP3 family gene expression as a marker of differentiation.
Collapse
Affiliation(s)
- Riku Asano
- Stem Cells & Reprogramming Laboratory, Department of Biology, Faculty of Science, Toho University, Chiba, Japan
| | - Yohei Iizaka
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Toho University, Chiba, Japan
| | - Makoto Kashima
- Department of Biomolecular Science, Faculty of Science, Toho University, Chiba, Japan
| | - Yojiro Anzai
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Toho University, Chiba, Japan
| | - Shinpei Yamaguchi
- Stem Cells & Reprogramming Laboratory, Department of Biology, Faculty of Science, Toho University, Chiba, Japan
| | - Masako Tada
- Stem Cells & Reprogramming Laboratory, Department of Biology, Faculty of Science, Toho University, Chiba, Japan
| |
Collapse
|
11
|
Budi NYP, Lai WY, Huang YH, Ho HN. 3D organoid cultivation improves the maturation and functional differentiation of cholangiocytes from human pluripotent stem cells. Front Cell Dev Biol 2024; 12:1361084. [PMID: 39040044 PMCID: PMC11260683 DOI: 10.3389/fcell.2024.1361084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 05/29/2024] [Indexed: 07/24/2024] Open
Abstract
Idiopathic cholangiopathies are diseases that affect cholangiocytes, and they have unknown etiologies. Currently, orthotopic liver transplantation is the only treatment available for end-stage liver disease. Limited access to the bile duct makes it difficult to model cholangiocyte diseases. In this study, by mimicking the embryonic development of cholangiocytes and using a robust, feeder- and serum-free protocol, we first demonstrate the generation of unique functional 3D organoids consisting of small and large cholangiocytes derived from human pluripotent stem cells (PSCs), as opposed to traditional 2D culture systems. At day 28 of differentiation, the human PSC-derived cholangiocytes expressed markers of mature cholangiocytes, such as CK7, CK19, and cystic fibrosis transmembrane conductance regulator (CFTR). Compared with the 2D culture system-generated cholangiocytes, the 3D cholangiocyte organoids (COs) showed higher expression of the region-specific markers of intrahepatic cholangiocytes YAP1 and JAG1 and extrahepatic cholangiocytes AQP1 and MUC1. Furthermore, the COs had small-large tube-like structures and functional assays revealed that they exhibited characteristics of mature cholangiocytes, such as multidrug resistance protein 1 transporter function and CFTR channel activity. In addition to the extracellular matrix supports, the epidermal growth factor receptor (EGFR)-mediated signaling regulation might be involved in this cholangiocyte maturation and differentiation. These results indicated the successful generation of intrahepatic and extrahepatic cholangiocytes by using our 3D organoid protocol. The results highlight the advantages of our 3D culture system over the 2D culture system in promoting the functional differentiation and maturation of cholangiocytes. In summary, in advance of the previous works, our study provides a possible concept of small-large cholangiocyte transdifferentiation of human PSCs under cost-effective 3D culture conditions. The study findings have implications for the development of effective cell-based therapy using COs for patients with cholangiopathies.
Collapse
Affiliation(s)
- Nova Yuli Prasetyo Budi
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Pediatric Surgery Division, Department of Surgery/Genetics Working Group, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada/Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Wei-Yu Lai
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yen-Hua Huang
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hong-Nerng Ho
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Obstetrics and Gynecology, Taipei Municipal Wanfang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
12
|
Vasisth R, Gurao A, Chitkara M, Kumar G, Sriranga KR, Mukesh M, Dige MS, Singh P, Aggarwal RAK, Kataria RS. Selection of reference genes for normalizing gene expression data across seasons in spermatozoa of water buffalo (Bubalus bubalis). INTERNATIONAL JOURNAL OF BIOMETEOROLOGY 2024; 68:1397-1409. [PMID: 38602552 DOI: 10.1007/s00484-024-02675-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/04/2023] [Accepted: 03/01/2024] [Indexed: 04/12/2024]
Abstract
Selection of the most stably expressed reference genes is key to monitoring accurate target gene expression across any tissue or cell type. The mRNA in spermatozoa stores valuable information related to changes in spermatogenesis due to variations in environmental conditions, especially during heat stress, which affects various sperm functions. Semen quality in buffalo bulls is significantly influenced by the seasons. In the study, a panel of nine genes was evaluated to identify the most stably expressed internal control gene (ICG) for the normalization of real-time gene expression data generated across various seasons for Murrah buffalo bulls' spermatozoa. Sperm cells were purified from the semen samples collected during different seasons, with temperature-humidity index (THI) ranging from 80.80 ± 1.47 (hot summer) to 55.88 ± 1.98 (winter), using the BoviPure™ gradient purification method. The RNA isolated from the purified spermatozoa fraction was quality checked prior to reverse transcription and subjected to qPCR (quantitative real-time PCR) based expression analysis. An automated 'endoGene' pipeline was employed to apply the geNorm, NormFinder, and BestKeeper algorithms for data analysis. The result indicated that GAPDH and PP1A were the most stably expressed among the gene panel, whereas ATPSF1 and ACTB were the two least stable expressed reference genes. Further, the most suitable ICGs identified were validated by normalization of real time expression data of heat stress and sperm quality genes, HSFY2 and AKAP4, respectively. The genes identified would help in generating the most reliable results for the expression profiling of the genes dictating sperm quality and heat stress cope-up mechanism in buffalo spermatozoa, collected during different seasons.
Collapse
Affiliation(s)
- Rashi Vasisth
- ICAR- National Bureau of Animal Genetic Resources, Karnal, Haryana, 132001, India
- ICAR- National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Ankita Gurao
- ICAR- National Bureau of Animal Genetic Resources, Karnal, Haryana, 132001, India
| | - Meenakshi Chitkara
- ICAR- National Bureau of Animal Genetic Resources, Karnal, Haryana, 132001, India
- ICAR- National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Gautam Kumar
- ICAR- National Bureau of Animal Genetic Resources, Karnal, Haryana, 132001, India
- ICAR- National Dairy Research Institute, Karnal, Haryana, 132001, India
| | | | - Manishi Mukesh
- ICAR- National Bureau of Animal Genetic Resources, Karnal, Haryana, 132001, India
| | | | - Pawan Singh
- ICAR- National Dairy Research Institute, Karnal, Haryana, 132001, India
| | | | - Ranjit Singh Kataria
- ICAR- National Bureau of Animal Genetic Resources, Karnal, Haryana, 132001, India.
| |
Collapse
|
13
|
Yang L, Gao YL, Jiang S, Qian B, Che L, Wu JS, Du ZB, Wang MZ, Yang Y, Lin YC, Liu G, Lin ZN. Aflatoxin B 1-exposed hepatocyte-derived extracellular vesicles: Initiating hepatic stellate cell-mediated liver fibrosis through a p53-Parkin-dependent mitophagy pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 277:116363. [PMID: 38663190 DOI: 10.1016/j.ecoenv.2024.116363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 02/27/2024] [Accepted: 04/19/2024] [Indexed: 05/30/2024]
Abstract
Environmental aflatoxin B1 (AFB1) exposure has been proposed to contribute to hepatocellular carcinoma by promoting liver fibrosis, but the potential mechanisms remain to be further elucidated. Extracellular vesicles (EVs) were recognized as crucial traffickers for hepatic intercellular communication and play a vital role in the pathological process of liver fibrosis. The AFB1-exposed hepatocyte-derived EVs (AFB1-EVs) were extracted, and the functional effects of AFB1-EVs on the activation of hepatic stellate cells (HSCs) were explored to investigate the molecular mechanism of AFB1 exposure-induced liver fibrogenesis. Our results revealed that an environment-level AFB1 exposure induced liver fibrosis via HSCs activation in mice, while the AFB1-EVs mediated hepatotoxicity and liver fibrogenesis in vitro and in vivo. AFB1 exposure in vitro increased PINK1/Parkin-dependent mitophagy in hepatocytes, where upregulated transcription of the PARK2 gene via p53 nuclear translocation and mitochondrial recruitment of Parkin, and promoted AFB1-EVs-mediated mitochondria-trafficking communication between hepatocytes and HSCs. The knockdown of Parkin in HepaRG cells reversed HSCs activation by blocking the mitophagy-related AFB1-EVs trafficking. This study further revealed that the hepatic fibrogenesis of AFB1 exposure was rescued by genetic intervention with siPARK2 or p53's Pifithrin-α (PFTα) inhibitors. Furthermore, AFB1-EVs-induced HSCs activation was relieved by GW4869 pharmaceutic inhibition of EVs secretion. These results revealed a novel mechanism that AFB1 exposure-induced p53-Parkin signal axis regulated mitophagy-dependent hepatocyte-derived EVs to mediate the mitochondria-trafficking intercellular communication between hepatocytes and HSCs in the local hepatotoxic microenvironment to promote the activated HSCs-associated liver fibrogenesis. Our study provided insight into p53-Parkin-dependent pathway regulation and promised an advanced strategy targeting intervention to EVs-mediated mitochondria trafficking for preventing xenobiotics-induced liver fibrosis.
Collapse
Affiliation(s)
- Lei Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yun-Lu Gao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Shan Jiang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Bo Qian
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Lin Che
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Jia-Shen Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Ze-Bang Du
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Ming-Zhu Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yun Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yu-Chun Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Zhong-Ning Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
14
|
Seo JE, Le Y, Revollo J, Miranda-Colon J, Xu H, McKinzie P, Mei N, Chen T, Heflich RH, Zhou T, Robison T, Bonzo JA, Guo X. Evaluating the mutagenicity of N-nitrosodimethylamine in 2D and 3D HepaRG cell cultures using error-corrected next generation sequencing. Arch Toxicol 2024; 98:1919-1935. [PMID: 38584193 PMCID: PMC11106104 DOI: 10.1007/s00204-024-03731-4] [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: 11/12/2023] [Accepted: 03/07/2024] [Indexed: 04/09/2024]
Abstract
Human liver-derived metabolically competent HepaRG cells have been successfully employed in both two-dimensional (2D) and 3D spheroid formats for performing the comet assay and micronucleus (MN) assay. In the present study, we have investigated expanding the genotoxicity endpoints evaluated in HepaRG cells by detecting mutagenesis using two error-corrected next generation sequencing (ecNGS) technologies, Duplex Sequencing (DS) and High-Fidelity (HiFi) Sequencing. Both HepaRG 2D cells and 3D spheroids were exposed for 72 h to N-nitrosodimethylamine (NDMA), followed by an additional incubation for the fixation of induced mutations. NDMA-induced DNA damage, chromosomal damage, and mutagenesis were determined using the comet assay, MN assay, and ecNGS, respectively. The 72-h treatment with NDMA resulted in concentration-dependent increases in cytotoxicity, DNA damage, MN formation, and mutation frequency in both 2D and 3D cultures, with greater responses observed in the 3D spheroids compared to 2D cells. The mutational spectrum analysis showed that NDMA induced predominantly A:T → G:C transitions, along with a lower frequency of G:C → A:T transitions, and exhibited a different trinucleotide signature relative to the negative control. These results demonstrate that the HepaRG 2D cells and 3D spheroid models can be used for mutagenesis assessment using both DS and HiFi Sequencing, with the caveat that severe cytotoxic concentrations should be avoided when conducting DS. With further validation, the HepaRG 2D/3D system may become a powerful human-based metabolically competent platform for genotoxicity testing.
Collapse
Affiliation(s)
- Ji-Eun Seo
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Yuan Le
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Javier Revollo
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Jaime Miranda-Colon
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Hannah Xu
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Page McKinzie
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Nan Mei
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Tao Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Robert H Heflich
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Tong Zhou
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Rockville, MD, 20855, USA
| | - Timothy Robison
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Jessica A Bonzo
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Xiaoqing Guo
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA.
| |
Collapse
|
15
|
Preiss LC, Georgi K, Lauschke VM, Petersson C. Comparison of Human Long-Term Liver Models for Clearance Prediction of Slowly Metabolized Compounds. Drug Metab Dispos 2024; 52:539-547. [PMID: 38604730 DOI: 10.1124/dmd.123.001638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/06/2024] [Accepted: 03/19/2024] [Indexed: 04/13/2024] Open
Abstract
The accurate prediction of human clearance is an important task during drug development. The proportion of low clearance compounds has increased in drug development pipelines across the industry since such compounds may be dosed in lower amounts and at lower frequency. These type of compounds present new challenges to in vitro systems used for clearance extrapolation. In this study, we compared the accuracy of clearance predictions of suspension culture to four different long-term stable in vitro liver models, including HepaRG sandwich culture, the Hµrel stochastic co-culture, the Hepatopac micropatterned co-culture (MPCC), and a micro-array spheroid culture. Hepatocytes in long-term stable systems remained viable and active over several days of incubation. Although intrinsic clearance values were generally high in suspension culture, clearance of low turnover compounds could frequently not be determined using this method. Metabolic activity and intrinsic clearance values from HepaRG cultures were low and, consequently, many compounds with low turnover did not show significant decline despite long incubation times. Similarly, stochastic co-cultures occasionally failed to show significant turnover for multiple low and medium turnover compounds. Among the different methods, MPCCs and spheroids provided the most consistent measurements. Notably, all culture methods resulted in underprediction of clearance; this could, however, be compensated for by regression correction. Combined, the results indicate that spheroid culture as well as the MPCC system provide adequate in vitro tools for human extrapolation for compounds with low metabolic turnover. SIGNIFICANCE STATEMENT: In this study, we compared suspension cultures, HepaRG sandwich cultures, the Hµrel liver stochastic co-cultures, the Hepatopac micropatterned co-cultures (MPCC), and micro-array spheroid cultures for low clearance determination and prediction. Overall, HepaRG and suspension cultures showed modest value for the low determination and prediction of clearance compounds. The micro-array spheroid culture resulted in the most robust clearance measurements, whereas using the MPCC resulted in the most accurate prediction for low clearance compounds.
Collapse
Affiliation(s)
- Lena C Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Darmstadt, Germany (L.C.P., K.G., C.P.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.); and University of Tuebingen, Tuebingen, Germany (V.M.L.)
| | - Katrin Georgi
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Darmstadt, Germany (L.C.P., K.G., C.P.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.); and University of Tuebingen, Tuebingen, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Darmstadt, Germany (L.C.P., K.G., C.P.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.); and University of Tuebingen, Tuebingen, Germany (V.M.L.)
| | - Carl Petersson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (L.C.P., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Darmstadt, Germany (L.C.P., K.G., C.P.); Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.); and University of Tuebingen, Tuebingen, Germany (V.M.L.)
| |
Collapse
|
16
|
Guo X, Xu H, Seo JE. Application of HepaRG cells for genotoxicity assessment: a review. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2024; 42:214-237. [PMID: 38566478 DOI: 10.1080/26896583.2024.2331956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
There has been growing interest in the use of human-derived metabolically competent cells for genotoxicity testing. The HepaRG cell line is considered one of the most promising cell models because it is TP53-proficient and retains many characteristics of primary human hepatocytes. In recent years, HepaRG cells, cultured in both a traditional two-dimensional (2D) format and as more advanced in-vivo-like 3D spheroids, have been employed in assays that measure different types of genetic toxicity endpoints, including DNA damage, mutations, and chromosomal damage. This review summarizes published studies that have used HepaRG cells for genotoxicity assessment, including cell model evaluation studies and risk assessment for various compounds. Both 2D and 3D HepaRG models can be adapted to several high-throughput genotoxicity assays, generating a large number of data points that facilitate quantitative benchmark concentration modeling. With further validation, HepaRG cells could serve as a unique, human-based new alternative methodology for in vitro genotoxicity testing.
Collapse
Affiliation(s)
- Xiaoqing Guo
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Jefferson, AR, USA
| | - Hannah Xu
- 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
| |
Collapse
|
17
|
Wei J, Zhang B, Tang J, Cao J, Du C, Wang Z, Zhang Y, Xie M, Zhou Z, Hou S. Embryonic growth and effect of embryonic age on quantitative and functional characteristics of duck primary hepatocytes. Poult Sci 2024; 103:103531. [PMID: 38417329 PMCID: PMC10909911 DOI: 10.1016/j.psj.2024.103531] [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: 11/30/2023] [Revised: 01/22/2024] [Accepted: 02/01/2024] [Indexed: 03/01/2024] Open
Abstract
Primary hepatocytes (PH) have been widely used in metabolic and disease-resistance mechanism research. However, hepatocyte isolation (HI) remains challenging in ducks. This study aimed to explore embryonic growth and the effect of embryonic age (EA) on the quantitative and functional characteristics of PH in ducks. For embryonic growth, the size and weight of the embryo and liver were determined from 6 to 28 EA (E6-E28, similar below). As EA increased, the corresponding size and weight grew significantly. Specifically, embryonic length varied from 12.5 mm to 133.0 mm, and liver width varied from 2.0 mm to 26.2 mm. Embryonic weight ranged from 0.259 g to 53.58 g, and liver weight ranged from 0.007 g to 1.765 g. Liver index initially decreased and then increased with a ratio ranging from 1.06 to 3.29%. For quantitative and functional characteristics, they were determined from E6 to E22, as there were no obvious liver features before E6 and few cells obtained after E22. The number of cells isolated in liver increased from E6 to E16 and then sharply decreased from E16 to E22. The viability remained relatively stable from E6 to E10 and then decreased from E12 to E22. The comprehensive intensity of hepatic glycogen was stronger at E8 and E14. Albumin expression increased markedly from E6 to E18 by qPCR, and the overall albumin expression was stronger at E8 and E14 by immunofluorescence assay. Hepatocyte purity exceeded 90% except for E20 and E22. During culture, cell clusters appeared after 24-h culture, which were identified as nonhepatocytes. The growth curve showed an initial increase in cell quantity followed by a decrease, another increase, and then remaining stable. In conclusion, EA had a significant effect on the quantitative and functional characteristics of PH, and the suitable EA for HI were E8 and E14. Considering better operability and quantity, E14 was the optimal EA, laying a solid foundation for further hepatocyte purification, nutrient metabolism, and disease-resistance mechanism explorations in ducks.
Collapse
Affiliation(s)
- Jie Wei
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Bo Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| | - Jing Tang
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Junting Cao
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Chenchen Du
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Zhen Wang
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yunsheng Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ming Xie
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Zhengkui Zhou
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Shuisheng Hou
- State Key Laboratory of Animal Nutrition and Feeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
18
|
Recoules C, Mirey G, Audebert M. Effect of cell treatment procedures on in vitro genotoxicity assessment. Arch Toxicol 2024; 98:1225-1236. [PMID: 38427119 DOI: 10.1007/s00204-024-03690-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/23/2024] [Indexed: 03/02/2024]
Abstract
So far, the majority of in vitro toxicological experiments are conducted after an acute 24 h treatment that does not represent a realistic human chemical exposure. Recently, new in vitro approaches have been proposed to study the chemical toxicological effect over several days in order to be more predictive of a representative exposure scenario. In this study, we investigated the genotoxic potential of chemicals (direct or bioactived clastogen, aneugen and apoptotic inducer) with the γH2AX and pH3 biomarkers, in the human liver-derived HepaRP cell line. We used different treatment durations, with or without a three-day recovery stage (release period), before genotoxicity measurement. Data were analysed with the Benchmark Dose approach. We observed that the detection of clastogenic compounds (notably for DNA damaging agents) was more sensitive after three days of repeated treatment compared to one or three treatments over 24 h. In contrast, aneugenic chemicals were detected as genotoxic in a similar manner whether after a 24 h exposure or a three-day repeated treatment. Globally, the release period decreases the genotoxicity measurement substantially. For DNA damaging agents, after high concentration treatments, γH2AX induction was always observed after a three-day release period. In contrast, for DNA topoisomerase inhibitors, no effect could be observed after the release period. In conclusion, in the HepaRP cell line, there are some important differences between a one-day acute and a three-day repeated treatment protocol, indicating that different cell treatment procedures may differentiate chemical genotoxic mechanisms of action more efficiently.
Collapse
Affiliation(s)
- Cynthia Recoules
- Toxalim, INRAE-UMR1331, INP-ENVT, INP-EI-Purpan, Université de Toulouse 3 Paul Sabatier, 180 Chemin de Tournefeuille, BP 93173, 31027, Toulouse Cedex 3, France
| | - Gladys Mirey
- Toxalim, INRAE-UMR1331, INP-ENVT, INP-EI-Purpan, Université de Toulouse 3 Paul Sabatier, 180 Chemin de Tournefeuille, BP 93173, 31027, Toulouse Cedex 3, France
| | - Marc Audebert
- Toxalim, INRAE-UMR1331, INP-ENVT, INP-EI-Purpan, Université de Toulouse 3 Paul Sabatier, 180 Chemin de Tournefeuille, BP 93173, 31027, Toulouse Cedex 3, France.
| |
Collapse
|
19
|
Senthilkumar S, Solan ME, Fernandez-Luna MT, Lavado R. Cannabidiol and Indole-3-carbinol Reduce Intracellular Lipid Droplet
Accumulation in HepaRG, A Human Liver Cell Line, as well as in Human
Adipocytes. THE NATURAL PRODUCTS JOURNAL 2024; 14. [DOI: 10.2174/2210315513666230526100544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/22/2023] [Accepted: 03/02/2023] [Indexed: 01/03/2025]
Abstract
Introduction:
An increase in obesity-related diseases is becoming an alarming worldwide problem. Therefore, new therapeutic methods are constantly sought to prevent, treat, and alleviate symptoms of the diseases associated with obesity.
Method:
This study investigates the effects of two natural compounds (indole-3-carbinol, I3C, a bioactive indolic compound found in cruciferous vegetables; cannabidiol, CBD, the active ingredient derived from the hemp plant) on the fatty acid accumulation in the human liver cell line HepaRG, a well-established model for non-alcoholic fatty liver disease (NAFLD) and in human pre-adipocytes (adipose-derived mesenchymal stem cells, MSC).
Results:
EC50s of each compound were in the high µM range (approximately 30 mg/L), showing the low toxicity of these compounds. Determination of the selected compounds in cell media showed no significant differences during the exposure, suggesting that no significant metabolism or degradation happened during the exposure time. Quantification of the bioaccumulation of lipid droplets on exposed HepaRG revealed a significant reduction and mitigation of fatty acid accumulation when exposed to 1 nM of I3C and 100 nM of CBD.). On MSC cells a significant inhibition of lipogenesis and adipocyte differentiation was observed in cells exposed to 0.1 nM of I3C and 1 nM of CBD.
Conclusion:
This study provides a significant contribution to advancing the understanding of preventative dietary strategies that target adipocyte differentiation and NAFLD.
Collapse
Affiliation(s)
| | - Megan E. Solan
- Department of Environmental Science, Baylor University, Waco, TX 76798, USA
| | | | - Ramon Lavado
- Department of Environmental Science, Baylor University, Waco, TX 76798, USA
| |
Collapse
|
20
|
Hanaoka K, Ikeno T, Iwaki S, Deguchi S, Takayama K, Mizuguchi H, Tao F, Kojima N, Ohno H, Sasaki E, Komatsu T, Ueno T, Maeda K, Kusuhara H, Urano Y. A general fluorescence off/on strategy for fluorogenic probes: Steric repulsion-induced twisted intramolecular charge transfer (sr-TICT). SCIENCE ADVANCES 2024; 10:eadi8847. [PMID: 38363840 PMCID: PMC10871538 DOI: 10.1126/sciadv.adi8847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 01/17/2024] [Indexed: 02/18/2024]
Abstract
Various control strategies are available for building fluorogenic probes to visualize biological events in terms of a fluorescence change. Here, we performed the time-dependent density functional theory (TD-DFT) computational analysis of the twisted intramolecular charge transfer (TICT) process in rhodamine dyes. On the basis of the results, we designed and synthesized a series of rhodamine dyes and established a fluorescence quenching strategy that we call steric repulsion-induced TICT (sr-TICT), in which the fluorescence quenching process is greatly accelerated by simple intramolecular twisting. As proof of concept of this design strategy, we used it to develop a fluorogenic probe, 2-Me PeER (pentyloxyethylrhodamine), for the N-dealkylation activity of CYP3A4. We applied 2-Me PeER for CYP3A4 activity-based fluorescence-activated cell sorting (FACS), providing access to homogeneous, highly functional human-induced pluripotent stem cell (hiPSC)-derived hepatocytes and intestinal epithelial cells. Our results suggest that sr-TICT represents a general fluorescence control method for fluorogenic probes.
Collapse
Affiliation(s)
- Kenjiro Hanaoka
- Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minoto-ku, Tokyo 105-8512, Japan
| | - Takayuki Ikeno
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shimpei Iwaki
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Sayaka Deguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kazuo Takayama
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka 565-0871, Japan
- National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Fumiya Tao
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan
| | - Nobuhiko Kojima
- Department of Life and Environmental System Science, Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan
| | - Hisashi Ohno
- Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minoto-ku, Tokyo 105-8512, Japan
| | - Eita Sasaki
- Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minoto-ku, Tokyo 105-8512, Japan
| | - Toru Komatsu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tasuku Ueno
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kazuya Maeda
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiroyuki Kusuhara
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yasuteru Urano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
21
|
Bronsard J, Savary C, Massart J, Viel R, Moutaux L, Catheline D, Rioux V, Clement B, Corlu A, Fromenty B, Ferron PJ. 3D multi-cell-type liver organoids: A new model of non-alcoholic fatty liver disease for drug safety assessments. Toxicol In Vitro 2024; 94:105728. [PMID: 37951556 DOI: 10.1016/j.tiv.2023.105728] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 10/23/2023] [Accepted: 11/03/2023] [Indexed: 11/14/2023]
Abstract
The development of in vitro models that recapitulate critical liver functions is essential for accurate assessments of drug toxicity. Although liver organoids can be used for drug discovery and toxicology, they are limited by (i) the lack of expression and activity of xenobiotic-metabolizing enzymes, and (ii) the difficulty of mimicking non-alcoholic fatty liver disease (NAFLD, which influences the expression of these enzymes) in vitro. Here, we generated three-dimensional multi-cell-type liver organoids (hereafter "HML organoids") from HepaRG cells, primary human macrophages, and hepatic-stellate-cell-derived LX-2 cells. We also developed an NAFLD model by culturing HML organoids for 9 days with a mixture of stearic and oleic acids. The exposed organoids showed typical features of steatosis and expressed fibrosis markers. We subsequently used HML and NAFLD-HML organoids to model drug-induced liver injury. By estimating the IC50 and benchmark doses, we were able to improve the in vitro detection of drugs likely to be toxic in fatty livers. Thus, HML and NAFLD-HML organoids exhibited most of the liver's functions and are relevant in vitro models of drug metabolism, drug toxicity, and adverse drug event in NAFLD.
Collapse
Affiliation(s)
- J Bronsard
- INSERM, Université de Rennes, INRAE, Institut NuMeCan UMR1317 (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France
| | - C Savary
- Univ Angers, CHU Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - J Massart
- INSERM, Université de Rennes, INRAE, Institut NuMeCan UMR1317 (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France
| | - R Viel
- Univ Rennes, CNRS, Inserm, Biosit UAR 3480 US_S 018, France-BioImaging (ANR-10-INBS-04), plateforme H2P2, F-35000 Rennes, France
| | - L Moutaux
- INSERM, Université de Rennes, INRAE, Institut NuMeCan UMR1317 (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France
| | - D Catheline
- INSERM, Université de Rennes, INRAE, Institut NuMeCan UMR1317 (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France
| | - V Rioux
- INSERM, Université de Rennes, INRAE, Institut NuMeCan UMR1317 (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France
| | - B Clement
- INSERM, Université de Rennes, INRAE, Institut NuMeCan UMR1317 (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France
| | - A Corlu
- INSERM, Université de Rennes, INRAE, Institut NuMeCan UMR1317 (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France
| | - B Fromenty
- INSERM, Université de Rennes, INRAE, Institut NuMeCan UMR1317 (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France
| | - P J Ferron
- INSERM, Université de Rennes, INRAE, Institut NuMeCan UMR1317 (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France.
| |
Collapse
|
22
|
Petitjean K, Verres Y, Bristeau S, Ribault C, Aninat C, Olivier C, Leroyer P, Ropert M, Loréal O, Herault O, Amalric L, Baran N, Fromenty B, Corlu A, Loyer P. Low concentrations of ethylene bisdithiocarbamate pesticides maneb and mancozeb impair manganese and zinc homeostasis to induce oxidative stress and caspase-dependent apoptosis in human hepatocytes. CHEMOSPHERE 2024; 346:140535. [PMID: 37923018 DOI: 10.1016/j.chemosphere.2023.140535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 10/02/2023] [Accepted: 10/23/2023] [Indexed: 11/07/2023]
Abstract
The worldwide and intensive use of phytosanitary compounds results in environmental and food contamination by chemical residues. Human exposure to multiple pesticide residues is a major health issue. Considering that the liver is not only the main organ for metabolizing pesticides but also a major target of toxicities induced by xenobiotics, we studied the effects of a mixture of 7 pesticides (chlorpyrifos-ethyl, dimethoate, diazinon, iprodione, imazalil, maneb, mancozeb) often detected in food samples. Effects of the mixture was investigated using metabolically competent HepaRG cells and human hepatocytes in primary culture. We report the strong cytotoxicity of the pesticide mixture towards hepatocytes-like HepaRG cells and human hepatocytes upon acute and chronic exposures at low concentrations extrapolated from the Acceptable Daily Intake (ADI) of each compound. Unexpectedly, we demonstrated that the manganese (Mn)-containing dithiocarbamates (DTCs) maneb and mancozeb were solely responsible for the cytotoxicity induced by the mixture. The mechanism of cell death involved the induction of oxidative stress, which led to cell death by intrinsic apoptosis involving caspases 3 and 9. Importantly, this cytotoxic effect was found only in cells metabolizing these pesticides. Herein, we unveil a novel mechanism of toxicity of the Mn-containing DTCs maneb and mancozeb through their metabolization in hepatocytes generating the main metabolite ethylene thiourea (ETU) and the release of Mn leading to intracellular Mn overload and depletion in zinc (Zn). Alteration of the Mn and Zn homeostasis provokes the oxidative stress and the induction of apoptosis, which can be prevented by Zn supplementation. Our data demonstrate the hepatotoxicity of Mn-containing fungicides at very low doses and unveil their adverse effect in disrupting Mn and Zn homeostasis and triggering oxidative stress in human hepatocytes.
Collapse
Affiliation(s)
- Kilian Petitjean
- Inserm, INRAE, Univ Rennes, Institut NUMECAN (Nutrition Métabolismes et Cancer) UMR-A 1341, UMR-S 1317, F-35000 Rennes, France
| | - Yann Verres
- Inserm, INRAE, Univ Rennes, Institut NUMECAN (Nutrition Métabolismes et Cancer) UMR-A 1341, UMR-S 1317, F-35000 Rennes, France
| | - Sébastien Bristeau
- BRGM, Direction Eau, Environnement, Procédés et Analyses (DEPA), 3 Avenue Claude-Guillemin - BP 36009, 45060 Orléans Cedex 2, France
| | - Catherine Ribault
- Inserm, INRAE, Univ Rennes, Institut NUMECAN (Nutrition Métabolismes et Cancer) UMR-A 1341, UMR-S 1317, F-35000 Rennes, France
| | - Caroline Aninat
- Inserm, INRAE, Univ Rennes, Institut NUMECAN (Nutrition Métabolismes et Cancer) UMR-A 1341, UMR-S 1317, F-35000 Rennes, France
| | - Christophe Olivier
- Cancéropole Grand Ouest (CGO), NET "Niches and Epigenetics of Tumors" Network, 44000 Nantes, France; INSERM UMR 1232 CRCINA, 44000 Nantes-Angers, France; Faculty of Pharmaceutical and Biological Sciences, Nantes University, 44000 Nantes, France
| | - Patricia Leroyer
- Inserm, INRAE, Univ Rennes, Institut NUMECAN (Nutrition Métabolismes et Cancer) UMR-A 1341, UMR-S 1317, F-35000 Rennes, France
| | - Martine Ropert
- Inserm, INRAE, Univ Rennes, Institut NUMECAN (Nutrition Métabolismes et Cancer) UMR-A 1341, UMR-S 1317, F-35000 Rennes, France; AEM2 Platform, CHU Pontchaillou, 2 Rue Henri le Guilloux, 35033 Rennes, France
| | - Olivier Loréal
- Inserm, INRAE, Univ Rennes, Institut NUMECAN (Nutrition Métabolismes et Cancer) UMR-A 1341, UMR-S 1317, F-35000 Rennes, France
| | - Olivier Herault
- Cancéropole Grand Ouest (CGO), NET "Niches and Epigenetics of Tumors" Network, 44000 Nantes, France; Department of Biological Hematology, Tours University Hospital, 37000 Tours, France; CNRS ERL 7001 LNOx, EA 7501, Tours University, 37000 Tours, France; CNRS GDR3697 Micronit "Microenvironment of Tumor Niches", Tours, France
| | - Laurence Amalric
- BRGM, Direction Eau, Environnement, Procédés et Analyses (DEPA), 3 Avenue Claude-Guillemin - BP 36009, 45060 Orléans Cedex 2, France
| | - Nicole Baran
- BRGM, Direction Eau, Environnement, Procédés et Analyses (DEPA), 3 Avenue Claude-Guillemin - BP 36009, 45060 Orléans Cedex 2, France
| | - Bernard Fromenty
- Inserm, INRAE, Univ Rennes, Institut NUMECAN (Nutrition Métabolismes et Cancer) UMR-A 1341, UMR-S 1317, F-35000 Rennes, France
| | - Anne Corlu
- Inserm, INRAE, Univ Rennes, Institut NUMECAN (Nutrition Métabolismes et Cancer) UMR-A 1341, UMR-S 1317, F-35000 Rennes, France; Cancéropole Grand Ouest (CGO), NET "Niches and Epigenetics of Tumors" Network, 44000 Nantes, France.
| | - Pascal Loyer
- Inserm, INRAE, Univ Rennes, Institut NUMECAN (Nutrition Métabolismes et Cancer) UMR-A 1341, UMR-S 1317, F-35000 Rennes, France; Cancéropole Grand Ouest (CGO), NET "Niches and Epigenetics of Tumors" Network, 44000 Nantes, France.
| |
Collapse
|
23
|
Pelletier R, Bourdais A, Fabresse N, Ferron PJ, Morel I, Gicquel T, Le Daré B. In silico and in vitro metabolism studies of the new synthetic opiate AP-237 (bucinnazine) using bioinformatics tools. Arch Toxicol 2024; 98:165-179. [PMID: 37839054 DOI: 10.1007/s00204-023-03617-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/27/2023] [Indexed: 10/17/2023]
Abstract
The recent emergence of new synthetic opioids (NSOs) compounds in the illicit market is increasingly related to fatal cases. Identification and medical care of NSO intoxication cases are challenging, particularly due to high frequency of new products and extensive metabolism. As the study of NSO metabolism is crucial for the identification of these drugs in cases of intoxication, we aimed to investigate the metabolism of the piperazine NSO AP-237 (= bucinnazine). Two complementary approaches (in silico and in vitro) were used to identify putative AP-237 metabolites which could be used as consumption markers. In silico metabolism studies were realized by combining four open access softwares (MetaTrans, SyGMa, Glory X, Biotransformer 3.0). In vitro experiments were performed by incubating AP-237 (20 µM) in differentiated HepaRG cells during 0 h, 8 h, 24 h or 48 h. Cell supernatant were extracted and analyzed by liquid chromatography coupled to high-resolution mass spectrometry and data were reprocessed using three strategies (MetGem, GNPS or Compound Discoverer®). A total of 28 phase I and six phase II metabolites was predicted in silico. Molecular networking identified seven putative phase I metabolites (m/z 203.154, m/z 247.180, m/z 271.180, two m/z 289.191 isomers, m/z 305.186, m/z 329.222), including four previously unknown metabolites. Overall, this cross-disciplinary approach with molecular networking on data acquired in vitro and in silico prediction enabled to propose relevant candidate as AP-237 consumption markers that could be added to mass spectrometry libraries to help diagnose intoxication.
Collapse
Affiliation(s)
- Romain Pelletier
- NuMeCan Institute (Nutrition, Metabolisms and Cancer), CHU Rennes, Univ Rennes, INSERM, INRAE, UMR_A 1341, UMR_S 1317, 35033, Rennes, France.
- Clinical and Forensic Toxicology Laboratory, Rennes University Hospital, 35033, Rennes, France.
| | - Alexis Bourdais
- NuMeCan Institute (Nutrition, Metabolisms and Cancer), CHU Rennes, Univ Rennes, INSERM, INRAE, UMR_A 1341, UMR_S 1317, 35033, Rennes, France
| | - Nicolas Fabresse
- Laboratory of Pharmacokinetics and Toxicology, La Timone University Hospital, 264 rue Saint Pierre, 13385, Marseille Cedex 5, France
- Aix Marseille University, INSERM, IRD, SESSTIM, Economic and Social Sciences of Health and Medical Information Processing, Marseille, France
| | - Pierre-Jean Ferron
- NuMeCan Institute (Nutrition, Metabolisms and Cancer), CHU Rennes, Univ Rennes, INSERM, INRAE, UMR_A 1341, UMR_S 1317, 35033, Rennes, France
| | - Isabelle Morel
- NuMeCan Institute (Nutrition, Metabolisms and Cancer), CHU Rennes, Univ Rennes, INSERM, INRAE, UMR_A 1341, UMR_S 1317, 35033, Rennes, France
- Clinical and Forensic Toxicology Laboratory, Rennes University Hospital, 35033, Rennes, France
| | - Thomas Gicquel
- NuMeCan Institute (Nutrition, Metabolisms and Cancer), CHU Rennes, Univ Rennes, INSERM, INRAE, UMR_A 1341, UMR_S 1317, 35033, Rennes, France
- Clinical and Forensic Toxicology Laboratory, Rennes University Hospital, 35033, Rennes, France
| | - Brendan Le Daré
- NuMeCan Institute (Nutrition, Metabolisms and Cancer), CHU Rennes, Univ Rennes, INSERM, INRAE, UMR_A 1341, UMR_S 1317, 35033, Rennes, France
- Pharmacy Department, Rennes University Hospital, 35033, Rennes, France
| |
Collapse
|
24
|
Du X, Zou R, Du K, Huang D, Miao C, Qiu B, Ding W, Li C. Modeling Colorectal Cancer-Induced Liver Portal Vein Microthrombus on a Hepatic Lobule Chip. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 38033197 DOI: 10.1021/acsami.3c14417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Colorectal cancer is one of the most common malignant tumors. At the advanced stage of colorectal cancer, cancer cells migrate with the blood to the liver from the hepatic portal vein, eventually resulting in a portal vein tumor thrombus (PVTT). To date, the progression of the early onset of PVTT [portal vein microthrombus (PVmT) induced by tumors] is unclear. Herein, we developed an on-chip PVmT model by loading the spheroid of colorectal cancer cells into the portal vein of a hepatic lobule chip (HLC). On the HLC, the progression of PVmT was presented, and early changes in metabolites of hepatic cells and in structures of hepatic plates and sinusoids induced by PVmT were analyzed. We replicated intrahepatic angiogenesis, thickened blood vessels, an increased number of hepatocytes, disordered hepatic plates, and decreased concentrations of biomarkers of hepatic cell functions in PVmT progression on a microfluidic chip for the first time. In addition, the combined therapy of thermo-ablation and chemo-drug for PVmT was preliminarily demonstrated. This study provides a promising method for understanding PVTT evolution and offers a valuable reference for PVTT therapy.
Collapse
Affiliation(s)
- Xiaofang Du
- School of Information Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Rong Zou
- School of Information Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Kun Du
- Department of Medical Equipment, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Dabing Huang
- Department of Oncology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Chunguang Miao
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei 230027, China
| | - Bensheng Qiu
- School of Information Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Weiping Ding
- Department of Oncology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Chengpan Li
- School of Information Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
- Department of Oncology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| |
Collapse
|
25
|
Kaden T, Graf K, Rennert K, Li R, Mosig AS, Raasch M. Evaluation of drug-induced liver toxicity of trovafloxacin and levofloxacin in a human microphysiological liver model. Sci Rep 2023; 13:13338. [PMID: 37587168 PMCID: PMC10432496 DOI: 10.1038/s41598-023-40004-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 08/03/2023] [Indexed: 08/18/2023] Open
Abstract
Drug-induced liver injury induced by already approved substances is a major threat to human patients, potentially resulting in drug withdrawal and substantial loss of financial resources in the pharmaceutical industry. Trovafloxacin, a broad-spectrum fluoroquinolone, was found to have unexpected side effects of severe hepatotoxicity, which was not detected by preclinical testing. To address the limitations of current drug testing strategies mainly involving 2D cell cultures and animal testing, a three-dimensional microphysiological model of the human liver containing expandable human liver sinusoidal endothelial cells, monocyte-derived macrophages and differentiated HepaRG cells was utilized to investigate the toxicity of trovafloxacin and compared it to the structurally-related non-toxic drug levofloxacin. In the model, trovafloxacin elicited vascular and hepatocellular toxicity associated with pro-inflammatory cytokine release already at clinically relevant concentrations, whereas levofloxacin did not provoke tissue injury. Similar to in vivo, cytokine secretion was dependent on a multicellular immune response, highlighting the potential of the complex microphysiological liver model for reliably detecting drug-related cytotoxicity in preclinical testing. Moreover, hepatic glutathione depletion and mitochondrial ROS formation were elucidated as intrinsic toxicity mechanisms contributing to trovafloxacin toxicity.
Collapse
Affiliation(s)
- Tim Kaden
- Dynamic42 GmbH, Jena, Germany
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | | | | | - Ruoya Li
- Biopredic International, St Gregoire, France
| | - Alexander S Mosig
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | | |
Collapse
|
26
|
Morgan K, Morley SD, Raja AK, Vandeputte M, Samuel K, Waterfall M, Homer NZM, Hayes PC, Fallowfield JA, Plevris JN. Metabolism of Acetaminophen by Enteric Epithelial Cells Mitigates Hepatocellular Toxicity In Vitro. J Clin Med 2023; 12:3995. [PMID: 37373688 DOI: 10.3390/jcm12123995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/25/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
The gut-liver axis is defined by dietary and environmental communication between the gut, microbiome and the liver with its redox and immune systems, the overactivation of which can lead to hepatic injury. We used media preconditioning to mimic some aspects of the enterohepatic circulation by treating the human Caco-2 intestinal epithelial cell line with 5, 10 and 20 mM paracetamol (N-acetyl-para-aminophenol; APAP) for 24 h, after which cell culture supernatants were transferred to differentiated human hepatic HepaRG cells for a further 24 h. Cell viability was assessed by mitochondrial function and ATP production, while membrane integrity was monitored by cellular-based impedance. Metabolism by Caco-2 cells was determined by liquid chromatography with tandem mass spectrometry. Caco-2 cell viability was not affected by APAP, while cell membrane integrity and tight junctions were maintained and became tighter with increasing APAP concentrations, suggesting a reduction in the permeability of the intestinal epithelium. During 24 h incubation, Caco-2 cells metabolised 64-68% of APAP, leaving 32-36% of intact starting compound to be transferred to HepaRG cells. When cultured with Caco-2-preconditioned medium, HepaRG cells also showed no loss of cell viability or membrane integrity, completely in contrast to direct treatment with APAP, which resulted in a rapid loss of cell viability and membrane integrity and, ultimately, cell death. Thus, the pre-metabolism of APAP could mitigate previously observed hepatotoxicity to hepatic tight junctions caused by direct exposure to APAP. These observations could have important implications for the direct exposure of hepatic parenchyma to APAP, administered via the intravenous route.
Collapse
Affiliation(s)
- Katie Morgan
- Hepatology Laboratory, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Steven D Morley
- Hepatology Laboratory, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Arslan K Raja
- Hepatology Laboratory, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Martin Vandeputte
- Hepatology Laboratory, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Kay Samuel
- Scottish Blood Transfusion Service, Jack Copland Centre, 52 Research Avenue North, Edinburgh EH14 4BE, UK
| | - Martin Waterfall
- Flow Cytometry Facility, Ashworth Laboratories, Institute of Immunology & Infection Research, The University of Edinburgh, The Kings Buildings, Edinburgh EH9 3FL, UK
| | - Natalie Z M Homer
- Mass Spectrometry Facility, Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Peter C Hayes
- Hepatology Laboratory, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Jonathan A Fallowfield
- Hepatology Laboratory, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Institute for Regeneration and Repair, Edinburgh BioQuarter, The University of Edinburgh, 4-5 Little France Drive, Edinburgh EH16 4UU, UK
| | - John N Plevris
- Hepatology Laboratory, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| |
Collapse
|
27
|
Ivanova ON, Krasnov GS, Snezhkina AV, Kudryavtseva AV, Fedorov VS, Zakirova NF, Golikov MV, Kochetkov SN, Bartosch B, Valuev-Elliston VT, Ivanov AV. Transcriptome Analysis of Redox Systems and Polyamine Metabolic Pathway in Hepatoma and Non-Tumor Hepatocyte-like Cells. Biomolecules 2023; 13:714. [PMID: 37189460 PMCID: PMC10136275 DOI: 10.3390/biom13040714] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/10/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
Reactive oxygen species (ROS) play a major role in the regulation of various processes in the cell. The increase in their production is a factor contributing to the development of numerous pathologies, including inflammation, fibrosis, and cancer. Accordingly, the study of ROS production and neutralization, as well as redox-dependent processes and the post-translational modifications of proteins, is warranted. Here, we present a transcriptomic analysis of the gene expression of various redox systems and related metabolic processes, such as polyamine and proline metabolism and the urea cycle in Huh7.5 hepatoma cells and the HepaRG liver progenitor cell line, that are widely used in hepatitis research. In addition, changes in response to the activation of polyamine catabolism that contribute to oxidative stress were studied. In particular, differences in the gene expression of various ROS-producing and ROS-neutralizing proteins, the enzymes of polyamine metabolisms and proline and urea cycles, as well as calcium ion transporters between cell lines, are shown. The data obtained are important for understanding the redox biology of viral hepatitis and elucidating the influence of the laboratory models used.
Collapse
Affiliation(s)
- Olga N. Ivanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - George S. Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasiya V. Snezhkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anna V. Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Vyacheslav S. Fedorov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Natalia F. Zakirova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Michail V. Golikov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Sergey N. Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Birke Bartosch
- Lyon Cancer Research Center, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, 69008 Lyon, France
| | | | - Alexander V. Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
28
|
Ma Y, Hu L, Tang J, Guo W, Feng Y, Liu Y, Tang F. Three-Dimensional Cell Co-Culture Liver Models and Their Applications in Pharmaceutical Research. Int J Mol Sci 2023; 24:ijms24076248. [PMID: 37047220 PMCID: PMC10094553 DOI: 10.3390/ijms24076248] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/14/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
As the primary site for the biotransformation of drugs, the liver is the most focused on organ type in pharmaceutical research. However, despite being widely used in pharmaceutical research, animal models have inherent species differences, while two-dimensional (2D) liver cell monocultures or co-cultures and three-dimensional (3D) liver cell monoculture in vitro liver models do not sufficiently represent the complexity of the human liver’s structure and function, making the evaluation results from these tools less reliable. Therefore, there is a pressing need to develop more representative in vitro liver models for pharmaceutical research. Fortunately, an exciting new development in recent years has been the emergence of 3D liver cell co-culture models. These models hold great promise as in vitro pharmaceutical research tools, because they can reproduce liver structure and function more practically. This review begins by explaining the structure and main cell composition of the liver, before introducing the potential advantages of 3D cell co-culture liver models for pharmaceutical research. We also discuss the main sources of hepatocytes and the 3D cell co-culture methods used in constructing these models. In addition, we explore the applications of 3D cell co-culture liver models with different functional states and suggest prospects for their further development.
Collapse
|
29
|
Bi Y, Wang X, Ding H, He F, Han L, Zhang Y. Transporter-mediated Natural Product-Drug Interactions. PLANTA MEDICA 2023; 89:119-133. [PMID: 35304735 DOI: 10.1055/a-1803-1744] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The increasing use of natural products in clinical practice has raised great concerns about the potential natural product-drug interactions (NDIs). Drug transporters mediate the transmembrane passage of a broad range of drugs, and thus are important determinants for drug pharmacokinetics and pharmacodynamics. Generally, transporters can be divided into ATP binding cassette (ABC) family and solute carrier (SLC) family. Numerous natural products have been identified as inhibitors, substrates, inducers, and/or activators of drug transporters. This review article aims to provide a comprehensive summary of the recent progress on the research of NDIs, focusing on the main drug transporters, such as P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), organic anion transporter 1 and 3 (OAT1/OAT3), organic anion-transporting polypeptide 1B1 and 1B3 (OATP1B1/OATP1B3), organic cation transporter 2 (OCT2), multidrug and toxin extrusion protein 1 and 2-K (MATE1/MATE2-K). Additionally, the challenges and strategies of studying NDIs are also discussed.
Collapse
Affiliation(s)
- Yajuan Bi
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P. R. China
| | - Xue Wang
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, USA
| | - Hui Ding
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Feng He
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Lifeng Han
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Youcai Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P. R. China
| |
Collapse
|
30
|
Le Guilcher C, Merlen G, Dellaquila A, Labour MN, Aid R, Tordjmann T, Letourneur D, Simon-Yarza T. Engineered human liver based on pullulan-dextran hydrogel promotes mice survival after liver failure. Mater Today Bio 2023; 19:100554. [PMID: 36756209 PMCID: PMC9900439 DOI: 10.1016/j.mtbio.2023.100554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Liver tissue engineering approaches aim to support drug testing, assistance devices, or transplantation. However, their suitability for clinical application remains unsatisfactory. Herein, we demonstrate the beneficial and biocompatible use of porous pullulan-dextran hydrogel for the self-assembly of hepatocytes and biliary-like cells into functional 3D microtissues. Using HepaRG cells, we obtained 21 days maintenance of engineered liver polarity, functional detoxification and excretion systems, as well as glycogen storage in hydrogel. Implantation on two liver lobes in mice of hydrogels containing 3800 HepaRG 3D structures of 100 μm in diameter, indicated successful engraftment and no signs of liver toxicity after one month. Finally, after acetaminophen-induced liver failure, when mice were transplanted with engineered livers on left lobe and peritoneal cavity, the survival rate at 7 days significantly increased by 31.8% compared with mice without cell therapy. These findings support the clinical potential of pullulan-dextran hydrogel for liver failure management.
Collapse
Affiliation(s)
- Camille Le Guilcher
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France,Corresponding author.
| | - Grégory Merlen
- Université Paris-Saclay, INSERM U1193, F- 94800 Villejuif, France
| | - Alessandra Dellaquila
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France
| | - Marie-Noëlle Labour
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France,ICGM, Université de Montpellier, CNRS, ENSCM, F- 34293 Montpellier, France,École Pratique des Hautes Études, Université Paris Sciences et Lettres, F-75014 Paris, France
| | - Rachida Aid
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France
| | | | - Didier Letourneur
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France,Corresponding author.
| | - Teresa Simon-Yarza
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, U1148, F-75018 Paris, France,Corresponding author.
| |
Collapse
|
31
|
Bellanti F, Mangieri D, di Bello G, Lo Buglio A, Pannone G, Pedicillo MC, Fersini A, Dobrakowski M, Kasperczyk A, Kasperczyk S, Vendemiale G. Redox-Dependent Modulation of Human Liver Progenitor Cell Line Fate. Int J Mol Sci 2023; 24:1934. [PMID: 36768260 PMCID: PMC9916526 DOI: 10.3390/ijms24031934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Redox homeostasis is determinant in the modulation of quiescence/self-renewal/differentiation of stem cell lines. The aim of this study consisted of defining the impact of redox modifications on cell fate in a human hepatic progenitor line. To achieve this, the HepaRG cell line, which shows oval ductular bipotent characteristics, was used. The impact of redox status on the balance between self-renewal and differentiation of HepaRG cells was investigated using different methodological approaches. A bioinformatic analysis initially proved that the trans-differentiation of HepaRG toward bipotent progenitors is associated with changes in redox metabolism. We then exposed confluent HepaRG (intermediate differentiation phase) to oxidized (H2O2) or reduced (N-acetylcysteine) extracellular environments, observing that oxidation promotes the acquisition of a mature HepaRG phenotype, while a reduced culture medium stimulates de-differentiation. These results were finally confirmed through pharmacological modulation of the nuclear factor (erythroid-derived 2)-like 2 (NRF2), a principal modulator of the antioxidant response, in confluent HepaRG. NRF2 inhibition led to intracellular pro-oxidative status and HepaRG differentiation, while its activation was associated with low levels of reactive species and de-differentiation. In conclusion, this study shows that both intra- and extracellular redox balance are crucial in the determination of HepaRG fate. The impact of redox status in the differentiation potential of HepaRG cells is significant on the utilization of this cell line in pre-clinical studies.
Collapse
Affiliation(s)
- Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Domenica Mangieri
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Giorgia di Bello
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Aurelio Lo Buglio
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Giuseppe Pannone
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | | | - Alberto Fersini
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Michał Dobrakowski
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-800 Katowice, Poland
| | - Aleksandra Kasperczyk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-800 Katowice, Poland
| | - Sławomir Kasperczyk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-800 Katowice, Poland
| | - Gianluigi Vendemiale
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
32
|
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: 2] [Impact Index Per Article: 0.7] [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
|
33
|
A Transversal Approach Combining In Silico, In Vitro and In Vivo Models to Describe the Metabolism of the Receptor Interacting Protein 1 Kinase Inhibitor Sibiriline. Pharmaceutics 2022; 14:pharmaceutics14122665. [PMID: 36559159 PMCID: PMC9787481 DOI: 10.3390/pharmaceutics14122665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/21/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022] Open
Abstract
Sibiriline is a novel drug inhibiting receptor-interacting protein 1 kinase (RIPK1) and necroptosis, a regulated form of cell death involved in several disease models. In this study, we aimed to investigate the metabolic fate of sibiriline in a cross-sectional manner using an in silico prediction, coupled with in vitro and in vivo experiments. In silico predictions were performed using GLORYx and Biotransformer 3.0 freeware; in vitro incubation was performed on differentiated human HepaRG cells, and in vivo experiments including a pharmacokinetic study were performed on mice treated with sibiriline. HepaRG culture supernatants and mice plasma samples were analyzed with ultra-high-performance liquid chromatography, coupled with tandem mass spectrometry (LC-HRMS/MS). The molecular networking bioinformatics tool applied to LC-HRMS/MS data allowed us to visualize the sibiriline metabolism kinetics. Overall, 14 metabolites, mostly produced by Phase II transformations (glucuronidation and sulfation) were identified. These data provide initial reassurance regarding the toxicology of this new RIPK1 inhibitor, although further studies are required.
Collapse
|
34
|
Hammour MM, Othman A, Aspera-Werz R, Braun B, Weis-Klemm M, Wagner S, Nadalin S, Histing T, Ruoß M, Nüssler AK. Optimisation of the HepaRG cell line model for drug toxicity studies using two different cultivation conditions: advantages and limitations. Arch Toxicol 2022; 96:2511-2521. [PMID: 35748891 DOI: 10.1007/s00204-022-03329-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 06/07/2022] [Indexed: 11/26/2022]
Abstract
The HepaRG cell line represents a successful model for hepatotoxicity studies. These cells are of human origin and are differentiated in vitro into mature and functional hepatocyte-like cells. The objective of this research was to compare two different culture protocols, Sison-Young et al. 2017 (hereinafter referred as Sison) and Gripon et al. 2002 (hereinafter referred as Biopredic) for HepaRG cells in order to optimise this model for drug metabolism and toxicity testing studies. HepaRG cells obtained from the same batch were cultured according to the described protocols. Using both protocols, differentiated HepaRG cells retained their drug metabolic capacity (major phase I/II enzymes) and transporters, as well as their morphological characteristics. Morphologically, HepaRG cells cultured after the Biopredic protocol formed more apical membranes and small ductular-like structures, than those cultivated using the Sison protocol. Also, the efflux activity of multidrug resistance protein 1 (MDR1) and multidrug resistance-associated protein 1 (MRP1) as well as the activity of uridine-glucuronosyltransferase (UGT) and glutathione S-transferase (GST) were significantly reduced in HepaRG cultured using the Sison protocol. Applying well-established drug cocktails to measure cytochrome P450 (CYPs) activity, we found that production of the corresponding metabolites was hampered in Sison-cultured HepaRG cells, indicating that the activity of CYP1A2, CYP2C9, CYP3A4, CYP2B6 and CYP2C19 was significantly reduced. Moreover, HepaRG sensitivity to well-known drugs, namely diclofenac, amiodarone, imipramine and paracetamol, revealed some differences between the two culture protocols. Furthermore, the HepaRG cells can be maintained with higher viability and sufficient CYPs activity and expression (i.e. CYP3A4, CYP1A2 and CYP2B6) as well as liver-specific functions, using Biopredic compared with the Sison culture protocol. These maintained liver-specific functions might be dependent on the prolongation of the culture conditions in the case of the Biopredic protocol. In conclusion, based on the metabolic activity of HepaRG cells using the standard protocol from Biopredic, we believe that this protocol is optimal for investigating drug metabolism and pharmacokinetic screening studies.
Collapse
Affiliation(s)
- Mohammad Majd Hammour
- Department of Traumatology, Siegfried Weller Institute, BG Klinik Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Amnah Othman
- Department of Traumatology, Siegfried Weller Institute, BG Klinik Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Romina Aspera-Werz
- Department of Traumatology, Siegfried Weller Institute, BG Klinik Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Bianca Braun
- Department of Traumatology, Siegfried Weller Institute, BG Klinik Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Michaela Weis-Klemm
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Silvia Wagner
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Silvio Nadalin
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Tina Histing
- Department of Traumatology, Siegfried Weller Institute, BG Klinik Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Marc Ruoß
- Department of Traumatology, Siegfried Weller Institute, BG Klinik Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Andreas K Nüssler
- Department of Traumatology, Siegfried Weller Institute, BG Klinik Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany.
| |
Collapse
|
35
|
Scheffschick A, Babel J, Sperling S, Nerusch J, Herzog N, Seehofer D, Damm G. Primary-like Human Hepatocytes Genetically Engineered to Obtain Proliferation Competence as a Capable Application for Energy Metabolism Experiments in In Vitro Oncologic Liver Models. BIOLOGY 2022; 11:biology11081195. [PMID: 36009822 PMCID: PMC9405410 DOI: 10.3390/biology11081195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/21/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022]
Abstract
Simple Summary Fatty liver disease is an increasing health concern in Westernized countries. A fatty liver can lead to hepatocellular carcinoma (HCC), a type of liver cancer arising from hepatocytes, the major cells of the liver. How HCC may develop from the fatty liver is not known, and good cellular systems to investigate this are lacking. Recently, hepatocytes that can multiply continuously have been generated and suggested for hepatocyte research. In this study, we compared these continuously multiplying human hepatocytes to normal human hepatocytes and liver cancer cells, both within the state of fatty liver or not. We identified that these multiplying hepatocytes displayed many similarities to the liver cancer cells in terms of energy metabolism and concluded that these hepatocytes could be a pre-cancer model for liver cancer research and would be a valuable tool for HCC research. Abstract Non-alcoholic fatty liver disease (NAFLD), characterized by lipid accumulation in the liver, is the most common cause of liver diseases in Western countries. NAFLD is a major risk factor for developing hepatocellular carcinoma (HCC); however, in vitro evaluation of hepatic cancerogenesis fails due to a lack of liver models displaying a proliferation of hepatocytes. Originally designed to overcome primary human hepatocyte (PHH) shortages, upcyte hepatocytes were engineered to obtain continuous proliferation and, therefore, could be a suitable tool for HCC research. We generated upcyte hepatocytes, termed HepaFH3 cells, and compared their metabolic characteristics to HepG2 hepatoma cells and PHHs isolated from resected livers. For displaying NAFLD-related HCCs, we induced steatosis in all liver models. Lipid accumulation, lipotoxicity and energy metabolism were characterized using biochemical assays and Western blot analysis. We showed that proliferating HepaFH3 cells resemble HepG2, both showing a higher glucose uptake rate, lactate levels and metabolic rate compared to PHHs. Confluent HepaFH3 cells displayed some similarities to PHHs, including higher levels of the transaminases AST and ALT compared to proliferating HepaFH3 cells. We recommend proliferating HepaFH3 cells as a pre-malignant cellular model for HCC research, while confluent HepaFH3 cells could serve as PHH surrogates for energy metabolism studies.
Collapse
Affiliation(s)
- Andrea Scheffschick
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
| | - Jonas Babel
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany
| | - Sebastian Sperling
- Department of General, Visceral and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Julia Nerusch
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
| | - Natalie Herzog
- Faculty of Science, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany
- Department of General, Visceral and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
- Department of General, Visceral and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
- Correspondence: ; Tel.: +49-341-97-39656
| |
Collapse
|
36
|
Zuo Q, Xu W, Wan Y, Feng D, He C, Lin C, Huang D, Chen F, Han L, Sun Q, Chen D, Du H, Huang L. Efficient generation of a CYP3A4-T2A-luciferase knock-in HepaRG subclone and its optimized differentiation. Biomed Pharmacother 2022; 152:113243. [PMID: 35687910 DOI: 10.1016/j.biopha.2022.113243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 11/02/2022] Open
Abstract
CRISPR/Cas9 has allowed development of better and easier-to-use ADME models than traditional methods by complete knockout or knock-in of genes. However, gene editing in HepaRG cells remains challenging because long-term monoclonal cultivation may alter their differentiation capacity to a large extent. Here, CRISPR/Cas9 was used to generate a CYP3A4-T2A-luciferase knock-in HepaRG subclone by Cas9-mediated homologous recombination and monoclonal cultivation. The knock-in HepaRG-#9 subclone retained a similar differentiation potential to wildtype HepaRG cells (HepaRG-WT). To further improve differentiation and expand the applications of knock-in HepaRG cells, two optimized differentiation procedures were evaluated by comparison with the standard differentiation procedure using the knock-in HepaRG-#9 subclone and HepaRG-WT. The results indicated that addition of forskolin (an adenylate cyclase activator) and SB431542 (a TGF-β pathway inhibitor) to the first optimized differentiation procedure led to better differentiation consequence in terms of not only the initiation time for differentiation and morphological characterization, but also the mRNA levels of hepatocyte-specific genes. These data may contribute to more extensive applications of genetically modified HepaRG cells in ADME studies.
Collapse
Affiliation(s)
- Qingxia Zuo
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Wanqing Xu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Yanbin Wan
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Dongyan Feng
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Changsheng He
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Cailing Lin
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Dongchao Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Feng Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Liya Han
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Qi Sun
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Dong Chen
- Fangrui Institute of Innovative Drugs, South China University of Technology, Guangzhou 510006, China
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Lizhen Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China.
| |
Collapse
|
37
|
Dubois-Pot-Schneider H, Aninat C, Kattler K, Fekir K, Jarnouen K, Cerec V, Glaise D, Salhab A, Gasparoni G, Takashi K, Ishida S, Walter J, Corlu A. Transcriptional and Epigenetic Consequences of DMSO Treatment on HepaRG Cells. Cells 2022; 11:cells11152298. [PMID: 35892596 PMCID: PMC9331440 DOI: 10.3390/cells11152298] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
Dimethyl sulfoxide (DMSO) is used to sustain or favor hepatocyte differentiation in vitro. Thus, DMSO is used in the differentiation protocol of the HepaRG cells that present the closest drug-metabolizing enzyme activities to primary human hepatocytes in culture. The aim of our study is to clarify its influence on liver-specific gene expression. For that purpose, we performed a large-scale analysis (gene expression and histone modification) to determine the global role of DMSO exposure during the differentiation process of the HepaRG cells. The addition of DMSO drives the upregulation of genes mainly regulated by PXR and PPARα whereas genes not affected by this addition are regulated by HNF1α, HNF4α, and PPARα. DMSO-differentiated-HepaRG cells show a differential expression for genes regulated by histone acetylation, while differentiated-HepaRG cells without DMSO show gene signatures associated with histone deacetylases. In addition, we observed an interplay between cytoskeleton organization and EMC remodeling with hepatocyte maturation.
Collapse
Affiliation(s)
- Hélène Dubois-Pot-Schneider
- INSERM, Université de Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France; (C.A.); (K.F.); (K.J.); (V.C.); (D.G.); (A.C.)
- Correspondence: ; Tel.: +33-372746115
| | - Caroline Aninat
- INSERM, Université de Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France; (C.A.); (K.F.); (K.J.); (V.C.); (D.G.); (A.C.)
| | - Kathrin Kattler
- Department of Genetics, University of Saarland (UdS), 66123 Saarbrücken, Germany; (K.K.); (A.S.); (G.G.); (J.W.)
| | - Karim Fekir
- INSERM, Université de Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France; (C.A.); (K.F.); (K.J.); (V.C.); (D.G.); (A.C.)
| | - Kathleen Jarnouen
- INSERM, Université de Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France; (C.A.); (K.F.); (K.J.); (V.C.); (D.G.); (A.C.)
| | - Virginie Cerec
- INSERM, Université de Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France; (C.A.); (K.F.); (K.J.); (V.C.); (D.G.); (A.C.)
| | - Denise Glaise
- INSERM, Université de Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France; (C.A.); (K.F.); (K.J.); (V.C.); (D.G.); (A.C.)
| | - Abdulrahman Salhab
- Department of Genetics, University of Saarland (UdS), 66123 Saarbrücken, Germany; (K.K.); (A.S.); (G.G.); (J.W.)
| | - Gilles Gasparoni
- Department of Genetics, University of Saarland (UdS), 66123 Saarbrücken, Germany; (K.K.); (A.S.); (G.G.); (J.W.)
| | - Kubo Takashi
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki-ku, Kawasaki 2109501, Japan; (K.T.); (S.I.)
| | - Seiichi Ishida
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki-ku, Kawasaki 2109501, Japan; (K.T.); (S.I.)
| | - Jörn Walter
- Department of Genetics, University of Saarland (UdS), 66123 Saarbrücken, Germany; (K.K.); (A.S.); (G.G.); (J.W.)
| | - Anne Corlu
- INSERM, Université de Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000 Rennes, France; (C.A.); (K.F.); (K.J.); (V.C.); (D.G.); (A.C.)
| |
Collapse
|
38
|
de Bruijn VMP, Wang Z, Bakker W, Zheng W, Spee B, Bouwmeester H. Hepatic bile acid synthesis and secretion: Comparison of in vitro methods. Toxicol Lett 2022; 365:46-60. [PMID: 35724847 DOI: 10.1016/j.toxlet.2022.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/30/2022] [Accepted: 06/09/2022] [Indexed: 12/12/2022]
Abstract
Reliable hepatic in vitro systems are crucial for the safety assessment of xenobiotics. Certain xenobiotics decrease the hepatic bile efflux, which can ultimately result in cholestasis. Preclinical animal models and the currently available in vitro systems poorly predict a xenobiotic's cholestatic potential. Here, we compared the phenotype and capacity of three liver derived in vitro systems to emulate human functionality to synthesize and secrete bile acids (BAs). To this end, basal BA production of sandwich cultured human hepatocytes (SCHHs), HepaRG cells (HepaRGs) and hepatocyte-like intrahepatic cholangiocyte organoids (ICO-heps) were analysed, and the effect of the known BSEP (Bile Salt Export Pump)-inhibitors bosentan and lopinavir on BA disposition in SCHHs and HepaRGs was quantified. RT-qPCR of selected target genes involved in maturation status, synthesis, transport and conjugation of BAs was performed to mechanistically underpin the observed differences in BA homeostasis. ICO-heps produced a (very) low amount of BAs. SCHHs are a powerful tool in cholestasis-testing due to their high basal BA production and high transporter expression compared to the other models tested. HepaRGs were responsive to both selected BSEP-inhibitors and produced a BA profile that is most similar to the human in vivo situation, making them a suitable and practical candidate for cholestasis-testing.
Collapse
Affiliation(s)
| | - Zhenguo Wang
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Wouter Bakker
- Division of Toxicology, Wageningen University & Research, the Netherlands
| | - Weijia Zheng
- Division of Toxicology, Wageningen University & Research, the Netherlands
| | - Bart Spee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University & Research, the Netherlands
| |
Collapse
|
39
|
Sblano S, Cerchia C, Laghezza A, Piemontese L, Brunetti L, Leuci R, Gilardi F, Thomas A, Genovese M, Santi A, Tortorella P, Paoli P, Lavecchia A, Loiodice F. A chemoinformatics search for peroxisome proliferator-activated receptors ligands revealed a new pan-agonist able to reduce lipid accumulation and improve insulin sensitivity. Eur J Med Chem 2022; 235:114240. [DOI: 10.1016/j.ejmech.2022.114240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/03/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022]
|
40
|
Molecular Networking for Drug Toxicities Studies: The Case of Hydroxychloroquine in COVID-19 Patients. Int J Mol Sci 2021; 23:ijms23010082. [PMID: 35008505 PMCID: PMC8744768 DOI: 10.3390/ijms23010082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/17/2021] [Accepted: 12/18/2021] [Indexed: 12/18/2022] Open
Abstract
Using drugs to treat COVID-19 symptoms may induce adverse effects and modify patient outcomes. These adverse events may be further aggravated in obese patients, who often present different illnesses such as metabolic-associated fatty liver disease. In Rennes University Hospital, several drug such as hydroxychloroquine (HCQ) have been used in the clinical trial HARMONICOV to treat COVID-19 patients, including obese patients. The aim of this study is to determine whether HCQ metabolism and hepatotoxicity are worsened in obese patients using an in vivo/in vitro approach. Liquid chromatography high resolution mass spectrometry in combination with untargeted screening and molecular networking were employed to study drug metabolism in vivo (patient’s plasma) and in vitro (HepaRG cells and RPTEC cells). In addition, HepaRG cells model were used to reproduce pathophysiological features of obese patient metabolism, i.e., in the condition of hepatic steatosis. The metabolic signature of HCQ was modified in HepaRG cells cultured under a steatosis condition and a new metabolite was detected (carboxychloroquine). The RPTEC model was found to produce only one metabolite. A higher cytotoxicity of HCQ was observed in HepaRG cells exposed to exogenous fatty acids, while neutral lipid accumulation (steatosis) was further enhanced in these cells. These in vitro data were compared with the biological parameters of 17 COVID-19 patients treated with HCQ included in the HARMONICOV cohort. Overall, our data suggest that steatosis may be a risk factor for altered drug metabolism and possibly toxicity of HCQ.
Collapse
|
41
|
Rose S, Cuvellier M, Ezan F, Carteret J, Bruyère A, Legagneux V, Nesslany F, Baffet G, Langouët S. DMSO-free highly differentiated HepaRG spheroids for chronic toxicity, liver functions and genotoxicity studies. Arch Toxicol 2021; 96:243-258. [PMID: 34762139 DOI: 10.1007/s00204-021-03178-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/06/2021] [Indexed: 12/23/2022]
Abstract
The liver is essential in the elimination of environmental and food contaminants. Given the interspecies differences between rodents and humans, the development of relevant in vitro human models is crucial to investigate liver functions and toxicity in cells that better reflect pathophysiological processes. Classically, the differentiation of the hepatic HepaRG cell line requires high concentration of dimethyl sulfoxide (DMSO), which restricts its usefulness for drug-metabolism studies. Herein, we describe undifferentiated HepaRG cells embedded in a collagen matrix in DMSO-free conditions that rapidly organize into polarized hollow spheroids of differentiated hepatocyte-like cells (Hepoid-HepaRG). Our conditions allow concomitant proliferation with high levels of liver-specific functions and xenobiotic metabolism enzymes expression and activities after a few days of culture and for at least 4 weeks. By studying the toxicity of well-known injury-inducing drugs by treating cells with 1- to 100-fold of their plasmatic concentrations, we showed appropriate responses and demonstrate the sensitivity to drugs known to induce various degrees of liver injury. Our results also demonstrated that the model is well suited to estimate cholestasis and steatosis effects of drugs following chronic treatment. Additionally, DNA alterations caused by four genotoxic compounds (Aflatoxin B1 (AFB1), Benzo[a]Pyrene (B[a]P), Cyclophosphamide (CPA) and Methyl methanesulfonate (MMS)) were quantified in a dose-dependent manner by the comet and micronucleus assays. Their genotoxic effects were significantly increased after either an acute 24 h treatment (AFB1: 1.5-6 μM, CPA: 2.5-10 μM, B[a]P: 12.5-50 μM, MMS: 90-450 μM) or after a 14-day treatment at much lower concentrations (AFB1: 0.05-0.2 μM, CPA: 0.125-0.5 μM, B[a]P: 0.125-0.5 μM) representative to human exposure. Altogether, the DMSO-free 3D culture of Hepoid-HepaRG provides highly differentiated and proliferating cells relevant for various toxicological in vitro assays, especially for drug-preclinical studies and environmental chemicals risk assessment.
Collapse
Affiliation(s)
- Sophie Rose
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Marie Cuvellier
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Frédéric Ezan
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Jennifer Carteret
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Arnaud Bruyère
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Vincent Legagneux
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Fabrice Nesslany
- Genotoxicology Department, Institut Pasteur de Lille, 1, Rue du Professeur Calmette, 59000, Lille, France
| | - Georges Baffet
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France.
| | - Sophie Langouët
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France.
| |
Collapse
|
42
|
Bellanti F, di Bello G, Tamborra R, Amatruda M, Lo Buglio A, Dobrakowski M, Kasperczyk A, Kasperczyk S, Serviddio G, Vendemiale G. Impact of senescence on the transdifferentiation process of human hepatic progenitor-like cells. World J Stem Cells 2021; 13:1595-1609. [PMID: 34786160 PMCID: PMC8567448 DOI: 10.4252/wjsc.v13.i10.1595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/14/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Senescence is characterized by a decline in hepatocyte function, with impairment of metabolism and regenerative capacity. Several models that duplicate liver functions in vitro are essential tools for studying drug metabolism, liver diseases, and organ regeneration. The human HepaRG cell line represents an effective model for the study of liver metabolism and hepatic progenitors. However, the impact of senescence on HepaRG cells is not yet known. AIM To characterize the effects of senescence on the transdifferentiation capacity and mitochondrial metabolism of human HepaRG cells. METHODS We compared the transdifferentiation capacity of cells over 10 (passage 10 [P10]) vs P20. Aging was evaluated by senescence-associated (SA) beta-galactosidase activity and the comet assay. HepaRG transdifferentiation was analyzed by confocal microscopy and flow cytometry (expression of cluster of differentiation 49a [CD49a], CD49f, CD184, epithelial cell adhesion molecule [EpCAM], and cytokeratin 19 [CK19]), quantitative PCR analysis (expression of albumin, cytochrome P450 3A4 [CYP3A4], γ-glutamyl transpeptidase [γ-GT], and carcinoembryonic antigen [CEA]), and functional analyses (albumin secretion, CYP3A4, and γ-GT). Mitochondrial respiration and the ATP and nicotinamide adenine dinucleotide (NAD+)/NAD with hydrogen (NADH) content were also measured. RESULTS SA β-galactosidase staining was higher in P20 than P10 HepaRG cells; in parallel, the comet assay showed consistent DNA damage in P20 HepaRG cells. With respect to P10, P20 HepaRG cells exhibited a reduction of CD49a, CD49f, CD184, EpCAM, and CK19 after the induction of transdifferentiation. Furthermore, lower gene expression of albumin, CYP3A4, and γ-GT, as well as reduced albumin secretion capacity, CYP3A4, and γ-GT activity were reported in transdifferentiated P20 compared to P10 cells. By contrast, the gene expression level of CEA was not reduced by transdifferentiation in P20 cells. Of note, both cellular and mitochondrial oxygen consumption was lower in P20 than in P10 transdifferentiated cells. Finally, both ATP and NAD+/NADH were depleted in P20 cells with respect to P10 cells. CONCLUSION SA mitochondrial dysfunction may limit the transdifferentiation potential of HepaRG cells, with consequent impairment of metabolic and regenerative properties, which may alter applications in basic studies.
Collapse
Affiliation(s)
- Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy.
| | - Giorgia di Bello
- Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy
| | - Rosanna Tamborra
- Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy
| | - Marco Amatruda
- Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy
| | - Aurelio Lo Buglio
- Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy
| | - Michał Dobrakowski
- Department of Biochemistry, Medical University of Silesia, Zabrze 41-808, Poland
| | | | - Sławomir Kasperczyk
- Department of Biochemistry, Medical University of Silesia, Zabrze 41-808, Poland
| | - Gaetano Serviddio
- Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy
| | - Gianluigi Vendemiale
- Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy
| |
Collapse
|
43
|
Du K, Li S, Li C, Li P, Miao C, Luo T, Qiu B, Ding W. Modeling nonalcoholic fatty liver disease on a liver lobule chip with dual blood supply. Acta Biomater 2021; 134:228-239. [PMID: 34265474 DOI: 10.1016/j.actbio.2021.07.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/21/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as a public health concern. To date, the mechanism of NAFLD progression remains unclear, and pharmacological treatment options are scarce. Traditional animal NAFLD models are limited in helping address these problems due to interspecies differences. Liver chips are promising for modeling NAFLD. However, pre-existing liver chips cannot reproduce complex physicochemical microenvironments of the liver effectively; thus, NAFLD modeling based on these chips is incomplete. Herein, we develop a biomimetic liver lobule chip (LC) and then establish a more accurate on-chip NAFLD model. The self-developed LC achieves dual blood supply through the designed hepatic portal vein and hepatic artery and the microtissue cultured on the LC forms multiple structures similar to in vivo liver. Based on the LC, NAFLD is modeled. Steatosis is successfully induced and more importantly, changing lipid zonation in a liver lobule with the progression of NAFLD is demonstrated for the first time on a microfluidic chip. In addition, the application of the induced NAFLD model has been preliminarily demonstrated in the prevention and reversibility of promising drugs. This study provides a promising platform to understand NAFLD progression and identify drugs for treating NAFLD. STATEMENT OF SIGNIFICANCE: Liver chips are promising for modeling nonalcoholic fatty liver disease. However, on-chip replicating liver physicochemical microenvironments is still a challenge. Herein, we developed a liver lobule chip with dual blood supply, achieving self-organized liver microtissue that is similar to in vivo tissue. Based on the chip, we successfully modeled NAFLD under physiologically differentiated nutrient supplies. For the first time, the changing lipid zonation in a single liver lobule with the early-stage progression of NAFLD was demonstrated on a liver chip. This study provides a promising platform for modeling liver-related diseases.
Collapse
Affiliation(s)
- Kun Du
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, China
| | - Shibo Li
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, China
| | - Chengpan Li
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, China
| | - Ping Li
- Chinese Integrative Medicine Oncology Department, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Chunguang Miao
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, China
| | - Tianzhi Luo
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, 230027, China
| | - Bensheng Qiu
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, China.
| | - Weiping Ding
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
44
|
The Performance of HepG2 and HepaRG Systems through the Glass of Acetaminophen-Induced Toxicity. Life (Basel) 2021; 11:life11080856. [PMID: 34440600 PMCID: PMC8400973 DOI: 10.3390/life11080856] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/07/2021] [Accepted: 08/18/2021] [Indexed: 12/30/2022] Open
Abstract
Investigation of drug-induced liver injuries requires appropriate in vivo and in vitro toxicological model systems. In our study, an attempt was made to compare the hepatocarcinoma HepG2 and the stem cell-derived HepaRG cell lines both in two- and three-dimensional culture conditions to find the most suitable model. Comparison of the liver-specific characteristics of these models was performed via the extent and mechanism of acetaminophen (APAP)-induced hepatotoxicity. Investigating the detailed mechanism of APAP-induced hepatotoxicity, different specific cell death inhibitors were used: the pan-caspase inhibitor zVAD-fmk and dabrafenib significantly protected both cell lines from APAP-induced cell death. However, the known specific inhibitors of necroptosis (necrostatin-1 and MDIVI) were only effective in differentiated HepaRG, which suggest a differential execution of activated pathways in the two models. By applying 3D culture methods, CYP2E1 mRNA levels could be elevated, but we failed to achieve a significant increase in hepatocyte function; hence, the 3D cultivation especially in APAP toxicity studies is not necessarily worth the complicated maintenance. Based on our findings, the hepatocyte functions of HepaRG may stand between the properties of HepG2 cells and primary hepatocytes (PHHs). However, it should be noted that in contrast to PHHs having many limitations, HepaRG cells are relatively immortal, having a stable phenotype and CYP450 expression.
Collapse
|
45
|
Polidoro MA, Ferrari E, Marzorati S, Lleo A, Rasponi M. Experimental liver models: From cell culture techniques to microfluidic organs-on-chip. Liver Int 2021; 41:1744-1761. [PMID: 33966344 DOI: 10.1111/liv.14942] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022]
Abstract
The liver is one of the most studied organs of the human body owing to its central role in xenobiotic and drug metabolism. In recent decades, extensive research has aimed at developing in vitro liver models able to mimic liver functions to study pathophysiological clues in high-throughput and reproducible environments. Two-dimensional (2D) models have been widely used in screening potential toxic compounds but have failed to accurately reproduce the three-dimensionality (3D) of the liver milieu. To overcome these limitations, improved 3D culture techniques have been developed to recapitulate the hepatic native microenvironment. These models focus on reproducing the liver architecture, representing both parenchymal and nonparenchymal cells, as well as cell interactions. More recently, Liver-on-Chip (LoC) models have been developed with the aim of providing physiological fluid flow and thus achieving essential hepatic functions. Given their unprecedented ability to recapitulate critical features of the liver cellular environments, LoC have been extensively adopted in pathophysiological modelling and currently represent a promising tool for tissue engineering and drug screening applications. In this review, we discuss the evolution of experimental liver models, from the ancient 2D hepatocyte models, widely used for liver toxicity screening, to 3D and LoC culture strategies adopted for mirroring a more physiological microenvironment for the study of liver diseases.
Collapse
Affiliation(s)
- Michela Anna Polidoro
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Erika Ferrari
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Simona Marzorati
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| |
Collapse
|
46
|
Le Daré B, Ferron PJ, Bellamri N, Ribault C, Delpy E, Zal F, Lagente V, Gicquel T. A therapeutic oxygen carrier isolated from Arenicola marina decreases amanitin-induced hepatotoxicity. Toxicon 2021; 200:87-91. [PMID: 34274377 DOI: 10.1016/j.toxicon.2021.07.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/18/2021] [Accepted: 07/10/2021] [Indexed: 11/29/2022]
Abstract
The amanitins (namely α- and β-amanitin) contained in certain mushrooms are bicyclic octapeptides that, when ingested, are responsible for potentially lethal hepatotoxicity. M101 is an extracellular hemoglobin extracted from the marine worm Arenicola marina. It has intrinsic Cu/Zn-SOD-like activity and is currently used as an oxygen carrier in organ preservation solutions. Our present results suggest that M101 might be effective in reducing amanitin-induced hepatotoxicity and may have potential for therapeutic development.
Collapse
Affiliation(s)
- Brendan Le Daré
- Pharmacy, Pontchaillou University Hospital, F-35000, Rennes, France; Univ Rennes, INSERM, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000, Rennes, France.
| | - Pierre-Jean Ferron
- Univ Rennes, INSERM, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000, Rennes, France
| | - Nessrine Bellamri
- Univ Rennes, INSERM, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000, Rennes, France
| | - Catherine Ribault
- Univ Rennes, INSERM, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000, Rennes, France
| | - Eric Delpy
- Hemarina, Aéropôle Centre, F-29600, Morlaix, France
| | - Franck Zal
- Hemarina, Aéropôle Centre, F-29600, Morlaix, France
| | - Vincent Lagente
- Univ Rennes, INSERM, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000, Rennes, France
| | - Thomas Gicquel
- Forensic and Toxicology Laboratory, Pontchaillou University Hospital, F-35000, Rennes, France; Univ Rennes, INSERM, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer), F-35000, Rennes, France
| |
Collapse
|
47
|
Abbott A, Coburn JM. HepaRG Maturation in Silk Fibroin Scaffolds: Toward Developing a 3D In Vitro Liver Model. ACS Biomater Sci Eng 2021. [PMID: 34105934 DOI: 10.1021/acsbiomaterials.0c01584] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In vitro liver models are necessary tools for the development of new therapeutics. HepaRG cells are a commonly used cell line to produce hepatic progenitor cells and hepatocytes. This study demonstrates for the first time the suitability of 3% silk scaffolds to support HepaRG growth and differentiation. The modulus and pore size of 3% silk scaffolds were shown to be within the desired range for liver cell growth. The optimal seeding density for HepaRG cells on silk scaffolds was determined. The growth and maturation of scaffolded HepaRG cells was evaluated for 28 days, where the first 14 days of culture were a proliferation period and the last 14 days of culture were a differentiation period using dimethyl sulfoxide (DMSO) treatment. After the first 14 days of culture, the scaffolded HepaRG cells exhibited increased metabolic activity and albumin secretion compared to monolayer cultured controls and preserved these attributes through the duration of culture. Additionally, after the first 14 days of culture, the scaffolded HepaRG cells displayed a significantly reduced expression of genes associated with hepatocyte maturation. This difference in expression was no longer apparent after 28 days of culture, suggesting that the cells underwent rapid differentiation within the scaffold. The functionalization of silk scaffolds with extracellular matrix (ECM) components (type I collagen and/or an arginylglycylaspartic acid (RGD)-containing peptide) was investigated to determine the impact on HepaRG cell attachment and maturation. The inclusion of ECM components had no noticeable impact on cell attachment but did significantly influence CYP3A4 expression and albumin secretion. Finally, the matrix support provided by the 3% silk scaffolds could prime the HepaRG cells for steatosis liver model applications, as evidenced by lipid droplet accumulation and expression of steatosis-related genes after 24 h of exposure to oleic acid. Overall, our work demonstrates the utility of silk scaffolds in providing a modifiable platform for liver cell growth.
Collapse
Affiliation(s)
- Alycia Abbott
- Department of Biomedical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, Massachusetts 01609, United States
| | - Jeannine M Coburn
- Department of Biomedical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, Massachusetts 01609, United States
| |
Collapse
|
48
|
Bellanti F, di Bello G, Iannelli G, Pannone G, Pedicillo MC, Boulter L, Lu WY, Tamborra R, Villani R, Vendemiale G, Forbes SJ, Serviddio G. Inhibition of nuclear factor (erythroid-derived 2)-like 2 promotes hepatic progenitor cell activation and differentiation. NPJ Regen Med 2021; 6:28. [PMID: 34039998 PMCID: PMC8155039 DOI: 10.1038/s41536-021-00137-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 04/28/2021] [Indexed: 02/08/2023] Open
Abstract
The stem cell ability to self-renew and lead regeneration relies on the balance of complex signals in their microenvironment. The identification of modulators of hepatic progenitor cell (HPC) activation is determinant for liver regeneration and may improve cell transplantation for end-stage liver disease. This investigation used different models to point out the Nuclear factor (erythroid-derived 2)-like 2 (NRF2) as a key regulator of the HPC fate. We initially proved that in vivo models of biliary epithelial cells (BECs)/HPC activation show hepatic oxidative stress, which activates primary BECs/HPCs in vitro. NRF2 downregulation and silencing were associated with morphological, phenotypic, and functional modifications distinctive of differentiated cells. Furthermore, NRF2 activation in the biliary tract repressed the ductular reaction in injured liver. To definitely assess the importance of NRF2 in HPC biology, we applied a xenograft model by inhibiting NRF2 in the human derived HepaRG cell line and transplanting into SCID/beige mice administered with anti-Fas antibody to induce hepatocellular apoptosis; this resulted in effective human hepatocyte repopulation with reduced liver injury. To conclude, NRF2 inhibition leads to the activation and differentiation of liver progenitors. This redox-dependent transcription factor represents a potential target to regulate the commitment of undifferentiated hepatic progenitors into specific lineages.
Collapse
Affiliation(s)
- Francesco Bellanti
- Centre for Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| | - Giorgia di Bello
- Centre for Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Giuseppina Iannelli
- Centre for Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Giuseppe Pannone
- Anatomical Pathology Unit, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Maria Carmela Pedicillo
- Anatomical Pathology Unit, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Luke Boulter
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Wei-Yu Lu
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston Birmingham, UK
| | - Rosanna Tamborra
- Centre for Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Rosanna Villani
- Centre for Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Gianluigi Vendemiale
- Centre for Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Stuart J Forbes
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Gaetano Serviddio
- Centre for Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
49
|
Duivenvoorde LPM, Louisse J, Pinckaers NET, Nguyen T, van der Zande M. Comparison of gene expression and biotransformation activity of HepaRG cells under static and dynamic culture conditions. Sci Rep 2021; 11:10327. [PMID: 33990636 PMCID: PMC8121841 DOI: 10.1038/s41598-021-89710-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 04/27/2021] [Indexed: 11/08/2022] Open
Abstract
Flow conditions have been shown to be important in improving longevity and functionality of primary hepatocytes, but the impact of flow on HepaRG cells is largely unknown. We studied the expression of genes encoding CYP enzymes and transporter proteins and CYP1 and CYP3A4 activity during 8 weeks of culture in HepaRG cells cultured under static conditions (conventional 24-/96-well plate culture with common bicarbonate/CO2 buffering) and under flow conditions in an organ-on-chip (OOC) device. Since the OOC-device is a closed system, bicarbonate/CO2 buffering was not possible, requiring application of another buffering agent, such as HEPES. In order to disentangle the effects of HEPES from the effects of flow, we also applied HEPES-supplemented medium in static cultures and studied gene expression and CYP activity. We found that cells cultured under flow conditions in the OOC-device, as well as cells cultured under static conditions with HEPES-supplemented medium, showed more stable gene expression levels. Furthermore, only cells cultured in the OOC-device showed relatively high baseline CYP1 activity, and their gene expression levels of selected CYPs and transporters were most similar to gene expression levels in human primary hepatocytes. However, there was a decrease in baseline CYP3A4 activity under flow conditions compared to HepaRG cells cultured under static conditions. Altogether, the present study shows that HepaRG cells cultured in the OOC-device were more stable than in static cultures, being a promising in vitro model to study hepatoxicity of chemicals upon chronic exposure.
Collapse
Affiliation(s)
- Loes P M Duivenvoorde
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands.
| | - Jochem Louisse
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| | - Nicole E T Pinckaers
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| | - Tien Nguyen
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| | - Meike van der Zande
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| |
Collapse
|
50
|
In vivo and in vitro α-amanitin metabolism studies using molecular networking. Toxicol Lett 2021; 346:1-6. [PMID: 33872745 DOI: 10.1016/j.toxlet.2021.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/17/2021] [Accepted: 04/13/2021] [Indexed: 01/09/2023]
Abstract
Amanitin poisonings are among the most life-threatening mushroom poisonings, and are mainly caused by the genus Amanita. Hepatotoxicity is the hallmark of amanitins, powerful toxins contained in these mushrooms, and can require liver transplant. Among amatoxins, α-amanitin is the most studied. However, the hypothesis of a possible metabolism of amanitins is still controversial in this pathophysiology. Therefore, there is a need of clarification using cutting-edge tools allowing metabolism study. Molecular network has emerged as powerful tool allowing metabolism study through organization and representation of untargeted tandem mass spectrometry (MS/MS) data in a graphical form. The aim of this study is to investigate amanitin metabolism using molecular networking. In vivo (four positive amanitin urine samples) and in vitro (differentiated HepaRG cells supernatant incubated with α-amanitin 2 μM for 24 h) samples were extracted and analyzed by LC-HRMS/MS using a Q Exactive™ Orbitrap mass spectrometer. Using molecular networking on both in vitro and in vivo, we have demonstrated that α-amanitin does not undergo metabolism in human. Thus, we provide solid evidence that a possible production of amanitin metabolites cannot be involved in its toxicity pathways. These findings can help to settle the debate on amanitin metabolism and toxicity.
Collapse
|