1
|
Jin HJ, Qiu ZD, Qian C, Zhang CY, Peng Y. Identification of the metabolomic alterations associated with the formation of bisphenol-A sulfate metabolite in HepG2 cells. Food Chem Toxicol 2025; 200:115382. [PMID: 40058625 DOI: 10.1016/j.fct.2025.115382] [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: 11/20/2024] [Revised: 02/05/2025] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
The elucidation of the causal relationship between bisphenol-A (BPA) exposure and hepatoxic outcomes is challenging because of the complexity in both the BPA-derived metabolites formed in the liver and the associated endogenous molecular responses. We performed parallel metabolism experiments with BPA to characterize the BPA sulfate formation and the associated alterations in the metabolome level in HepG2 cells using mass spectrometry-based metabolome wide association study. Briefly, HepG2 cells were exposed for 8 or 24 h to 1 or 10 μM BPA in DMSO or DMSO alone. The levels of BPA sulfate in the cell culture media were quantified, and the sulfation efficiency was about 0.4 % observed for both 1 and 10 μM BPA in HepG2 cells. Targeted metabolomic analyses revealed alterations belonging to forty metabolic pathways following BPA exposure. Featured by the decreasing of estrone sulfate, estrogen metabolism was observed as the top 1 enriched pathway in response to BPA exposure. MWAS suggests that BPA sulfate formation in HepG2 cells resulted in vitamin B6 deficiency and dysregulated vitamin B6-dependent processes, for example, the kynurenine pathway in tryptophan metabolism. These findings collectively provide insights into the linkage between exogenous and endogenous metabolism and the potential initial events in BPA exposure-relevant hepatoxicity.
Collapse
Affiliation(s)
- Hui-Jun Jin
- Hubei Key Laboratory of Regional Development and Environmental Response, Faculty of Resources and Environmental Science, Hubei University, Wuhan, 430062, PR China
| | - Zi-Dong Qiu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Centre for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Cen Qian
- Hubei Key Laboratory of Regional Development and Environmental Response, Faculty of Resources and Environmental Science, Hubei University, Wuhan, 430062, PR China
| | - Chun-Yun Zhang
- Hubei Key Laboratory of Regional Development and Environmental Response, Faculty of Resources and Environmental Science, Hubei University, Wuhan, 430062, PR China.
| | - Yu Peng
- Hubei Key Laboratory of Regional Development and Environmental Response, Faculty of Resources and Environmental Science, Hubei University, Wuhan, 430062, PR China.
| |
Collapse
|
2
|
Watanabe M, Salvadori A, Markovic M, Sudo R, Ovsianikov A. Advanced liver-on-chip model mimicking hepatic lobule with continuous microvascular network via high-definition laser patterning. Mater Today Bio 2025; 32:101643. [PMID: 40206147 PMCID: PMC11979415 DOI: 10.1016/j.mtbio.2025.101643] [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: 07/31/2024] [Revised: 11/09/2024] [Accepted: 03/06/2025] [Indexed: 04/11/2025] Open
Abstract
There is a great demand for development of advanced in vitro liver models to predict the efficacy and safety of drug candidates accurately in the preclinical drug development. Despite the great efforts to develop biomimetic models, it remains challenging to precisely mimic a functional unit of the liver (i.e., hepatic lobule) with a continuous microvascular network. Recent progress in laser patterning has allowed us to create arbitrary biomimetic structures with high resolution. Here, we propose an advanced liver-on-chip model mimicking the hepatic lobule with a continuous microvascular network, ranging from the microvessels to the central vein of the liver, utilizing femtosecond laser patterning. Firstly, we optimize the laser power to pattern microchannels mimicking the microvessel and central vein of the hepatic lobule by using a femtosecond laser within a collagen-based hydrogel containing hepatic cells. Secondly, we construct continuous microvessels with luminal structures by comparing different microchannel sizes in diameter. Finally, we assemble a millimeter-scale hepatic lobule-like structure with multiple layers of microvascular networks in the liver-on-chip. Furthermore, our liver-on-chip model exhibits major liver functions and drug-induced hepatotoxicity, as evidenced by albumin and urea productions and by a toxic response to acetaminophen, respectively. Our approach provides valuable strategies for the development of advanced physiological and pathological liver-on-chip models for pharmaceutical and toxicological studies.
Collapse
Affiliation(s)
- Masafumi Watanabe
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), 1060 Vienna, Austria
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Austria
- Japan Society for the Promotion of Science (JSPS) Overseas Research Fellow, Japan
| | - Alice Salvadori
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), 1060 Vienna, Austria
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Austria
| | - Marica Markovic
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), 1060 Vienna, Austria
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Austria
| | - Ryo Sudo
- Department of System Design Engineering, Keio University, 223-8522 Yokohama, Japan
| | - Aleksandr Ovsianikov
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), 1060 Vienna, Austria
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Austria
| |
Collapse
|
3
|
Carlsson MJ, Herzog N, Felske C, Ackermann G, Regier A, Wittmann S, Fernández Cereijo R, Sturla SJ, Küpper JH, Fahrer J. The DNA Repair Protein MGMT Protects against the Genotoxicity of N-Nitrosodimethylamine, but Not N-Nitrosodiethanolamine and N-Nitrosomethylaniline, in Human HepG2 Liver Cells with CYP2E1 Expression. Chem Res Toxicol 2025. [PMID: 40390554 DOI: 10.1021/acs.chemrestox.5c00133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
N-nitrosamines are genotoxic contaminants that occur in the diet, consumer products, and the environment. More recently, N-nitrosamines were also detected as drug impurities. After uptake, N-nitrosamines undergo metabolic activation by cytochrome P450 monooxygenases (CYPs), resulting in DNA damage and tumor formation. In this study, the genotoxicity and cytotoxicity of three N-nitrosamines with structurally distinct substituents, N-nitrosodimethylamine (NDMA), N-nitrosodiethanolamine (NDELA) and N-nitrosomethylaniline (NMA), were analyzed in human HepG2 liver cell models proficient or deficient in CYP2E1 biotransformation. Furthermore, the impact of the DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT) was investigated. The novel genetically engineered HepG2-CYP2E1 cell line strongly expressed CYP2E1, which was not detectable in wildtype (WT) HepG2 cells. We then confirmed that the CYP2E1 substrate NDMA caused O6-methyldesoxyguanosine adducts and DNA strand breaks in a CYP2E1-dependent manner, leading to cytotoxicity. By the same approach, we demonstrated that NDELA induced DNA strand breaks in HepG2-CYP2E1 cells, whereas no effect was observed for NMA. However, NMA was revealed to cause DNA cross-links. Furthermore, both NDELA and NMA were cytotoxic in HepG2-CYP2E1 cells, but not in WT cells. Subsequently, the pharmacological MGMT inhibitor O6-benzylguanine was used to deplete MGMT in both HepG2 cell models. MGMT inhibition clearly increased DNA strand break levels due to NDMA exposure, whereas DNA strand break formation by NDELA and NMA were not affected by inhibiting MGMT. In line with these findings, the clastogenic effects of NDMA were potentiated in the absence of MGMT. In contrast to that, NDELA- and NMA-induced clastogenicity was not influenced by MGMT inhibition. Taken together, our study revealed that all three structurally diverse N-nitrosamines are cytotoxic and clastogenic in a CYP2E1-dependent manner, while only NDMA and NDELA caused DNA strand breaks. Furthermore, we demonstrated for the first time that DNA repair by MGMT does not confer protection against NDELA and NMA-triggered DNA strand break induction and clastogenicity.
Collapse
Affiliation(s)
- Max J Carlsson
- Division of Food Chemistry and Toxicology, Department of Chemistry, RPTU Kaiserslautern-Landau, 67663 Kaiserslautern, Germany
| | - Natalie Herzog
- Division of Molecular Cell Biology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Christina Felske
- Division of Food Chemistry and Toxicology, Department of Chemistry, RPTU Kaiserslautern-Landau, 67663 Kaiserslautern, Germany
| | - Gabriel Ackermann
- Division of Food Chemistry and Toxicology, Department of Chemistry, RPTU Kaiserslautern-Landau, 67663 Kaiserslautern, Germany
| | - Alexander Regier
- Division of Food Chemistry and Toxicology, Department of Chemistry, RPTU Kaiserslautern-Landau, 67663 Kaiserslautern, Germany
| | - Simon Wittmann
- Division of Food Chemistry and Toxicology, Department of Chemistry, RPTU Kaiserslautern-Landau, 67663 Kaiserslautern, Germany
| | | | - Shana J Sturla
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland
| | - Jan-Heiner Küpper
- Division of Molecular Cell Biology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Jörg Fahrer
- Division of Food Chemistry and Toxicology, Department of Chemistry, RPTU Kaiserslautern-Landau, 67663 Kaiserslautern, Germany
| |
Collapse
|
4
|
Zhou Y, Zhong Y, Lauschke VM. Evaluating the synergistic use of advanced liver models and AI for the prediction of drug-induced liver injury. Expert Opin Drug Metab Toxicol 2025; 21:563-577. [PMID: 39893552 DOI: 10.1080/17425255.2025.2461484] [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: 10/28/2024] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
INTRODUCTION Drug-induced liver injury (DILI) is a leading cause of acute liver failure. Hepatotoxicity typically occurs only in a subset of individuals after prolonged exposure and constitutes a major risk factor for the termination of drug development projects. AREAS COVERED We provide an overview of available human liver models for DILI research and discuss how they have been used to aid in early risk assessments and to mitigate the risk of project closures due to DILI in clinical stages. We summarize the different data that can be provided by such models and illustrate how these diverse data types can be interfaced with machine learning strategies to improve predictions of liver safety liabilities. EXPERT OPINION Advanced human liver models closely mimic human liver phenotypes and functions for many weeks, allowing for the recapitulation of hepatotoxicity events in vitro. Integration of the biochemical, histological, and toxicogenomic output data from these models with physicochemical compound properties using different machine learning architectures holds promise to enhance preclinical DILI predictions. However, to realize this aim, it is important to benchmark the available liver models on test sets of DILI positive and negative compounds and to carefully annotate and share the resulting data.
Collapse
Affiliation(s)
- Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Yi Zhong
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
5
|
Nosaka T, Naito T, Akazawa Y, Takahashi K, Matsuda H, Ohtani M, Nishizawa T, Okamoto H, Nakamoto Y. Identification of novel antiviral host factors by functional gene expression analysis using in vitro HBV infection assay systems. PLoS One 2025; 20:e0314581. [PMID: 40048440 PMCID: PMC11884705 DOI: 10.1371/journal.pone.0314581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/12/2024] [Indexed: 03/09/2025] Open
Abstract
To cure hepatitis B virus (HBV) infection, it is essential to elucidate the function of hepatocyte host factors in regulating the viral life cycle. Signaling and transcription activator of transcription (STAT)1 play important roles in immune responses, but STAT1-independent pathways have also been shown to have important biological reactivity. Using an in vitro HBV infection assay system, the current study aimed to investigate the STAT1-independent host factors that contribute to the control of viral infection by comprehensive functional screening. The in vitro HBV infection system was established using primary human hepatocytes (PXB cells) infected with HBV derived from a plasmid containing the 1.3-mer HBV genome. Comprehensive functional studies were performed using small interfering RNA (siRNA) and vector transfection and analyzed using microarrays. Knockdown of STAT1 increased viral products in HBV-transfected HepG2 cells, but decreased in HBV-infected PXB cells. RNA microarray was performed using HBV-infected PXB cells with STAT1 knockdown. Fumarylacetoacetate hydrolase (FAH) was extracted by siRNA of genes in PXB cells altered by STAT1 knockdown. Transfection of FAH inhibited HBV replication. Dimethyl fumarate (DMF), the methyl ester of FAH metabolite, showed antiviral effects by inducing autophagy and anti-HBV-related genes. Independently of STAT1, FAH was identified as a host factor that contributes to the control of viral infection, and its metabolite, DMF, exhibited antiviral activity. These results suggest that the novel host factor FAH and its metabolites may be an innovative therapeutic strategy to control the HBV life cycle.
Collapse
Affiliation(s)
- Takuto Nosaka
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Tatsushi Naito
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Yu Akazawa
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Kazuto Takahashi
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Hidetaka Matsuda
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Masahiro Ohtani
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Tsutomu Nishizawa
- Division of Virology, Department of Infection and Immunity, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Hiroaki Okamoto
- Division of Virology, Department of Infection and Immunity, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Yasunari Nakamoto
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| |
Collapse
|
6
|
Rentschler S, Doss S, Kaiser L, Weinschrott H, Kohl M, Deigner HP, Sauer M. Metabolic Biomarkers of Liver Failure in Cell Models and Patient Sera: Toward Liver Damage Evaluation In Vitro. Int J Mol Sci 2024; 25:13739. [PMID: 39769500 PMCID: PMC11677895 DOI: 10.3390/ijms252413739] [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: 10/31/2024] [Revised: 11/29/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Recent research has concentrated on the development of suitable in vitro cell models for the early identification of hepatotoxicity during drug development in order to reduce the number of animal models and to obtain a better predictability for hepatotoxic reactions in humans. The aim of the presented study was to identify translational biomarkers for acute liver injury in human patients that can serve as biomarkers for hepatocellular injury in vivo and in vitro in simple cell models. Therefore, 188 different metabolites from patients with acute-on-chronic liver failure before and after liver transplantation were analyzed with mass spectrometry. The identified potential metabolic biomarker set, including acylcarnitines, phosphatidylcholines and sphingomyelins, was used to screen primary and permanent hepatocyte culture models for their ability to model hepatotoxic responses caused by different drugs with known and unknown hepatotoxic potential. The results obtained suggest that simple in vitro cell models have the capability to display metabolic responses in biomarkers for liver cell damage in course of the treatment with different drugs and therefore can serve as a basis for in vitro models for metabolic analysis in drug toxicity testing. The identified metabolites should further be evaluated for their potential to serve as a metabolic biomarker set indicating hepatocellular injury in vitro as well as in vivo.
Collapse
Affiliation(s)
- Simone Rentschler
- Institute of Precision Medicine, Furtwangen University, Jakob-Kienzle-Straße 17, 78054 VS-Schwenningen, Germany
| | - Sandra Doss
- Fraunhofer Institute IZI (Leipzig), Department Rostock, Schillingallee 68, 18057 Rostock, Germany
| | - Lars Kaiser
- Institute of Precision Medicine, Furtwangen University, Jakob-Kienzle-Straße 17, 78054 VS-Schwenningen, Germany
| | - Helga Weinschrott
- Institute of Precision Medicine, Furtwangen University, Jakob-Kienzle-Straße 17, 78054 VS-Schwenningen, Germany
| | - Matthias Kohl
- Institute of Precision Medicine, Furtwangen University, Jakob-Kienzle-Straße 17, 78054 VS-Schwenningen, Germany
| | - Hans-Peter Deigner
- Institute of Precision Medicine, Furtwangen University, Jakob-Kienzle-Straße 17, 78054 VS-Schwenningen, Germany
- Faculty of Science, Tuebingen University, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Rostock, Schillingallee 35, 18057 Rostock, Germany
| | - Martin Sauer
- Fraunhofer Institute IZI (Leipzig), Department Rostock, Schillingallee 68, 18057 Rostock, Germany
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Rostock, Schillingallee 35, 18057 Rostock, Germany
- Center for Anesthesiology and Intensive Care Medicine, Hospital of Magdeburg, Birkenallee 34, 39130 Magdeburg, Germany
| |
Collapse
|
7
|
Singla B. Fanlian Huazhuo Formula: A promising herbal preparation for metabolic liver disease. World J Gastroenterol 2024; 30:4964-4968. [PMID: 39679304 PMCID: PMC11612710 DOI: 10.3748/wjg.v30.i46.4964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/17/2024] [Accepted: 11/05/2024] [Indexed: 11/21/2024] Open
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) has increased significantly in recent decades and is projected to increase further due to the rising obesity rates. MASLD patients are at higher risk of developing advanced liver diseases "cirrhosis and hepatocellular carcinoma" as well as liver- or cardiovascular-related mortality. Existing lipid-lowering therapies failed to reduce the risk of mortality in these patients. Therefore, there is an urgent need for pharmacotherapies that can control and even reverse this disease. Fanlian Huazhuo Formula (FLHZF) is a combination herbal preparation, and its various individual constituents regulate hepatic lipid metabolism, adipose tissue inflammation, and gut microbiota. Despite, these useful effects, limited information is available on its benefits in diet-induced hepatosteatosis. In this article, we discuss the research findings recently published about the therapeutic effects of FLHZF in suppressing MASLD development and underlying mechanisms. Utilizing a series of in vitro and in vivo experiments, the authors demonstrated for the first time that FLHZF suppresses MASLD in male mice possibly by inhibiting hepatic de novo lipogenesis pathways and reducing hepatocyte death. This study paves the way for future investigations aimed at investigating FLHZF's role in inhibiting lipogenesis particularly using radioactively-labeled glucose and acetate, and governing hepatocyte mitochondrial function, gut microbiome profile, and its effects in other models of MASLD, and female mice.
Collapse
Affiliation(s)
- Bhupesh Singla
- Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38103, United States
| |
Collapse
|
8
|
Xin Y, Ligorio C, O'brien M, Collins R, Dong S, Miller AF, Saiani A, Gough JE. Effect of supramolecular peptide hydrogel scaffold charge on HepG2 viability and spheroid formation. J Mater Chem B 2024; 12:12553-12566. [PMID: 39502032 DOI: 10.1039/d4tb01701c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
Supramolecular bioinspired self-assembling peptide hydrogel (SAPH) scaffolds represent a class of fully defined synthetic materials whose chemical and mechanical properties can be finely engineered. In this study, the relationship between SAPHs physicochemical properties and HepG2 cells viability, spheroid formation and function are discussed. We first report that negatively charged SAPHs promote hepatocyte proliferation and spheroids formation in vitro 3D culture while positively charged SAPHs lead to hepatocyte death irrespective of the hydrogel mechanical properties. More specifically HepG2 cultured in 3D in E(FKFE)2 negatively charged SAPH maintained a differentiated phenotype and assembled into well-defined spheroids with strong cell-cell interactions. Furthermore, HepG2 spheroids responded to acetaminophen exposure with upregulation of key CYP450 enzymes expression clearly showing their potential for drug toxicity testing. These findings demonstrate how fine-tuned functional SAPH scaffolds can be used to identify key scaffolds parameters affecting cells. In this case we demonstrated the potential of negatively charged SAPHs for the 3D culture of HepG2 with potential applications in drug screening.
Collapse
Affiliation(s)
- Yu Xin
- Department of Materials & Henry Royce Institute, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, UK.
| | - Cosimo Ligorio
- Department of Materials & Manchester Institute of Biotechnology, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, UK
| | - Marie O'brien
- Department of Materials & Henry Royce Institute, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, UK.
| | - Richard Collins
- Electron Microscopy Core Facility, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| | - Siyuan Dong
- Department of Chemical Engineering & Manchester Institute of Biotechnology, School of Engineering, Faculty of Science and Engineering, The University of Manchester, UK
| | - Aline F Miller
- Department of Chemical Engineering & Manchester Institute of Biotechnology, School of Engineering, Faculty of Science and Engineering, The University of Manchester, UK
| | - Alberto Saiani
- Division of Pharmacy and Optometry & Manchester Institute of Biotechnology, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| | - Julie E Gough
- Department of Materials & Henry Royce Institute, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, UK.
| |
Collapse
|
9
|
Jiang X, Zhu Y, Dong S, Lin R, Zhu P, Mao J, Cao Y, Yin X, Dong F, He K, Wang N. Combination of biotransformation and metabolomics reveals tolfenpyrad-induced hepatocytotoxicity. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175320. [PMID: 39111429 DOI: 10.1016/j.scitotenv.2024.175320] [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: 04/15/2024] [Revised: 06/10/2024] [Accepted: 08/04/2024] [Indexed: 08/18/2024]
Abstract
Tolfenpyrad (TFP) is an extensively used pesticide that inevitably leads to human exposure to both TFP and its transformation product residues. However, the biotransformation of TFP in humans has not been elucidated, and the toxicity of TFP along with its biotransformation products remains largely unknown. In this study, the biotransformation process of TFP was investigated using human liver microsomes and human hepatic cells. Endogenous metabolic changes in the cells were studied to investigate the hepatocytotoxicity of TFP at environmentally relevant concentrations. Fourteen phase I biotransformation products and four phase II TFP products were characterized, among which twelve products were identified for the first time. The oxidative product tolfenpyrad-benzoic acid (PT-CA) was particularly abundant and stable. Further hepatotoxicity assessments and metabolic studies demonstrated comparable metabolic profiles for TFP and PT-CA in HepG2 cells, with both significantly disrupting purine and glutathione metabolism. These processes are closely associated with oxidative stress, mitochondrial damage, and cell death. Our results provide novel perspectives on the biotransformation, metabolism, and hepatotoxicity of TFP, thereby highlighting the non-negligible toxicity of its crucial biotransformation product PT-CA in environmental risk assessments.
Collapse
Affiliation(s)
- Xin Jiang
- National Center of Biomedical Analysis, Beijing 100039, China
| | - Yingjie Zhu
- National Center of Biomedical Analysis, Beijing 100039, China
| | - Suhe Dong
- National Center of Biomedical Analysis, Beijing 100039, China
| | - Runfeng Lin
- National Center of Biomedical Analysis, Beijing 100039, China
| | - Peihong Zhu
- National Center of Biomedical Analysis, Beijing 100039, China
| | - Jie Mao
- National Center of Biomedical Analysis, Beijing 100039, China
| | - Yanqing Cao
- National Center of Biomedical Analysis, Beijing 100039, China
| | - Xiaoyao Yin
- National Center of Biomedical Analysis, Beijing 100039, China
| | - Fangting Dong
- National Center of Biomedical Analysis, Beijing 100039, China
| | - Kun He
- National Center of Biomedical Analysis, Beijing 100039, China.
| | - Na Wang
- National Center of Biomedical Analysis, Beijing 100039, China.
| |
Collapse
|
10
|
Yang NV, Chao JY, Garton KA, Tran T, King SM, Orr J, Oei JH, Crawford A, Kang M, Zalpuri R, Jorgens DM, Konchadi P, Chorba JS, Theusch E, Krauss RM. TOMM40 regulates hepatocellular and plasma lipid metabolism via an LXR-dependent pathway. Mol Metab 2024; 90:102056. [PMID: 39489289 PMCID: PMC11600064 DOI: 10.1016/j.molmet.2024.102056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
OBJECTIVE The gene encoding TOMM40 (Transporter of Outer Mitochondrial Membrane 40) is adjacent to that encoding APOE, which has a central role in lipid and lipoprotein metabolism. While human genetic variants near APOE and TOMM40 have been shown to be strongly associated with plasma lipid levels, a specific role for TOMM40 in lipid metabolism has not been established, and the present study was aimed at assessing this possibility. METHODS TOMM40 was knocked down by siRNA in human hepatoma HepG2 cells, and effects on mitochondrial function, lipid phenotypes, and crosstalk between mitochondria, ER, and lipid droplets were examined. Additionally, hepatic and plasma lipid levels were measured in mice following shRNA-induced knockdown of Tomm40 shRNA. RESULTS In HepG2 cells, TOMM40 knockdown upregulated expression of APOE and LDLR in part via activation of LXRB (NR1H2) by oxysterols, with consequent increased uptake of VLDL and LDL. This is in part due to disruption of mitochondria-endoplasmic reticulum contact sites, with resulting accrual of reactive oxygen species and non-enzymatically derived oxysterols. With TOMM40 knockdown, cellular triglyceride and lipid droplet content were increased, effects attributable in part to receptor-mediated VLDL uptake, since lipid staining was significantly reduced by concomitant suppression of either LDLR or APOE. In contrast, cellular cholesterol content was reduced due to LXRB-mediated upregulation of the ABCA1 transporter as well as increased production and secretion of oxysterol-derived cholic acid. Consistent with the findings in hepatoma cells, in vivo knockdown of TOMM40 in mice resulted in significant reductions of plasma triglyceride and cholesterol concentrations, reduced hepatic cholesterol and increased triglyceride content, and accumulation of lipid droplets leading to development of steatosis. CONCLUSIONS These findings demonstrate a role for TOMM40 in regulating hepatic lipid and plasma lipoprotein levels and identify mechanisms linking mitochondrial function with lipid metabolism.
Collapse
Affiliation(s)
- Neil V Yang
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, CA, USA; Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Justin Y Chao
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Kelly A Garton
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, CA, USA
| | - Tommy Tran
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Sarah M King
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Joseph Orr
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Jacob H Oei
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Alexandra Crawford
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Misun Kang
- Electron Microscope Laboratory, University of California, Berkeley, CA, USA
| | - Reena Zalpuri
- Electron Microscope Laboratory, University of California, Berkeley, CA, USA
| | - Danielle M Jorgens
- Electron Microscope Laboratory, University of California, Berkeley, CA, USA
| | - Pranav Konchadi
- Department of Medicine, University of California, San Francisco, CA, USA
| | - John S Chorba
- Department of Medicine, University of California, San Francisco, CA, USA; Division of Cardiology, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - Elizabeth Theusch
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Ronald M Krauss
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, CA, USA; Department of Pediatrics, University of California, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, CA, USA.
| |
Collapse
|
11
|
Kim C, Zhu Z, Barbazuk WB, Bacher RL, Vulpe CD. Time-course characterization of whole-transcriptome dynamics of HepG2/C3A spheroids and its toxicological implications. Toxicol Lett 2024; 401:125-138. [PMID: 39368564 DOI: 10.1016/j.toxlet.2024.10.004] [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: 06/14/2024] [Revised: 09/10/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
Physiologically relevant in vitro models are a priority in predictive toxicology to replace and/or reduce animal experiments. The compromised toxicant metabolism of many immortalized human liver cell lines grown as monolayers as compared to in vivo metabolism limits their physiological relevance. However, recent efforts to culture liver cells in a 3D environment, such as spheroids, to better mimic the in vivo conditions, may enhance the toxicant metabolism of human liver cell lines. In this study, we characterized the dynamic changes in the transcriptome of HepG2/C3A hepatocarcinoma cell spheroids maintained in a clinostat system (CelVivo) to gain insight into the metabolic capacity of this model as a function of spheroid size and culture time. We assessed morphological changes (size, necrotic core), cell health, and proliferation rate from initial spheroid seeding to 35 days of continuous culture in conjunction with a time-course (0, 3, 7, 10, 14, 21, 28 days) of the transcriptome (TempO-Seq, BioSpyder). The phenotypic characteristics of HepG2/C3A growing in spheroids were comparable to monolayer growth until ∼Day 12 (Day 10-14) when a significant decrease in cell doubling rate was noted which was concurrent with down-regulation of cell proliferation and cell cycle pathways over this time period. Principal component analysis of the transcriptome data suggests that the Day 3, 7, and 10 spheroids are pronouncedly different from the Day 14, 21, and 28 spheroids in support of a biological transition time point during the long-term 3D spheroid cultures. The expression of genes encoding cellular components involved in toxicant metabolism and transport rapidly increased during the early time points of spheroids to peak at Day 7 or Day 10 as compared to monolayer cultures with a gradual decrease in expression with further culture, suggesting the most metabolically responsive time window for exposure studies. Overall, we provide baseline information on the cellular and molecular characterization, with a particular focus on toxicant metabolic capacity dynamics and cell growth, of HepG2/C3A 3D spheroid cultures over time.
Collapse
Affiliation(s)
- Chanhee Kim
- Center for Human and Environmental Toxicology, Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Zhaohan Zhu
- Department of Biostatistics, University of Florida, Gainesville, FL, United States
| | - W Brad Barbazuk
- Department of Biology, University of Florida, Gainesville, FL, United States; University of Florida Genetics Institute, University of Florida, Gainesville, FL, United States
| | - Rhonda L Bacher
- Department of Biostatistics, University of Florida, Gainesville, FL, United States
| | - Christopher D Vulpe
- Center for Human and Environmental Toxicology, Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
12
|
Manuguerra S, Carli F, Scoditti E, Santulli A, Gastaldelli A, Messina CM. Effects of Mixtures of Emerging Pollutants and Drugs on Modulation of Biomarkers Related to Toxicity, Oxidative Stress, and Cancer. Metabolites 2024; 14:559. [PMID: 39452940 PMCID: PMC11509268 DOI: 10.3390/metabo14100559] [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: 08/13/2024] [Revised: 10/03/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Background/Objectives: Over time, the scientific community has developed a growing interest in the effects of mixtures of different compounds, for which there is currently no established evidence or knowledge, in relation to certain categories of xenobiotics. It is well known that exposure to pollutants causes oxidative stress, resulting in the overproduction of reactive oxygen species (ROS), which can affect signaling pathways that regulate the cell cycle, apoptosis, energy balance, and cellular metabolism. The aim of this study was to investigate the effects of sub-lethal concentrations of mixtures of emerging pollutants and pharmaceuticals on the modulation of biomarkers related to toxicity, oxidative stress, and cancer. Methods: In this study, the hepatoma cell line HepG2 was exposed to increasing concentrations of polybrominated diphenyl ether 47 (BDE-47), cadmium chloride (CdCl2), and carbamazepine (CBZ), both individually and in mixtures, for 72 h to assess cytotoxicity using the MTT assay. The subsequent step, following the identification of the sub-lethal concentration, was to investigate the effects of exposure at the gene expression level, through the evaluation of molecular markers related to cell cycle and apoptosis (p53), oxidative stress (NRF2), conjugation and detoxification of xenobiotics (CYP2C9 and GST), DNA damage (RAD51 and γH2AFX), and SUMOylation processes (SUMO1 and UBC9) in order to identify any potential alterations in pathways that are normally activated at the cellular level. Results: The results showed that contaminants tend to affect the enzymatic detoxification and antioxidant system, influencing DNA repair defense mechanisms involved in resistance to oxidative stress. The combined effect of the compounds at sub-lethal doses results in a greater activation of these pathways compared to exposure to each compound alone, thereby exacerbating their cytotoxicity. Conclusions: The biomarkers analyzed could contribute to the definition of early warning markers useful for environmental monitoring, while simultaneously providing insight into the toxicity and hazard levels of these substances in the environment and associated health risks.
Collapse
Affiliation(s)
- Simona Manuguerra
- Laboratory of Marine Biochemistry and Ecotoxicology, Department of Earth and Marine Sciences DiSTeM, University of Palermo, Via G. Barlotta 4, 91100 Trapani, Italy; (S.M.); (A.S.)
| | - Fabrizia Carli
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy;
| | - Egeria Scoditti
- Institute of Clinical Physiology, National Research Council, 73100 Lecce, Italy;
| | - Andrea Santulli
- Laboratory of Marine Biochemistry and Ecotoxicology, Department of Earth and Marine Sciences DiSTeM, University of Palermo, Via G. Barlotta 4, 91100 Trapani, Italy; (S.M.); (A.S.)
| | - Amalia Gastaldelli
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy;
| | - Concetta Maria Messina
- Laboratory of Marine Biochemistry and Ecotoxicology, Department of Earth and Marine Sciences DiSTeM, University of Palermo, Via G. Barlotta 4, 91100 Trapani, Italy; (S.M.); (A.S.)
| |
Collapse
|
13
|
Zwickenpflug W, Hornung F, Hollaus A, Oswald MS, Chioato Z, Gudermann T, Högg C. Biosynthesis of vitamin B 3 and NAD +: incubating HepG2 cells with the alkaloid myosmine. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:6844-6854. [PMID: 38578648 DOI: 10.1002/jsfa.13513] [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: 10/12/2023] [Revised: 03/26/2024] [Accepted: 04/03/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND In the kynurenine pathway, it is reported that the essential amino acid tryptophan forms nicotinic acid (NA, vitamin B3) in biological systems. This pathway is part of the de novo pathway to perform nicotinamide adenine dinucleotide (NAD+) biosynthesis. Additionally, biosynthesis of NAD+ via the Preiss-Handler pathway involves NA and its analogue nicotinamide, both designated as niacin. Previous attempts were successful in converting myosmine (MYO) by organic synthesis to NA, and the assumption was that the alkaloid MYO, which is taken in from food, can be converted into NA by biological oxidation. RESULT Incubation of HepG2 cells with MYO yielded NA. Moreover, a significant increase of NAD+ compared with the control has been found. CONCLUSION Hence, MYO could be assumed to be the hitherto unknown origin of an alternative NA biosynthesis additionally influencing NAD+ biosynthesis positively. This novel MYO pathway may open new perspectives to improve knowledge and relevance of NA and NAD+ biosynthesis and bioactivation in cells and, moreover, in food staples, food, and diet. © 2024 The Authors. Journal of The Science of Food and Agriculture published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
Affiliation(s)
- Wolfgang Zwickenpflug
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Florian Hornung
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Alexandra Hollaus
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Michaela S Oswald
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Zoé Chioato
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Christof Högg
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
- Department of Conservative Dentistry and Periodontology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
14
|
Valente LC, Bacil GP, Riechelmann-Casarin L, Barbosa GC, Barbisan LF, Romualdo GR. Exploring in vitro modeling in hepatocarcinogenesis research: morphological and molecular features and similarities to the corresponding human disease. Life Sci 2024; 351:122781. [PMID: 38848937 DOI: 10.1016/j.lfs.2024.122781] [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: 01/06/2024] [Revised: 04/04/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
The hepatocellular carcinoma (HCC) features a remarkable epidemiological burden, ranking as the third most lethal cancer worldwide. As the HCC-related molecular and cellular complexity unfolds as the disease progresses, the use of a myriad of in vitro models available is mandatory in translational preclinical research setups. In this review paper, we will compile cutting-edge information on the in vitro bioassays for HCC research, (A) emphasizing their morphological and molecular parallels with human HCC; (B) delineating the advantages and limitations of their application; and (C) offering perspectives on their prospective applications. While bidimensional (2D) (co) culture setups provide a rapid low-cost strategy for metabolism and drug screening investigations, tridimensional (3D) (co) culture bioassays - including patient-derived protocols as organoids and precision cut slices - surpass some of the 2D strategies limitations, mimicking the complex microarchitecture and cellular and non-cellular microenvironment observed in human HCC. 3D models have become invaluable tools to unveil HCC pathophysiology and targeted therapy. In both setups, the recapitulation of HCC in different etiologies/backgrounds (i.e., viral, fibrosis, and fatty liver) may be considered as a fundamental guide for obtaining translational findings. Therefore, a "multimodel" approach - encompassing the advantages of different in vitro bioassays - is encouraged to circumvent "model-biased" outcomes in preclinical HCC research.
Collapse
Affiliation(s)
- Leticia Cardoso Valente
- São Paulo State University (UNESP), Medical School, Botucatu, Experimental Research Unit (UNIPEX), Brazil
| | - Gabriel Prata Bacil
- São Paulo State University (UNESP), Institute of Biosciences, Botucatu, Department of Structural and Functional Biology, Brazil
| | - Luana Riechelmann-Casarin
- São Paulo State University (UNESP), Medical School, Botucatu, Experimental Research Unit (UNIPEX), Brazil
| | | | - Luís Fernando Barbisan
- São Paulo State University (UNESP), Institute of Biosciences, Botucatu, Department of Structural and Functional Biology, Brazil
| | - Guilherme Ribeiro Romualdo
- São Paulo State University (UNESP), Medical School, Botucatu, Experimental Research Unit (UNIPEX), Brazil.
| |
Collapse
|
15
|
Carvalho LCF, Ferreira FM, Dias BV, Azevedo DCD, de Souza GHB, Milagre MM, de Lana M, Vieira PMDA, Carneiro CM, Paula-Gomes SD, Cangussu SD, Costa DC. Silymarin inhibits the lipogenic pathway and reduces worsening of non-alcoholic fatty liver disease (NAFLD) in mice. Arch Physiol Biochem 2024; 130:460-474. [PMID: 36328030 DOI: 10.1080/13813455.2022.2138445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/17/2022] [Accepted: 09/08/2022] [Indexed: 11/06/2022]
Abstract
CONTEXT The role of silymarin in hepatic lipid dysfunction and its possible mechanisms of action were investigated. OBJECTIVE To evaluate the effects of silymarin on hepatic and metabolic profiles in mice fed with 30% fructose for 8 weeks. METHODS We evaluated the antioxidant profile of silymarin; mice consumed 30% fructose and were treated with silymarin (120 mg/kg/day or 240 mg/kg/day). We performed biochemical, redox status, and histopathological assays. RT-qPCR was performed to detect ACC-1, ACC-2, FAS, and CS expression, and western blotting to detect PGC-1α levels. RESULTS Silymarin contains high levels of phenolic compounds and flavonoids and exhibited significant antioxidant capacity in vitro. In vivo, the fructose-fed groups showed increased levels of AST, ALT, SOD/CAT, TBARS, hepatic TG, and cholesterol, as well as hypertriglyceridaemia, hypercholesterolaemia, and increased ACC-1 and FAS. Silymarin treatment reduced these parameters and increased mRNA levels and activity of hepatic citrate synthase. CONCLUSIONS These results suggest that silymarin reduces worsening of NAFLD.
Collapse
Affiliation(s)
| | | | - Bruna Vidal Dias
- Laboratório de Bioquímica Metabólica, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | | | | | - Matheus Marque Milagre
- Laboratório Doença de Chagas, Universidade Federal de Ouro Preto, UFOP, Ouro Preto, Brazil
| | - Marta de Lana
- Laboratório Doença de Chagas, Universidade Federal de Ouro Preto, UFOP, Ouro Preto, Brazil
| | | | | | - Sílvia de Paula-Gomes
- Laboratório de Bioquímica e Biologia Molecular, Universidade Federal de Ouro Preto, UFOP, Ouro Preto, Brazil
| | - Silvia Dantas Cangussu
- Laboratório de Fisiopatologia Experimental, Universidade Federal de Ouro Preto, UFOP, Ouro Preto, Brazil
| | - Daniela Caldeira Costa
- Laboratório de Bioquímica Metabólica, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| |
Collapse
|
16
|
Kaito S, Takeshita JI, Iwata M, Sasaki T, Hosaka T, Shizu R, Yoshinari K. Utility of human cytochrome P450 inhibition data in the assessment of drug-induced liver injury. Xenobiotica 2024; 54:411-419. [PMID: 38315106 DOI: 10.1080/00498254.2024.2312505] [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: 12/13/2023] [Accepted: 01/28/2024] [Indexed: 02/07/2024]
Abstract
Drug-induced liver injury (DILI) is a major cause of drug development discontinuation and drug withdrawal from the market, but there are no golden standard methods for DILI risk evaluation. Since we had found the association between DILI and CYP1A1 or CYP1B1 inhibition, we further evaluated the utility of cytochrome P450 (P450) inhibition assay data for DILI risk evaluation using decision tree analysis.The inhibitory activity of drugs with DILI concern (DILI drugs) and no DILI concern (no-DILI drugs) against 10 human P450s was assessed using recombinant enzymes and luminescent substrates. The drugs were also subjected to cytotoxicity assays and high-content analysis using HepG2 cells. Molecular descriptors were calculated by alvaDesc.Decision tree analysis was performed with the data obtained as variables with or without P450-inhibitory activity to discriminate between DILI drugs and no-DILI drugs. The accuracy was significantly higher when P450-inhibitory activity was included. After the decision tree discrimination, the drugs were further discriminated with the P450-inhibitory activity. The results demonstrated that many false-positive and false-negative drugs were correctly discriminated by using the P450 inhibition data.These results suggest that P450 inhibition assay data are useful for DILI risk evaluation.
Collapse
Affiliation(s)
- Shunnosuke Kaito
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Jun-Ichi Takeshita
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
- Research Institute of Science for Safety and Sustainability, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Misaki Iwata
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Takamitsu Sasaki
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Takuomi Hosaka
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Ryota Shizu
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kouichi Yoshinari
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
17
|
Chung E, Wen X, Jia X, Ciallella HL, Aleksunes LM, Zhu H. Hybrid non-animal modeling: A mechanistic approach to predict chemical hepatotoxicity. JOURNAL OF HAZARDOUS MATERIALS 2024; 471:134297. [PMID: 38677119 PMCID: PMC11519847 DOI: 10.1016/j.jhazmat.2024.134297] [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: 01/08/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024]
Abstract
Developing mechanistic non-animal testing methods based on the adverse outcome pathway (AOP) framework must incorporate molecular and cellular key events associated with target toxicity. Using data from an in vitro assay and chemical structures, we aimed to create a hybrid model to predict hepatotoxicants. We first curated a reference dataset of 869 compounds for hepatotoxicity modeling. Then, we profiled them against PubChem for existing in vitro toxicity data. Of the 2560 resulting assays, we selected the mitochondrial membrane potential (MMP) assay, a high-throughput screening (HTS) tool that can test chemical disruptors for mitochondrial function. Machine learning was applied to develop quantitative structure-activity relationship (QSAR) models with 2536 compounds tested in the MMP assay for screening new compounds. The MMP assay results, including QSAR model outputs, yielded hepatotoxicity predictions for reference set compounds with a Correct Classification Ratio (CCR) of 0.59. The predictivity improved by including 37 structural alerts (CCR = 0.8). We validated our model by testing 37 reference set compounds in human HepG2 hepatoma cells, and reliably predicting them for hepatotoxicity (CCR = 0.79). This study introduces a novel AOP modeling strategy that combines public HTS data, computational modeling, and experimental testing to predict chemical hepatotoxicity.
Collapse
Affiliation(s)
- Elena Chung
- Department of Chemistry and Biochemistry, Rowan University, NJ, USA; Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA, USA
| | - Xia Wen
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
| | - Xuelian Jia
- Department of Chemistry and Biochemistry, Rowan University, NJ, USA; Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA, USA
| | - Heather L Ciallella
- Department of Toxicology, Cuyahoga County Medical Examiner's Office, Cleveland, OH, USA
| | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
| | - Hao Zhu
- Department of Chemistry and Biochemistry, Rowan University, NJ, USA; Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
18
|
Yu D, Hales BF, Robaire B. Organophosphate ester flame retardants and plasticizers affect the phenotype and function of HepG2 liver cells. Toxicol Sci 2024; 199:261-275. [PMID: 38518089 PMCID: PMC11131028 DOI: 10.1093/toxsci/kfae034] [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] [Indexed: 03/24/2024] Open
Abstract
Exposure to the organophosphate esters (OPEs), used as flame retardants and plasticizers, is associated with a variety of adverse health effects including an increase in the incidence of fatty liver diseases. The goal of this study was to investigate the effects of six OPEs, all detected in Canadian house dust, on the phenotype and function of HepG2 liver cells. We used high-content imaging to investigate the effects of these OPEs on cell survival, mitochondria, oxidative stress, lipid droplets, and lysosomes. Effects on the autophagy/lipophagy pathway were evaluated using confocal microscopy. The triaryl OPEs (isopropylated triphenylphosphate [IPPP], tris(methylphenyl) phosphate [TMPP], and triphenyl phosphate [TPHP]) were more cytotoxic than non-triaryl OPEs (tris(2-butoxyethyl) phosphate [TBOEP], tris(1-chloro-2-propyl) phosphate [TCIPP], and tris(1,3-dichloro-2-propyl) phosphate [TDCIPP]). Exposure to most OPEs increased total mitochondria, reduced reactive oxygen species, and increased total lipid droplet areas and lysosomal intensity. Potency ranking was done using the lowest benchmark concentration/administered equivalent dose method and toxicological prioritization index analyses to integrate all phenotypic endpoints. IPPP, TBOEP, and TPHP ranked as the most potent OPEs, whereas TMPP, TCIPP, and TDCIPP were relatively less bioactive. Confocal microscopic analysis demonstrated that IPPP reduced the colocalization of lipid droplets (PLIN2), lysosomes (LAMP1), and autophagosomes (p62), disrupting autophagy. In contrast, TBOEP rescued cells from bafilomycin A1-induced inhibition of autophagy and/or increased autophagic flux. Together, these data demonstrate that OPEs have adverse effects on HepG2 cells. Further, OPE-induced dysregulation of autophagy may contribute to the association between OPE exposure and adverse effects on liver lipid homeostasis.
Collapse
Affiliation(s)
- Dongwei Yu
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Barbara F Hales
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Bernard Robaire
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
- Department of Obstetrics and Gynecology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| |
Collapse
|
19
|
Dogra S, Koul B, Singh J, Mishra M, Yadav D. Phytochemical Analysis, Antimicrobial Screening and In Vitro Pharmacological Activity of Artemisia vestita Leaf Extract. Molecules 2024; 29:1829. [PMID: 38675649 PMCID: PMC11054168 DOI: 10.3390/molecules29081829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/03/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Artemisia vestita Wall. Ex Besser is a folklore medicinal plant that belongs to Asteraceae family and a treasure trove of drugs. The aim of this research study was to investigate the phytoconstituents, antimicrobial activity, antioxidant, anti-inflammatory, cytotoxicity and wound healing potential of A. vestita leaf extract (ALE). Phytochemical analysis of the ALE was carried out by Soxhlet extraction and GCMS (gas chromatography-mass spectrometry) analysis. Antimicrobial activity was performed by the agar well diffusion method against selected bacterial and fungal strains. Free radical scavenging potential was evaluated by DPPH (2,2-Diphenyl-1-picrylhydrazyl), ABTS (2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid)) and FRAP (Ferric reducing antioxidant power) assays. Anti-inflammatory activity was performed by enzyme inhibition assay-COXII. The cytotoxicity of ALE on HaCaT cells was studied via MTT (3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide) assay. An in vitro scratch assay was performed for the evaluation of the wound healing property of ALE. It showed satisfactory antimicrobial activity against Staphylococcus aureus (14.2 ± 0.28 mm), Escherichia coli (17.6 ± 0.52 mm), Bacillus subtilis (13.1 ± 0.37 mm), Streptococcus pyogenes (17.3 ± 0.64 mm), Proteus mirabilis (9.4 ± 0.56 mm), Aspergillus niger (12.7 ± 0.53 mm), Aspergilus flavus (15.3 ± 0.25 mm) and Candida albicans (17.6 ± 0.11 mm). In ALE, 36 phytochemicals were detected by GCMS analysis, but 22 were dominant. Moreover, the ALE was effective in scavenging free radicals with different assays and exhibited reasonable anti-inflammatory activity. The MTT assay revealed that ALE had a cytotoxic effect on the HaCaT cells. The scratch assay showed 94.6% wound closure (after 24 h incubation) compared to the positive control Cipladine, which is remarkable wound healing activity. This is the first report on the wound healing property of A. vestita, which can serve as a potential agent for wound healing and extends knowledge on its therapeutic potential.
Collapse
Affiliation(s)
- Shivani Dogra
- Department of Microbiology, Lovely Professional University, Phagwara 144411, Punjab, India;
| | - Bhupendra Koul
- Department of Biotechnology, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Joginder Singh
- Department of Botany, Nagaland University, Zunheboto 798627, Zunheboto, India;
| | - Meerambika Mishra
- Department of Infectious Disease and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA;
| | - Dhananjay Yadav
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
20
|
Gabriel V, Lincoln A, Zdyrski C, Ralston A, Wickham H, Honold S, Ahmed BH, Paukner K, Feauto R, Merodio MM, Piñeyro P, Meyerholz D, Allenspach K, Mochel JP. Evaluation of different media compositions promoting hepatocyte differentiation in the canine liver organoid model. Heliyon 2024; 10:e28420. [PMID: 38590903 PMCID: PMC10999936 DOI: 10.1016/j.heliyon.2024.e28420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 04/10/2024] Open
Abstract
Organoids are 3-dimensional (3D) self-assembled structures capable of replicating the microanatomy and physiology of the epithelial components of their organ of origin. Adult stem cell (ASC) derived organoids from the liver have previously been shown to differentiate into primarily mature cholangiocytes, and their partial differentiation into functional hepatocytes can be promoted using specific media compositions. While full morphological differentiation of mature hepatocytes from ASCs has not yet been reported for any species, the functional differentiation can be approximated using various media compositions. Six differentiation media formulations from published studies on hepatic organoids were used for the differentiation protocol. Target species for these protocols were humans, mice, cats, and dogs, and encompassed various combinations and concentrations of four major hepatocyte media components: Bone morphogenetic protein 7 (BMP7), Fibroblast Growth Factor 19 (FGF19), Dexamethasone (Dex), and Gamma-Secretase Inhibitor IX (DAPT). Additionally, removing R-spondin from basic organoid media has previously been shown to drive the differentiation of ASC into mature hepatocytes. Differentiation media (N = 20) were designed to encompass combinations of the four major hepatocyte media components. The preferred differentiation of ASC-derived organoids from liver tissue into mature hepatocytes over cholangiocytes was confirmed by albumin production in the culture supernatant. Out of the twenty media compositions tested, six media resulted in the production of the highest amounts of albumin in the supernatant of the organoids. The cell lines cultured using these six media were further characterized via histological staining, transmission electron microscopy, RNA in situ hybridization, analysis of gene expression patterns, immunofluorescence, and label-free proteomics. The results indicate that preferential hepatocyte maturation from canine ADC-derived organoids from liver tissue is mainly driven by Dexamethasone and DAPT components. FGF19 did not enhance organoid differentiation but improved cell culture survival. Furthermore, we confirm that removing R-spondin from the media is crucial for establishing mature hepatic organoid cultures.
Collapse
Affiliation(s)
- Vojtech Gabriel
- SMART Lab, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Addison Lincoln
- SMART Lab, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Christopher Zdyrski
- SMART Lab, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
- 3D Health Solutions Inc., Ames, IA, USA
- Precision One Health Initiative, Department of Pathology, University of Georgia College of Veterinary Medicine, 30602, Athens, GA, USA
| | | | - Hannah Wickham
- SMART Lab, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Sydney Honold
- SMART Lab, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Basant H. Ahmed
- SMART Lab, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Karel Paukner
- Laboratory for Atherosclerosis Research, Institute for Clinical and Experimental Medicine, Prague, CZ, Czech Republic
| | - Ryan Feauto
- SMART Lab, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Maria M. Merodio
- SMART Lab, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Pablo Piñeyro
- Veterinary Diagnostic Laboratory, Iowa State University, Ames, IA, USA
| | - David Meyerholz
- Department of Pathology, University of Iowa, Iowa City, IA, USA
| | - Karin Allenspach
- SMART Lab, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
- 3D Health Solutions Inc., Ames, IA, USA
- Precision One Health Initiative, Department of Pathology, University of Georgia College of Veterinary Medicine, 30602, Athens, GA, USA
| | - Jonathan P. Mochel
- 3D Health Solutions Inc., Ames, IA, USA
- Precision One Health Initiative, Department of Pathology, University of Georgia College of Veterinary Medicine, 30602, Athens, GA, USA
| |
Collapse
|
21
|
Liu G, Zhou W, Zhang X, Zhu J, Xu X, Li Y, Zhang J, Wen C, Liang L, Liu X, Xu X. Toxicity and oxidative stress of HepG2 and HL-7702 cells induced by PAH4 using oil as a carrier. Food Res Int 2024; 178:113988. [PMID: 38309887 DOI: 10.1016/j.foodres.2024.113988] [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: 09/19/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 02/05/2024]
Abstract
Polycyclic aromatic hydrocarbons (PAHs), a widespread class of food pollutants, are commonly exposed to humans along with edible oil. The dietary exposure pattern of PAH4 was simulated to study the toxicity and oxidative stress of oil-based PAH4 on hepatocytes. The findings demonstrated that oil-based PAH4 induced cell viability and mitochondrial membrane potential decreased and promoted apoptosis and oxidative stress in a concentration-dependent manner. Benzo[a]pyrene had the strongest toxicity and HL-7702 cells were more sensitive to toxicity than HepG2 cells, due to differences in induced CYP1A enzyme activity. Oil-based PAH4 had greater cytotoxicity than PAH4, attributed to the synergistic effect of oil and PAH4. Furthermore, oil-based PAH4 induced oxidative stress in HepG2 and HL-7702 cells through the same AHR-Nrf2-KEAP1 pathway, which was elucidated by detecting genes and proteins expression. This study lays the foundation for elucidating the harm of dietary exposure to PAHs and reminds us that food composition may increase the harm of PAHs.
Collapse
Affiliation(s)
- Guoyan Liu
- School of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Wanli Zhou
- School of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Xu Zhang
- School of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Jie Zhu
- School of Tourism and Cuisine, Yangzhou University, Yangzhou 225127, China
| | - Xiaowei Xu
- School of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Youdong Li
- School of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Jixian Zhang
- School of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Chaoting Wen
- School of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Li Liang
- School of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China
| | - Xiaofang Liu
- School of Tourism and Cuisine, Yangzhou University, Yangzhou 225127, China.
| | - Xin Xu
- School of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China.
| |
Collapse
|
22
|
Xing C, Kemas A, Mickols E, Klein K, Artursson P, Lauschke VM. The choice of ultra-low attachment plates impacts primary human and primary canine hepatocyte spheroid formation, phenotypes, and function. Biotechnol J 2024; 19:e2300587. [PMID: 38403411 DOI: 10.1002/biot.202300587] [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: 10/30/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 02/27/2024]
Abstract
Organotypic three-dimensional liver spheroid cultures in which hepatic cells retain their molecular phenotype and functionality have emerged as powerful tools for preclinical drug development. In recent years a multitude of culture systems have been developed; however, a thorough side-by-side benchmarking of the different methods is lacking. Here, we compared the performance of ten different 96- and 384-well microplate types to support spheroid formation and long-term culture. Specifically, we evaluated differences in spheroid formation kinetics, viability, functionality, expression patterns, and their utility for hepatotoxicity assessments using primary human hepatocytes (PHH) and primary canine hepatocytes (PCH). All 96-well plates enabled formation of PHH liver spheroids, albeit with differences between plates in spheroid size, geometry, and reproducibility. Performance of different 384-wells was less consistent. Only 6/10 microplates supported the formation of PCH aggregates. Interestingly, even if PCH aggregates in these six microplates were more loosely packed than PHH spheroids, they maintained their function and were compatible with long-term pharmacological and toxicological assays. Overall, Corning and Biofloat plates showed the best performance in the formation of both human and canine liver spheroids with highest viability, most physiologically relevant phenotypes, superior CYP activity and lowest coefficient of variation in toxicity assays. The presented data constitutes a valuable resource that demonstrates the impacts of current ultra-low attachment plates on liver spheroid metrics and can guide evidence-based plate selection. Combined, these results have important implications for the cross-comparison of different studies and can facilitate the standardization and reproducibility of three-dimensional liver culture experiments.
Collapse
Affiliation(s)
- Chen Xing
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Aurino Kemas
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | | | - Kathrin Klein
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Per Artursson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| |
Collapse
|
23
|
Liu Y, Ge Y, Wu Y, Feng Y, Liu H, Cao W, Xie J, Zhang J. High-Voltage Electrostatic Field Hydrogel Microsphere 3D Culture System Improves Viability and Liver-like Properties of HepG2 Cells. Int J Mol Sci 2024; 25:1081. [PMID: 38256154 PMCID: PMC10816196 DOI: 10.3390/ijms25021081] [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: 12/02/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Three-dimensional (3D) hepatocyte models have become a research hotspot for evaluating drug metabolism and hepatotoxicity. Compared to two-dimensional (2D) cultures, 3D cultures are better at mimicking the morphology and microenvironment of hepatocytes in vivo. However, commonly used 3D culture techniques are not suitable for high-throughput drug screening (HTS) due to their high cost, complex handling, and inability to simulate cell-extracellular matrix (ECM) interactions. This article describes a method for rapid and reproducible 3D cell cultures with ECM-cell interactions based on 3D culture instrumentation to provide more efficient HTS. We developed a microsphere preparation based on a high-voltage electrostatic (HVE) field and used sodium alginate- and collagen-based hydrogels as scaffolds for 3D cultures of HepG2 cells. The microsphere-generating device enables the rapid and reproducible preparation of bioactive hydrogel microspheres. This 3D culture system exhibited better cell viability, heterogeneity, and drug-metabolizing activity than 2D and other 3D culture models, and the long-term culture characteristics of this system make it suitable for predicting long-term liver toxicity. This system improves the overall applicability of HepG2 spheroids in safety assessment studies, and this simple and controllable high-throughput-compatible method shows potential for use in drug toxicity screening assays and mechanistic studies.
Collapse
Affiliation(s)
- Yi Liu
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (Y.L.); (Y.W.)
- The CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.G.); (Y.F.); (H.L.); (W.C.); (J.X.)
| | - Yang Ge
- The CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.G.); (Y.F.); (H.L.); (W.C.); (J.X.)
| | - Yanfan Wu
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (Y.L.); (Y.W.)
- The CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.G.); (Y.F.); (H.L.); (W.C.); (J.X.)
| | - Yongtong Feng
- The CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.G.); (Y.F.); (H.L.); (W.C.); (J.X.)
| | - Han Liu
- The CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.G.); (Y.F.); (H.L.); (W.C.); (J.X.)
| | - Wei Cao
- The CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.G.); (Y.F.); (H.L.); (W.C.); (J.X.)
| | - Jinsong Xie
- The CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.G.); (Y.F.); (H.L.); (W.C.); (J.X.)
| | - Jingzhong Zhang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (Y.L.); (Y.W.)
- The CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.G.); (Y.F.); (H.L.); (W.C.); (J.X.)
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
24
|
Smiriglia A, Lorito N, Serra M, Perra A, Morandi A, Kowalik MA. Sex difference in liver diseases: How preclinical models help to dissect the sex-related mechanisms sustaining NAFLD and hepatocellular carcinoma. iScience 2023; 26:108363. [PMID: 38034347 PMCID: PMC10682354 DOI: 10.1016/j.isci.2023.108363] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023] Open
Abstract
Only a few preclinical findings are confirmed in the clinic, posing a critical issue for clinical development. Therefore, identifying the best preclinical models can help to dissect molecular and mechanistic insights into liver disease pathogenesis while being clinically relevant. In this context, the sex relevance of most preclinical models has been only partially considered. This is particularly significant in NAFLD and HCC, which have a higher prevalence in men when compared to pre-menopause women but not to those in post-menopausal status, suggesting a role for sex hormones in the pathogenesis of the diseases. This review gathers the sex-relevant findings and the available preclinical models focusing on both in vitro and in vivo studies and discusses the potential implications and perspectives of introducing the sex effect in the selection of the best preclinical model. This is a critical aspect that would help to tailor personalized therapies based on sex.
Collapse
Affiliation(s)
- Alfredo Smiriglia
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Nicla Lorito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Marina Serra
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Andrea Perra
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Marta Anna Kowalik
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| |
Collapse
|
25
|
Liu Y, Wu K, Fu Y, Li W, Zhao XY. Slc7a11 stimulates glutathione synthesis to preserve fatty acid metabolism in primary hepatocytes. Redox Rep 2023; 28:2260646. [PMID: 37750478 PMCID: PMC10540662 DOI: 10.1080/13510002.2023.2260646] [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] [Indexed: 09/27/2023] Open
Abstract
Primary hepatocytes are widely used as a tool for studying metabolic function and regulation in the liver. However, the metabolic properties of primary hepatocytes are gradually lost after isolation. Here, we illustrated that fatty acid metabolism is the major compromised metabolic process in isolated primary hepatocytes, along with drastically decreased GSH and ROS content, while lipid peroxidation is increased. Gain- and loss-of-function studies revealed that Slc7a11 expression is critical in maintaining fatty acid metabolism and facilitating hormone-induced fatty acid metabolic events, which is synergistic with dexamethasone treatment. Intriguingly, Slc7a11 expression and dexamethasone treatment cooperatively upregulated AKT and AMPK signaling and mitochondrial complex expression in primary hepatocytes. Furthermore, direct treatment with reduced GSH or inhibition of ferroptosis is sufficient to drive protective effects on fatty acid metabolism in primary hepatocytes. Our results demonstrate that Slc7a11 expression in isolated primary hepatocytes induces GSH production, which protects against ferroptosis, to increase fatty acid metabolic gene expression, AKT and AMPK signaling and mitochondrial function in synergy with dexamethasone treatment, thereby efficiently preserving primary hepatocyte metabolic signatures, thus providing a promising approach to better reserve primary hepatocyte metabolic activities after isolation to potentially improve the understanding of liver biological functions from studies using primary hepatocytes.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Kaimin Wu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yinkun Fu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Wenyan Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Xu-Yun Zhao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
26
|
Omori H, Chikamoto J, Nagahara M, Hirata M, Otoi T. Evaluating variations in bilirubin glucuronidation activity by protease inhibitors in canine and human primary hepatocytes cultured in a 3D culture system. Toxicol In Vitro 2023; 93:105689. [PMID: 37660998 DOI: 10.1016/j.tiv.2023.105689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/18/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023]
Abstract
Bilirubin is excreted into the bile from hepatocytes, mainly as monoglucuronosyl and bisglucuronosyl conjugates, reflecting bilirubin glucuronidation activity. However, there is limited information on the in vitro evaluation of liver cell lines or primary hepatocytes. This study aimed to investigate variations in the bilirubin metabolic function of canine and human hepatocyte spheroids formed in a three-dimensional (3D) culture system indicated by the formation of bilirubin glucuronides when protease inhibitors such as atazanavir, indinavir, ritonavir, and nelfinavir were treated with bilirubin. The culture supernatant was collected for bilirubin glucuronidation assessment and the cells were used to evaluate viability. On day 8 of culture, both canine and human hepatocyte spheroids showed high albumin secretion and distinct spheroid formation, and their bilirubin glucuronidation activities were evaluated considering cell viability. Treatment with atazanavir and ritonavir remarkably inhibited bilirubin glucuronide formation, wherein atazanavir showed the highest inhibition, particularly in human hepatocyte spheroids. These results may reflect the effects on cellular uptake of bilirubin and its intracellular metabolic function. Thus, primary hepatocytes cultured in a 3D culture system may be a useful in vitro system for the comprehensive evaluation of bilirubin metabolic function and risk assessment in bilirubin metabolic disorders for drug development.
Collapse
Affiliation(s)
- Hisayoshi Omori
- Bio-Innovation Research Center, Tokushima University, Tokushima, Japan; Preclinical Basic Research, Taiho Pharmaceutical Co., Ltd., Tokushima, Japan
| | - Junko Chikamoto
- Preclinical Basic Research, Taiho Pharmaceutical Co., Ltd., Tokushima, Japan
| | - Megumi Nagahara
- Bio-Innovation Research Center, Tokushima University, Tokushima, Japan; Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Maki Hirata
- Bio-Innovation Research Center, Tokushima University, Tokushima, Japan; Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Takeshige Otoi
- Bio-Innovation Research Center, Tokushima University, Tokushima, Japan; Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan.
| |
Collapse
|
27
|
Gronert A, Zierau O, Thieme D, Keiler AM. Effect of HepG2 cell 3D cultivation on the metabolism of the anabolic androgenic steroid metandienone. Drug Test Anal 2023; 15:1319-1328. [PMID: 36772854 DOI: 10.1002/dta.3455] [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: 12/05/2022] [Revised: 01/23/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023]
Abstract
The elucidation of the metabolic fate of prohibited substances is crucial for the abuse detection. The human hepatocyte cell line HepG2 can be used to study biotransformation. In order to improve this in vitro model system, we compared the HepG2 spheroid generation using three different techniques: a forced floating, a scaffold-free and a scaffold-based method. We characterized the spheroids with regard to the expression levels of the proliferation marker Mki67, the liver-specific marker albumin and biotransformation enzymes. Moreover, the metandienone metabolite pattern was comparatively analysed by high-performance liquid chromatography mass spectrometry. With all three techniques, HepG2 spheroids were generated showing a degree of differentiation. The forced floating method resulted in very large spheroids (1 mm in diameter) showing signs of necrosis in the centre and a very low metandienone conversion rate. The spheroids formed by the two other techniques were comparable in size with 0.5 mm in diameter on average. Among the three different 3D cultivation methods, the HepG2 spheroids formed on Matrigel® as extracellular matrix were the most promising regarding biotransformation studies on anabolic androgenic steroids. Prospectively, HepG2 spheroids are a promising in vitro model system to study multidrug setups, drug-drug interactions and the biotransformation of other substance classes.
Collapse
Affiliation(s)
- Anika Gronert
- Institute of Doping Analysis and Sports Biochemistry Dresden, Kreischa, Germany
| | - Oliver Zierau
- Environmental Monitoring and Endocrinology, Faculty of Biology, Technische Universität Dresden, Dresden, Germany
| | - Detlef Thieme
- Institute of Doping Analysis and Sports Biochemistry Dresden, Kreischa, Germany
| | - Annekathrin Martina Keiler
- Institute of Doping Analysis and Sports Biochemistry Dresden, Kreischa, Germany
- Environmental Monitoring and Endocrinology, Faculty of Biology, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
28
|
Botte E, Mancini P, Magliaro C, Ahluwalia A. A sense of proximity: Cell packing modulates oxygen consumption. APL Bioeng 2023; 7:036111. [PMID: 37664826 PMCID: PMC10468216 DOI: 10.1063/5.0160422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023] Open
Abstract
Accurately modeling oxygen transport and consumption is crucial to predict metabolic dynamics in cell cultures and optimize the design of tissue and organ models. We present a methodology to characterize the Michaelis-Menten oxygen consumption parameters in vitro, integrating novel experimental techniques and computational tools. The parameters were derived for hepatic cell cultures with different dimensionality (i.e., 2D and 3D) and with different surface and volumetric densities. To quantify cell packing regardless of the dimensionality of cultures, we devised an image-based metric, referred to as the proximity index. The Michaelis-Menten parameters were related to the proximity index through an uptake coefficient, analogous to a diffusion constant, enabling the quantitative analysis of oxygen dynamics across dimensions. Our results show that Michaelis-Menten parameters are not constant for a given cell type but change with dimensionality and cell density. The maximum consumption rate per cell decreases significantly with cell surface and volumetric density, while the Michaelis-Menten constant tends to increase. In addition, the dependency of the uptake coefficient on the proximity index suggests that the oxygen consumption rate of hepatic cells is superadaptive, as they modulate their oxygen utilization according to its local availability and to the proximity of other cells. We describe, for the first time, how cells consume oxygen as a function of cell proximity, through a quantitative index, which combines cell density and dimensionality. This study enhances our understanding of how cell-cell interaction affects oxygen dynamics and enables better prediction of aerobic metabolism in tissue models, improving their translational value.
Collapse
Affiliation(s)
| | | | | | - Arti Ahluwalia
- Author to whom correspondence should be addressed:. Tel.: +39 0502217062
| |
Collapse
|
29
|
Todorova VK, Byrum SD, Mackintosh SG, Jamshidi-Parsian A, Gies AJ, Washam CL, Jenkins SV, Spiva T, Bowman E, Reyna NS, Griffin RJ, Makhoul I. Exosomal MicroRNA and Protein Profiles of Hepatitis B Virus-Related Hepatocellular Carcinoma Cells. Int J Mol Sci 2023; 24:13098. [PMID: 37685904 PMCID: PMC10487651 DOI: 10.3390/ijms241713098] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Infection with hepatitis B virus (HBV) is a main risk factor for hepatocellular carcinoma (HCC). Extracellular vesicles, such as exosomes, play an important role in tumor development and metastasis, including regulation of HBV-related HCC. In this study, we have characterized exosome microRNA and proteins released in vitro from hepatitis B virus (HBV)-related HCC cell lines SNU-423 and SNU-182 and immortalized normal hepatocyte cell lines (THLE2 and THLE3) using microRNA sequencing and mass spectrometry. Bioinformatics, including functional enrichment and network analysis, combined with survival analysis using data related to HCC in The Cancer Genome Atlas (TCGA) database, were applied to examine the prognostic significance of the results. More than 40 microRNAs and 200 proteins were significantly dysregulated (p < 0.05) in the exosomes released from HCC cells in comparison with the normal liver cells. The functional analysis of the differentially expressed exosomal miRNAs (i.e., mir-483, mir-133a, mir-34a, mir-155, mir-183, mir-182), their predicted targets, and exosomal differentially expressed proteins (i.e., POSTN, STAM, EXOC8, SNX9, COL1A2, IDH1, FN1) showed correlation with pathways associated with HBV, virus activity and invasion, exosome formation and adhesion, and exogenous protein binding. The results from this study may help in our understanding of the role of HBV infection in the development of HCC and in the development of new targets for treatment or non-invasive predictive biomarkers of HCC.
Collapse
Affiliation(s)
- Valentina K. Todorova
- Department of Internal Medicine/Division of Hematology/Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Stephanie D. Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.D.B.); (S.G.M.); (A.J.G.); (C.L.W.)
| | - Samuel G. Mackintosh
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.D.B.); (S.G.M.); (A.J.G.); (C.L.W.)
| | - Azemat Jamshidi-Parsian
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.J.-P.); (S.V.J.); (R.J.G.)
| | - Allen J. Gies
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.D.B.); (S.G.M.); (A.J.G.); (C.L.W.)
| | - Charity L. Washam
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.D.B.); (S.G.M.); (A.J.G.); (C.L.W.)
| | - Samir V. Jenkins
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.J.-P.); (S.V.J.); (R.J.G.)
| | - Timothy Spiva
- Biology Department, Ouachita Baptist University, Arkadelphia, AR 71998, USA; (T.S.); (E.B.); (N.S.R.)
| | - Emily Bowman
- Biology Department, Ouachita Baptist University, Arkadelphia, AR 71998, USA; (T.S.); (E.B.); (N.S.R.)
| | - Nathan S. Reyna
- Biology Department, Ouachita Baptist University, Arkadelphia, AR 71998, USA; (T.S.); (E.B.); (N.S.R.)
| | - Robert J. Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.J.-P.); (S.V.J.); (R.J.G.)
| | - Issam Makhoul
- Department of Internal Medicine/Division of Hematology/Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| |
Collapse
|
30
|
Lavrenova LG, Sukhikh TS, Glinskaya LA, Trubina SV, Zvereva VV, Lavrov AN, Klyushova LS, Artem’ev AV. Synthesis, Structure, and Magnetic and Biological Properties of Copper(II) Complexes with 1,3,4-Thiadiazole Derivatives. Int J Mol Sci 2023; 24:13024. [PMID: 37629205 PMCID: PMC10455747 DOI: 10.3390/ijms241613024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/11/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
New coordination compounds of copper(II) with 2,5-bis(ethylthio)-1,3,4-thiadiazole (L1) and 2,5-bis(pyridylmethylthio)-1,3,4-thiadiazole (L2) with compositions Cu(L1)2Br2, Cu(L1)(C2N3)2, Cu(L2)Cl2, and Cu(L2)Br2 were prepared. The complexes were identified and studied by CHN analysis, infrared (IR) spectroscopy, powder X-Ray diffraction (XRD), and static magnetic susceptibility. The crystal structures of Cu(II) complexes with L1 were determined. The structures of the coordination core of complexes Cu(L2)Cl2 and Cu(L2)Br2 were determined by Extended X-ray absorption fine structure (EXAFS) spectroscopy. Magnetization measurements have revealed various magnetic states in the studied complexes, ranging from an almost ideal paramagnet in Cu(L1)2Br2 to alternating-exchange antiferromagnetic chains in Cu(L1)(C2N3)2, where double dicyanamide bridges provide an unusually strong exchange interaction (J1/kB ≈ -23.5 K; J2/kB ≈ -20.2 K) between Cu(II) ions. The cytotoxic activity of copper(II) complexes with L2 was estimated on the human cell lines of breast adenocarcinoma (MCF-7) and hepatocellular carcinoma (HepG2).
Collapse
Affiliation(s)
- Lyudmila G. Lavrenova
- Nikolaev Institute of Inorganic Chemistry, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia; (T.S.S.); (L.A.G.); (S.V.T.); (V.V.Z.); (A.N.L.); (A.V.A.)
| | - Taisiya S. Sukhikh
- Nikolaev Institute of Inorganic Chemistry, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia; (T.S.S.); (L.A.G.); (S.V.T.); (V.V.Z.); (A.N.L.); (A.V.A.)
| | - Lyudmila A. Glinskaya
- Nikolaev Institute of Inorganic Chemistry, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia; (T.S.S.); (L.A.G.); (S.V.T.); (V.V.Z.); (A.N.L.); (A.V.A.)
| | - Svetlana V. Trubina
- Nikolaev Institute of Inorganic Chemistry, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia; (T.S.S.); (L.A.G.); (S.V.T.); (V.V.Z.); (A.N.L.); (A.V.A.)
| | - Valentina V. Zvereva
- Nikolaev Institute of Inorganic Chemistry, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia; (T.S.S.); (L.A.G.); (S.V.T.); (V.V.Z.); (A.N.L.); (A.V.A.)
| | - Alexander N. Lavrov
- Nikolaev Institute of Inorganic Chemistry, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia; (T.S.S.); (L.A.G.); (S.V.T.); (V.V.Z.); (A.N.L.); (A.V.A.)
| | - Lyubov S. Klyushova
- Institute of Molecular Biology and Biophysics, Federal Research Center for Fundamental and Translational Medicine, Novosibirsk 630117, Russia;
| | - Alexander V. Artem’ev
- Nikolaev Institute of Inorganic Chemistry, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia; (T.S.S.); (L.A.G.); (S.V.T.); (V.V.Z.); (A.N.L.); (A.V.A.)
| |
Collapse
|
31
|
Tamargo-Rubio I, Simpson AB, Hoogerland JA, Fu J. Human induced pluripotent stem cell-derived liver-on-a-chip for studying drug metabolism: the challenge of the cytochrome P450 family. Front Pharmacol 2023; 14:1223108. [PMID: 37448965 PMCID: PMC10338083 DOI: 10.3389/fphar.2023.1223108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
The liver is the primary organ responsible for the detoxification and metabolism of drugs. To date, a lack of preclinical models that accurately emulate drug metabolism by the human liver presents a significant challenge in the drug development pipeline, particularly for predicting drug efficacy and toxicity. In recent years, emerging microfluidic-based organ-on-a-chip (OoC) technologies, combined with human induced pluripotent stem cell (hiPSC) technology, present a promising avenue for the complete recapitulation of human organ biology in a patient-specific manner. However, hiPSC-derived organoids and liver-on-a-chip models have so far failed to sufficiently express cytochrome P450 monooxygenase (CYP450) enzymes, the key enzymes involved in first-pass metabolism, which limits the effectiveness and translatability of these models in drug metabolism studies. This review explores the potential of innovative organoid and OoC technologies for studying drug metabolism and discusses their existing drawbacks, such as low expression of CYP450 genes. Finally, we postulate potential approaches for enhancing CYP450 expression in the hope of paving the way toward developing novel, fully representative liver drug-metabolism models.
Collapse
Affiliation(s)
- Isabel Tamargo-Rubio
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Anna Bella Simpson
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Joanne A. Hoogerland
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
32
|
Otumala AE, Hellen DJ, Luna CA, Delgado P, Dissanayaka A, Ugwumadu C, Oshinowo O, Islam MM, Shen L, Karpen SJ, Myers DR. Opportunities and considerations for studying liver disease with microphysiological systems on a chip. LAB ON A CHIP 2023; 23:2877-2898. [PMID: 37282629 DOI: 10.1039/d2lc00940d] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Advances in microsystem engineering have enabled the development of highly controlled models of the liver that better recapitulate the unique in vivo biological conditions. In just a few short years, substantial progress has been made in creating complex mono- and multi-cellular models that mimic key metabolic, structural, and oxygen gradients crucial for liver function. Here we review: 1) the state-of-the-art in liver-centric microphysiological systems and 2) the array of liver diseases and pressing biological and therapeutic challenges which could be investigated with these systems. The engineering community has unique opportunities to innovate with new liver-on-a-chip devices and partner with biomedical researchers to usher in a new era of understanding of the molecular and cellular contributors to liver diseases and identify and test rational therapeutic modalities.
Collapse
Affiliation(s)
- Adiya E Otumala
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Dominick J Hellen
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - C Alessandra Luna
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Priscilla Delgado
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Anjana Dissanayaka
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Chidozie Ugwumadu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Oluwamayokun Oshinowo
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Md Mydul Islam
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Luyao Shen
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Saul J Karpen
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - David R Myers
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
33
|
Riddick DS. Fifty Years of Aryl Hydrocarbon Receptor Research as Reflected in the Pages of Drug Metabolism and Disposition. Drug Metab Dispos 2023; 51:657-671. [PMID: 36653119 DOI: 10.1124/dmd.122.001009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
The induction of multiple drug-metabolizing enzymes by halogenated and polycyclic aromatic hydrocarbon toxicants is mediated by the aryl hydrocarbon receptor (AHR). This fascinating receptor also has natural dietary and endogenous ligands, and much is now appreciated about the AHR's developmental and physiologic roles, as well as its importance in cancer and other diseases. The past several years has witnessed increasing emphasis on understanding the multifaceted roles of the AHR in the immune system. Most would agree that the "discovery" of the AHR occurred in 1976, with the report of specific binding of a high affinity radioligand in mouse liver, just three years after the launch of the journal Drug Metabolism and Disposition (DMD) in 1973. Over the ensuing 50 years, the AHR and DMD have led parallel and often intersecting lives. The overall goal of this mini-review is to provide a decade-by-decade overview of major historical landmark discoveries in the AHR field and to highlight the numerous contributions made by publications appearing in the pages of DMD. It is hoped that this historical tour might inspire current and future research in the AHR field. SIGNIFICANCE STATEMENT: With the launch of Drug Metabolism and Disposition (DMD) in 1973 and the discovery of the aryl hydrocarbon receptor (AHR) in 1976, the journal and the receptor have led parallel and often intersecting lives over the past 50 years. Tracing the history of the AHR can reveal how knowledge in the field has evolved to the present and highlight the important contributions made by discoveries reported in DMD. This may inspire additional DMD papers reporting future AHR landmark discoveries.
Collapse
Affiliation(s)
- David S Riddick
- Department of Pharmacology and Toxicology, Medical Sciences Building, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
34
|
Sukumaran S, Tan M, Ben-Uliel SF, Zhang H, De Zotti M, Chua MS, So SK, Qvit N. Rational design, synthesis and structural characterization of peptides and peptidomimetics to target Hsp90/Cdc37 interaction for treating hepatocellular carcinoma. Comput Struct Biotechnol J 2023; 21:3159-3172. [PMID: 37304004 PMCID: PMC10250827 DOI: 10.1016/j.csbj.2023.05.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/23/2023] [Accepted: 05/23/2023] [Indexed: 06/13/2023] Open
Abstract
Heat shock protein 90 (Hsp90) and cell division cycle 37 (Cdc37) work together as a molecular chaperone complex to regulate the activity of a multitude of client protein kinases. These kinases belong to a wide array of intracellular signaling networks that mediate multiple cellular processes including proliferation. As a result, Hsp90 and Cdc37 represent innovative therapeutic targets in various cancers (such as leukemia, multiple myeloma, and hepatocellular carcinoma (HCC)) in which their expression levels are elevated. Conventional small molecule Hsp90 inhibitors act by blocking the conserved adenosine triphosphate (ATP) binding site. However, by targeting less conserved sites in a more specific manner, peptides and peptidomimetics (modified peptides) hold potential as more efficacious and less toxic alternatives to the conventional small molecule inhibitors. Using a rational approach, we herein developed bioactive peptides targeting Hsp90/Cdc37 interaction. A six amino acid linear peptide derived from Cdc37, KTGDEK, was designed to target Hsp90. We used in silico computational docking to first define its mode of interaction, and binding orientation, and then conjugated the peptide with a cell penetrating peptide, TAT, and a fluorescent dye to confirm its ability to colocalize with Hsp90 in HCC cells. Based on the parent linear sequence, we developed a peptidomimetics library of pre-cyclic and cyclic derivatives. These peptidomimetics were evaluated for their binding affinity to Hsp90, and bioactivity in HCC cell lines. Among them, a pre-cyclic peptidomimetic demonstrates high binding affinity and bioactivity in HCC cells, causing reduced cell proliferation that is associated with induction of cell apoptosis, and down-regulation of phosphorylated MEK1/2. Overall, this generalized approach of rational design, structural optimization, and cellular validation of 'drug-like' peptidomimetics against Hsp90/Cdc37 offers a feasible and promising way to design novel therapeutic agents for malignancies and other diseases that are dependent on this molecular chaperone complex.
Collapse
Affiliation(s)
- Surya Sukumaran
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel
| | - Mingdian Tan
- Asian Liver Center, Department of Surgery, Stanford University School of Medicine, 1201 Welch Road, Palo Alto, CA 94305, USA
| | - Shulamit Fluss Ben-Uliel
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel
| | - Hui Zhang
- Asian Liver Center, Department of Surgery, Stanford University School of Medicine, 1201 Welch Road, Palo Alto, CA 94305, USA
| | - Marta De Zotti
- Department of Chemistry, University of Padova, Via Marzolo 1, 35131 Padova, Italy
| | - Mei-Sze Chua
- Asian Liver Center, Department of Surgery, Stanford University School of Medicine, 1201 Welch Road, Palo Alto, CA 94305, USA
| | - Samuel K. So
- Asian Liver Center, Department of Surgery, Stanford University School of Medicine, 1201 Welch Road, Palo Alto, CA 94305, USA
| | - Nir Qvit
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel
| |
Collapse
|
35
|
Zhou Z, Luo D, Li M, Lao G, Zhou Z, Dinnyés A, Xu W, Sun Q. A Novel Multicellular Placental Barrier Model to Investigate the Effect of Maternal Aflatoxin B 1 Exposure on Fetal-Side Neural Stem Cells. Toxins (Basel) 2023; 15:toxins15050312. [PMID: 37235346 DOI: 10.3390/toxins15050312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Ingestion of food toxins such as aflatoxin B1 (AFB1) during pregnancy may impair fetal neurodevelopment. However, animal model results may not be accurate due to the species' differences, and testing on humans is ethically impermissible. Here, we developed an in vitro human maternal-fetal multicellular model composed of a human hepatic compartment, a bilayer placental barrier, and a human fetal central nervous system compartment using neural stem cells (NSCs) to investigate the effect of AFB1 on fetal-side NSCs. AFB1 passed through the HepG2 hepatocellular carcinoma cells to mimic the maternal metabolic effects. Importantly, even at the limited concentration (0.0641 ± 0.0046 μM) of AFB1, close to the national safety level standard of China (GB-2761-2011), the mixture of AFB1 crossing the placental barrier induced NSC apoptosis. The level of reactive oxygen species in NSCs was significantly elevated and the cell membrane was damaged, causing the release of intracellular lactate dehydrogenase (p < 0.05). The comet experiment and γ-H2AX immunofluorescence assay showed that AFB1 caused significant DNA damage to NSCs (p < 0.05). This study provided a new model for the toxicological evaluation of the effect of food mycotoxin exposure during pregnancy on fetal neurodevelopment.
Collapse
Affiliation(s)
- Zhiwei Zhou
- Key Laboratory of Bio-Resources and Eco-Environment Ministry of the Education, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Dongmei Luo
- Key Laboratory of Bio-Resources and Eco-Environment Ministry of the Education, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Mengxue Li
- Key Laboratory of Bio-Resources and Eco-Environment Ministry of the Education, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Guangjie Lao
- Key Laboratory of Bio-Resources and Eco-Environment Ministry of the Education, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Zhiqiang Zhou
- Department of Food Engineering, Sichuan University, Chengdu 610064, China
| | - András Dinnyés
- Key Laboratory of Bio-Resources and Eco-Environment Ministry of the Education, College of Life Sciences, Sichuan University, Chengdu 610064, China
- BioTalentum Ltd., Aulich Lajos Str. 26, 2100 Godollo, Hungary
- Department of Cell Biology and Molecular Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Wenming Xu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610064, China
- Reproductive Endocrinology and Regulation Laboratory West China Second University Hospital, Sichuan University, Chengdu 610064, China
| | - Qun Sun
- Key Laboratory of Bio-Resources and Eco-Environment Ministry of the Education, College of Life Sciences, Sichuan University, Chengdu 610064, China
| |
Collapse
|
36
|
Rosario FJ, Chopra A, Biggar K, Powell TL, Gupta MB, Jansson T. Placental Remote Control of Fetal Metabolism: Trophoblast mTOR Signaling Regulates Liver IGFBP-1 Phosphorylation and IGF-1 Bioavailability. Int J Mol Sci 2023; 24:7273. [PMID: 37108437 PMCID: PMC10138459 DOI: 10.3390/ijms24087273] [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: 02/21/2023] [Revised: 04/02/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
The mechanisms mediating the restricted growth in intrauterine growth restriction (IUGR) remain to be fully established. Mechanistic target of rapamycin (mTOR) signaling functions as a placental nutrient sensor, indirectly influencing fetal growth by regulating placental function. Increased secretion and the phosphorylation of fetal liver IGFBP-1 are known to markedly decrease the bioavailability of IGF-1, a major fetal growth factor. We hypothesized that an inhibition of trophoblast mTOR increases liver IGFBP-1 secretion and phosphorylation. We collected conditioned media (CM) from cultured primary human trophoblast (PHT) cells with a silenced RAPTOR (specific inhibition of mTOR Complex 1), RICTOR (inhibition of mTOR Complex 2), or DEPTOR (activates both mTOR Complexes). Subsequently, HepG2 cells, a well-established model for human fetal hepatocytes, were cultured in CM from PHT cells, and IGFBP-1 secretion and phosphorylation were determined. CM from PHT cells with either mTORC1 or mTORC2 inhibition caused the marked hyperphosphorylation of IGFBP-1 in HepG2 cells as determined by 2D-immunoblotting while Parallel Reaction Monitoring-Mass Spectrometry (PRM-MS) identified increased dually phosphorylated Ser169 + Ser174. Furthermore, using the same samples, PRM-MS identified multiple CK2 peptides coimmunoprecipitated with IGFBP-1 and greater CK2 autophosphorylation, indicating the activation of CK2, a key enzyme mediating IGFBP-1 phosphorylation. Increased IGFBP-1 phosphorylation inhibited IGF-1 function, as determined by the reduced IGF-1R autophosphorylation. Conversely, CM from PHT cells with mTOR activation decreased IGFBP-1 phosphorylation. CM from non-trophoblast cells with mTORC1 or mTORC2 inhibition had no effect on HepG2 IGFBP-1 phosphorylation. Placental mTOR signaling may regulate fetal growth by the remote control of fetal liver IGFBP-1 phosphorylation.
Collapse
Affiliation(s)
- Fredrick J. Rosario
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Anand Chopra
- Institute of Biochemistry, Carleton University, Ottawa, ON K1S 5B6, Canada
| | - Kyle Biggar
- Institute of Biochemistry, Carleton University, Ottawa, ON K1S 5B6, Canada
| | - Theresa L. Powell
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Biochemistry, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Madhulika B. Gupta
- Department of Biochemistry, University of Western Ontario, London, ON N6A 3K7, Canada
- Department of Pediatrics, University of Western Ontario, London, ON N6A 3K7, Canada
- Children’s Health Research Institute, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
37
|
Onodera R, Jimma Y, Suzuki A, Habano W, Ozawa S, Terashima J. The Regulation Pathway of VEGF Gene Expression Is Different between 2D Cells and 3D Spheroids in Human Lung Cancer Cells. Biol Pharm Bull 2023; 46:608-613. [PMID: 37005305 DOI: 10.1248/bpb.b22-00772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Angiogenesis is involved in the malignant transformation of cancers. Vascular endothelial growth factor (VEGF) is important in inducing angiogenesis. Cultured cells play an important role in analyzing the regulation of VEGF expression, and it is revealed that VEGF expression is induced under hypoxia. However, it has been shown that there are differences in the pathway for gene expression between two-dimensional (2D) cells and in vivo cells. Three-dimensional (3D) spheroids constructed in 3D culture with a gene expression pattern more similar to that of in vivo cells than 2D cells have been used to solve this problem. This study analyzed the VEGF gene expression pathway in 3D spheroids of human lung cancer cells, A549 and H1703. Hypoxia-inducible factor-1α (HIF-1α) and aryl hydrocarbon receptor nuclear translocator (ARNT) regulated VEGF gene expression in 3D spheroids. However, VEGF gene expression was not regulated by HIF-1α in 2D cells. To conclude, we found that the regulatory pathway of VEGF gene expression is different between 2D cells and 3D spheroids in human lung cancer cells. These results suggest the possibility of a new VEGF gene expression regulation pathway in vivo. In addition, they show useful knowledge for the analysis of angiogenesis induction mechanisms and also demonstrate the usefulness of 3D spheroids.
Collapse
Affiliation(s)
- Ryo Onodera
- Division of Pharmacodynamics and Molecular Genetics, School of Pharmacy, Iwate Medical University
| | - Yoko Jimma
- Division of Pharmacodynamics and Molecular Genetics, School of Pharmacy, Iwate Medical University
| | - Anna Suzuki
- Division of Pharmacodynamics and Molecular Genetics, School of Pharmacy, Iwate Medical University
| | - Wataru Habano
- Division of Pharmacodynamics and Molecular Genetics, School of Pharmacy, Iwate Medical University
| | - Shogo Ozawa
- Division of Pharmacodynamics and Molecular Genetics, School of Pharmacy, Iwate Medical University
| | - Jun Terashima
- Division of Pharmacodynamics and Molecular Genetics, School of Pharmacy, Iwate Medical University
| |
Collapse
|
38
|
In Vitro Pharmacokinetic Behavior of Antiviral 3-Amidinophenylalanine Derivatives in Rat, Dog and Monkey Hepatocytes. Biomedicines 2023; 11:biomedicines11030682. [PMID: 36979660 PMCID: PMC10045298 DOI: 10.3390/biomedicines11030682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
Type II transmembrane serine proteases represent pharmacological targets for blocking entry and spread of influenza or coronaviruses. In this study, the depletion rates of the 3-amidinophenylalanine (3-APhA)-derived matriptase/TMPRSS2 inhibitors MI-463, MI-472, MI-485 or MI-1900 were determined by LC-MS/MS measurements over a period of 300 min using suspensions of rat, dog and cynomolgus monkey primary hepatocytes. From these in vitro pharmacokinetic (PK) experiments, intrinsic clearance values (Clint) were evaluated, and in vivo pharmacokinetic parameters (hepatic clearance, hepatic extraction ratio and bioavailability) were predicted. It was found that rat hepatocytes were the most active in the metabolism of 3-APhA derivatives (Clint 31.9–37.8 mL/min/kg), whereas dog and monkey cells displayed somewhat lower clearance of these compounds (Clint 6.6–26.7 mL/min/kg). These data support elucidation of important PK properties of anti-TMPRSS2/anti-matriptase 3-APhAs using mammalian hepatocyte models and thus contribute to the optimization of lead compounds.
Collapse
|
39
|
Huang YL, De Gregorio C, Silva V, Elorza ÁA, Léniz P, Aliaga-Tobar V, Maracaja-Coutinho V, Budini M, Ezquer F, Ezquer M. Administration of Secretome Derived from Human Mesenchymal Stem Cells Induces Hepatoprotective Effects in Models of Idiosyncratic Drug-Induced Liver Injury Caused by Amiodarone or Tamoxifen. Cells 2023; 12:cells12040636. [PMID: 36831304 PMCID: PMC9954258 DOI: 10.3390/cells12040636] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/19/2023] [Accepted: 02/07/2023] [Indexed: 02/18/2023] Open
Abstract
Drug-induced liver injury (DILI) is one of the leading causes of acute liver injury. While many factors may contribute to the susceptibility to DILI, obese patients with hepatic steatosis are particularly prone to suffer DILI. The secretome derived from mesenchymal stem cell has been shown to have hepatoprotective effects in diverse in vitro and in vivo models. In this study, we evaluate whether MSC secretome could improve DILI mediated by amiodarone (AMI) or tamoxifen (TMX). Hepatic HepG2 and HepaRG cells were incubated with AMI or TMX, alone or with the secretome of MSCs obtained from human adipose tissue. These studies demonstrate that coincubation of AMI or TMX with MSC secretome increases cell viability, prevents the activation of apoptosis pathways, and stimulates the expression of priming phase genes, leading to higher proliferation rates. As proof of concept, in a C57BL/6 mouse model of hepatic steatosis and chronic exposure to AMI, the MSC secretome was administered endovenously. In this study, liver injury was significantly attenuated, with a decrease in cell infiltration and stimulation of the regenerative response. The present results indicate that MSC secretome administration has the potential to be an adjunctive cell-free therapy to prevent liver failure derived from DILI caused by TMX or AMI.
Collapse
Affiliation(s)
- Ya-Lin Huang
- Centro de Medicina Regenerativa, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile
| | - Cristian De Gregorio
- Centro de Medicina Regenerativa, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile
| | - Verónica Silva
- Centro de Medicina Regenerativa, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile
| | - Álvaro A. Elorza
- Instituto de Ciencias Biomédicas, Facultad de Medicina y Ciencias de la Vida, Universidad Andres Bello, Santiago 7610658, Chile
| | - Patricio Léniz
- Unidad de Cirugía Plástica, Reparadora y Estética, Clínica Alemana, Santiago 7610658, Chile
| | - Víctor Aliaga-Tobar
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 7610658, Chile
- Centro de Modelamiento Molecular, Biofísica y Bioinformática (CM2B2), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 7610658, Chile
- Laboratorio de Bioingeniería, Instituto de Ciencias de la Ingeniería, Universidad de O’Higgins, Rancagua 7610658, Chile
| | - Vinicius Maracaja-Coutinho
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 7610658, Chile
- Centro de Modelamiento Molecular, Biofísica y Bioinformática (CM2B2), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 7610658, Chile
| | - Mauricio Budini
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 7610658, Chile
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile
- Correspondence: (F.E.); (M.E.); Tel.: +56-990-699-272 (F.E.); +56-976-629-880 (M.E.)
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile
- Correspondence: (F.E.); (M.E.); Tel.: +56-990-699-272 (F.E.); +56-976-629-880 (M.E.)
| |
Collapse
|
40
|
Sugahara G, Ishida Y, Lee JJ, Li M, Tanaka Y, Eoh H, Higuchi Y, Saito T. Long-term cell fate and functional maintenance of human hepatocyte through stepwise culture configuration. FASEB J 2023; 37:e22750. [PMID: 36607308 PMCID: PMC9830592 DOI: 10.1096/fj.202201292rr] [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: 08/10/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 01/07/2023]
Abstract
Human hepatocyte culture system represents by far the most physiologically relevant model for our understanding of liver biology and diseases; however, its versatility has been limited due to the rapid and progressive loss of genuine characteristics, indicating the inadequacy of in vitro milieu for fate maintenance. This study, therefore, is designed to define environmental requirements necessary to sustain the homeostasis of terminally differentiated hepatocytes. Our study reveals that the supplementation of dimethyl sulfoxide (DMSO) is indispensable in mitigating fate deterioration and promoting adaptation to the in vitro environment, resulting in the restoration of tight cell-cell contact, cellular architecture, and polarity. The morphological recovery was overall accompanied by the restoration of hepatocyte marker gene expression, highlighting the interdependence between the cellular architecture and the maintenance of cell fate. However, beyond the recovery phase culture, DMSO supplementation is deemed detrimental due to the potent inhibitory effect on a multitude of hepatocyte functionalities while its withdrawal results in the loss of cell fate. In search of DMSO substitute, our screening of organic substances led to the identification of dimethyl sulfone (DMSO2), which supports the long-term maintenance of proper morphology, marker gene expression, and hepatocytic functions. Moreover, hepatocytes maintained DMSO2 exhibited clinically relevant toxicity in response to prolonged exposure to xenobiotics as well as alcohol. These observations suggest that the stepwise culture configuration consisting of the consecutive supplementation of DMSO and DMSO2 confers the microenvironment essential for the fate and functional maintenance of terminally differentiated human hepatocytes.
Collapse
Affiliation(s)
- Go Sugahara
- Department of Medicine, Division of Gastrointestinal and Liver DiseasesUniversity of Southern California, Keck School of MedicineLos AngelesCaliforniaUSA
- Research and Development DepartmentPhoenixBio, Co., LtdHiroshimaJapan
| | - Yuji Ishida
- Department of Medicine, Division of Gastrointestinal and Liver DiseasesUniversity of Southern California, Keck School of MedicineLos AngelesCaliforniaUSA
- Research and Development DepartmentPhoenixBio, Co., LtdHiroshimaJapan
| | - Jae Jin Lee
- Department of Molecular Microbiology & ImmunologyUniversity of Southern California, Keck School of MedicineLos AngelesCaliforniaUSA
| | - Meng Li
- Bioinformatics Service ProgramUniversity of Southern California, Norris Medical LibraryLos AngelesCaliforniaUSA
| | - Yasuhito Tanaka
- Department of Gastroenterology and Hepatology, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Hyungjin Eoh
- Department of Molecular Microbiology & ImmunologyUniversity of Southern California, Keck School of MedicineLos AngelesCaliforniaUSA
| | - Yusuke Higuchi
- Department of Molecular MedicineBeckman Research Institute of City of HopeDuarteCaliforniaUSA
| | - Takeshi Saito
- Department of Medicine, Division of Gastrointestinal and Liver DiseasesUniversity of Southern California, Keck School of MedicineLos AngelesCaliforniaUSA
- Department of Molecular Microbiology & ImmunologyUniversity of Southern California, Keck School of MedicineLos AngelesCaliforniaUSA
- USC Research Center for Liver DiseasesLos AngelesCaliforniaUSA
| |
Collapse
|
41
|
Tian T, Liu J, Zhu H. Organ Chips and Visualization of Biological Systems. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1199:155-183. [PMID: 37460731 DOI: 10.1007/978-981-32-9902-3_8] [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: 07/20/2023]
Abstract
Organ-on-a-chip (OOC) is an emerging frontier cross-cutting science and technology developed in the past 10 years. It was first proposed by the Wyss Institute for Biologically Inspired Engineering of Harvard Medical School. It consists of a transparent flexible polymer the size of a computer memory stick, with hollow microfluidic channels lined with living human cells. Researchers used bionics methods to simulate the microenvironment of human cells on microfluidic chips, so as to realize the basic physiological functions of corresponding tissues and organs in vitro. Transparent chip materials can perform real-time visualization and high-resolution analysis of various human life processes in a way that is impossible in animal models, so as to better reproduce the microenvironment of human tissue and simulate biological systems in vitro to observe drug metabolism and other life processes. It provides innovative research systems and system solutions for in vitro bionics of biological systems. It also has gradually become a new tool for disease mechanism research and new drug development. In this chapter, we will take the current research mature single-organ-on-a-chip and multi-organ human-on-a-chip as examples; give an overview of the research background and underlying technologies in this field, especially the application of in vitro bionic models in visualized medicine; and look forward to the foreseeable future development prospects after the integration of organ-on-chip and organoid technology.
Collapse
Affiliation(s)
- Tian Tian
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.
| | - Jun Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - He Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
42
|
Cell Dome as an Evaluation Platform for Organized HepG2 Cells. Cells 2022; 12:cells12010069. [PMID: 36611862 PMCID: PMC9818560 DOI: 10.3390/cells12010069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/06/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Human-hepatoblastoma-derived cell line, HepG2, has been widely used in liver and liver cancer studies. HepG2 spheroids produced in a three-dimensional (3D) culture system provide a better biological model than cells cultured in a two-dimensional (2D) culture system. Since cells at the center of spheroids exhibit specific behaviors attributed to hypoxic conditions, a 3D cell culture system that allows the observation of such cells using conventional optical or fluorescence microscopes would be useful. In this study, HepG2 cells were cultured in "Cell Dome", a micro-dome in which cells are enclosed in a cavity consisting of a hemispherical hydrogel shell. HepG2 cells formed hemispherical cell aggregates which filled the cavity of Cell Domes on 18 days of culture and the cells could continue to be cultured for 29 days. The cells at the center of hemispherical cell aggregates were observed using a fluorescence microscope. The cells grew in Cell Domes for 18 days exhibited higher Pi-class Glutathione S-Transferase enzymatic activity, hypoxia inducible factor-1α gene expression, and higher tolerance to mitomycin C than those cultured in 2D on tissue culture dishes (* p < 0.05). These results indicate that the center of the glass adhesive surface of hemispherical cell aggregates which is expected to have the similar environment as the center of the spheroids can be directly observed through glass plates. In conclusion, Cell Dome would be useful as an evaluation platform for organized HepG2 cells.
Collapse
|
43
|
DNMT1/PKR double knockdowned HepG2 (HepG2-DP) cells have high hepatic function and differentiation ability. Sci Rep 2022; 12:21173. [PMID: 36476676 PMCID: PMC9729623 DOI: 10.1038/s41598-022-25777-z] [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: 05/31/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
HepG2 cells are widely used as a human hepatocytes model, but their functions, including drug metabolism, are inferior to primary hepatocytes. We previously reported that the hepatic gene expressions in HepG2 cells were upregulated by treatment with zebularine, which is an inhibitor of DNA methylation, through the inhibition of both DNA methyltransferase 1 (DNMT1) and double-stranded RNA-dependent protein kinase (PKR). In this study, we established a new HepG2 cell subline, HepG2-DP cells, by stable double knockdown of DNMT1 and PKR and evaluated its function. Albumin production, expression of CYP1A2 genes, and accumulation of lipid droplets were increased in HepG2-DP cells compared with the original HepG2 cells. Comprehensive gene expression analysis of transcription factors revealed that the expression of important genes for hepatic function, such as HNF1β, HNF4α, ONECUT1, FOXA1, FOXA2, FOXA3, and various nuclear receptors, was upregulated in HepG2-DP cells. These results indicate that the newly established HepG2-DP cells are a highly functional hepatocyte cell line. In addition, we investigated whether HepG2-DP cells are able to mature by differentiation induction, since HepG2 cells are derived from hepatoblastoma. The gene expression of major CYPs and Phase II, III drug-metabolizing enzyme genes was significantly increased in HepG2-DP cells cultured in differentiation induction medium. These results suggest that HepG2-DP cells can be further matured by the induction of differentiation and could therefore be applied to studies of drug metabolism and pharmacokinetics.
Collapse
|
44
|
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
|
45
|
Llabjani V, Siddique MR, Macos A, Abouzid A, Hoti V, Martin FL, Patel II, Raza A. Introducing CELLBLOKS ®: a novel organ-on-a-chip platform allowing a plug-and-play approach towards building organotypic models. IN VITRO MODELS 2022; 1:423-435. [PMID: 39872618 PMCID: PMC11756440 DOI: 10.1007/s44164-022-00027-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 01/30/2025]
Abstract
Human organs are structurally and functionally complex systems. Their function is driven by the interactions between many specialised cell types, which is difficult to unravel on a standard Petri dish format. Conventional "Petri dish" approaches to culturing cells are static and self-limiting. However, current organ-on-a-chip technologies are difficult to use, have a limited throughput and lack compatibility with standard workflow conditions. We developed CELLBLOKS® as a novel "plug-and-play" organ-on-a-chip platform that enables straightforward creation of multiple cell-type organ-specific microenvironments. Herein, we demonstrate its advantages by building a liver model representative of live tissue function. CELLBLOKS® allows one to systematically test and identify various cell combinations that replicate optimal hepatic relevance. The combined interactions of fibroblasts, endothelial cells and hepatocytes were analysed using hepatic biochemistry (CYP3A4 and urea), cellular proliferation indices and transporter activities (albumin). The results demonstrate that optimal liver function can be achieved by exploiting crosstalk in co-culture combinations compared to conventional mono-culture. The optimised CELLBLOKS® liver model was tested to analyse drug-induced liver toxicity using tamoxifen. The data suggests that our CELLBLOKS® liver model is highly sensitive to toxic insult compared to mono-culture liver models. In summary, CELLBLOKS® provides a novel cell culture technology for creating human-relevant organotypic models that are easy and straightforward to establish in laboratory settings. Supplementary Information The online version contains supplementary material available at 10.1007/s44164-022-00027-8.
Collapse
Affiliation(s)
- Valon Llabjani
- REVIVOCELL Limited, Sci-Tech Daresbury, Keckwick Lane, Daresbury, Warrington, WA4 4AD UK
| | - M. R. Siddique
- REVIVOCELL Limited, Sci-Tech Daresbury, Keckwick Lane, Daresbury, Warrington, WA4 4AD UK
| | - Anaïs Macos
- REVIVOCELL Limited, Sci-Tech Daresbury, Keckwick Lane, Daresbury, Warrington, WA4 4AD UK
| | - Afaf Abouzid
- REVIVOCELL Limited, Sci-Tech Daresbury, Keckwick Lane, Daresbury, Warrington, WA4 4AD UK
| | - Valmira Hoti
- REVIVOCELL Limited, Sci-Tech Daresbury, Keckwick Lane, Daresbury, Warrington, WA4 4AD UK
| | - Francis L. Martin
- REVIVOCELL Limited, Sci-Tech Daresbury, Keckwick Lane, Daresbury, Warrington, WA4 4AD UK
| | - Imran I. Patel
- REVIVOCELL Limited, Sci-Tech Daresbury, Keckwick Lane, Daresbury, Warrington, WA4 4AD UK
| | - Ahtasham Raza
- REVIVOCELL Limited, Sci-Tech Daresbury, Keckwick Lane, Daresbury, Warrington, WA4 4AD UK
| |
Collapse
|
46
|
Shedding light on non-alcoholic fatty liver disease: Pathogenesis, molecular mechanisms, models, and emerging therapeutics. Life Sci 2022; 312:121185. [PMID: 36375569 DOI: 10.1016/j.lfs.2022.121185] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disorder globally impacting an estimated 25% of the population associated with severe consequences such as cirrhosis, hepatocellular carcinoma (HCC), and overall mortality. Fatty liver disease is triggered through multiple pathways, but the most prominent cause is either diabetes or obesity, or a combination of both. Therefore, hepatic glucose, insulin and fatty acid signaling becomes a dire need to understand which is well elaborated in this review. This review summarizes the popular two-hit pathogenesis of NAFLD, the molecular mechanisms underlying hepatic insulin resistance. As fatty liver disease gets advanced, it requires in-vitro as well as in-vivo models closer to disease progression in humans for better understanding the pathological state and identifying a novel therapeutic target. This review summarizes in-vitro (2D cell-culture/co-culture, 3D spheroid/organoid/liver-on-a-chip) models as well as in-vivo (genetically/dietary/chemically induced fatty liver disease) research models. Fatty liver disease research has gathered lots of attention recently since there is no FDA approved therapy available so far. However, there have been numerous promising targets to treat fatty liver disease including potential therapeutic targets under clinical trials are listed in this review.
Collapse
|
47
|
Elizondo-Luévano JH, Gomez-Flores R, Verde-Star MJ, Tamez-Guerra P, Romo-Sáenz CI, Chávez-Montes A, Rodríguez-Garza NE, Quintanilla-Licea R. In Vitro Cytotoxic Activity of Methanol Extracts of Selected Medicinal Plants Traditionally Used in Mexico against Human Hepatocellular Carcinoma. PLANTS (BASEL, SWITZERLAND) 2022; 11:plants11212862. [PMID: 36365315 PMCID: PMC9659118 DOI: 10.3390/plants11212862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 05/14/2023]
Abstract
Medicinal plants are traditionally used in Mexico to treat diseases such as cancer. The present study aimed to evaluate the cytotoxic, antioxidant, and anti-hemolytic activity of 15 plants of ethnopharmacological use in Mexico. For this, plant methanol extracts were prepared by the Soxhlet method, after which their cytotoxic activity was evaluated against human hepatocellular carcinoma (HEP-G2) and monkey kidney epithelial (Vero) cells by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction colorimetric assay. The selectivity index (SI) of each extract was then determined by the IC50 ratio of normal to tumor cells. We showed that Ruta chalepensis extract possessed an IC50 of 1.79 µg/mL and 522.08 µg/mL against HEP-G2 and Vero cells, respectively, resulting in an SI of 291.50. Furthermore, antioxidant activity was evaluated by the 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical scavenging technique, where the best antioxidant potential was shown by the Heterotheca inuloides extract (IC50 = 19.24 µg/mL). Furthermore, the hemolytic potential was determined against human erythrocytes, which showed that the extracts with the highest anti-hemolytic activity were Smilax aspera (IC50 = 4.41 µg/mL) and Amphipterygium adstringens (IC50 = 5.35 µg/mL). In conclusion, we observed that R. chalepensis methanol extract possesses cytotoxic activity against HEP-G2 cells, without affecting non-tumorigenic Vero cells. Our results indicated the antitumor potential of medicinal plants used in Mexico.
Collapse
Affiliation(s)
- Joel H. Elizondo-Luévano
- Departamento de Química, Facultad de Ciencias Biológicas (FCB), Universidad Autónoma de Nuevo León (UANL), San Nicolás de los Garza, N.L., Monterrey 66455, Mexico
| | - Ricardo Gomez-Flores
- Departamento de Microbiología e Inmunología, Facultad de Ciencias Biológicas (FCB), Universidad Autónoma de Nuevo León (UANL), San Nicolás de los Garza, N.L., Monterrey 66455, Mexico
- Correspondence: (R.G.-F.); (R.Q.-L.); Tel.: +52-81-8020-7449 (R.G.-F.); +52-81-8376-3668 (ext. 1476) (R.Q.-L.)
| | - María J. Verde-Star
- Departamento de Química, Facultad de Ciencias Biológicas (FCB), Universidad Autónoma de Nuevo León (UANL), San Nicolás de los Garza, N.L., Monterrey 66455, Mexico
| | - Patricia Tamez-Guerra
- Departamento de Microbiología e Inmunología, Facultad de Ciencias Biológicas (FCB), Universidad Autónoma de Nuevo León (UANL), San Nicolás de los Garza, N.L., Monterrey 66455, Mexico
| | - César I. Romo-Sáenz
- Departamento de Microbiología e Inmunología, Facultad de Ciencias Biológicas (FCB), Universidad Autónoma de Nuevo León (UANL), San Nicolás de los Garza, N.L., Monterrey 66455, Mexico
| | - Abelardo Chávez-Montes
- Departamento de Química, Facultad de Ciencias Biológicas (FCB), Universidad Autónoma de Nuevo León (UANL), San Nicolás de los Garza, N.L., Monterrey 66455, Mexico
| | - Nancy E. Rodríguez-Garza
- Departamento de Química, Facultad de Ciencias Biológicas (FCB), Universidad Autónoma de Nuevo León (UANL), San Nicolás de los Garza, N.L., Monterrey 66455, Mexico
| | - Ramiro Quintanilla-Licea
- Departamento de Química, Facultad de Ciencias Biológicas (FCB), Universidad Autónoma de Nuevo León (UANL), San Nicolás de los Garza, N.L., Monterrey 66455, Mexico
- Correspondence: (R.G.-F.); (R.Q.-L.); Tel.: +52-81-8020-7449 (R.G.-F.); +52-81-8376-3668 (ext. 1476) (R.Q.-L.)
| |
Collapse
|
48
|
Mišík M, Nersesyan A, Ferk F, Holzmann K, Krupitza G, Herrera Morales D, Staudinger M, Wultsch G, Knasmueller S. Search for the optimal genotoxicity assay for routine testing of chemicals: Sensitivity and specificity of conventional and new test systems. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2022; 881:503524. [PMID: 36031336 DOI: 10.1016/j.mrgentox.2022.503524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/15/2022] [Accepted: 06/28/2022] [Indexed: 06/15/2023]
Abstract
Many conventional in vitro tests that are currently widely used for routine screening of chemicals have a sensitivity/specificity in the range between 60 % and 80 % for the detection of carcinogens. Most procedures were developed 30-40 years ago. In the last decades several assays became available which are based on the use of metabolically competent cell lines, improvement of the cultivation conditions and development of new endpoints. Validation studies indicate that some of these models may be more reliable for the detection of genotoxicants (i.e. many of them have sensitivity and specificity values between 80 % and 95 %). Therefore, they could replace conventional tests in the future. The bone marrow micronucleus (MN) assay with rodents is at present the most widely used in vivo test. The majority of studies indicate that it detects only 5-6 out of 10 carcinogens while experiments with transgenic rodents and comet assays seem to have a higher predictive value and detect genotoxic carcinogens that are negative in MN experiments. Alternatives to rodent experiments could be MN experiments with hen eggs or their replacement by combinations of new in vitro tests. Examples for promising candidates are ToxTracker, TGx-DDI, multiplex flow cytometry, γH2AX experiments, measurement of p53 activation and MN experiments with metabolically competent human derived liver cells. However, the realization of multicentric collaborative validation studies is mandatory to identify the most reliable tests.
Collapse
Affiliation(s)
- M Mišík
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - A Nersesyan
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - F Ferk
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - K Holzmann
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - G Krupitza
- Department of Pathology, Medical University of Vienna, A-1090 Vienna, Austria
| | - D Herrera Morales
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - M Staudinger
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - G Wultsch
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - S Knasmueller
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria.
| |
Collapse
|
49
|
Hoyle H, Stenger C, Przyborski S. Design considerations of benchtop fluid flow bioreactors for bio-engineered tissue equivalents in vitro. BIOMATERIALS AND BIOSYSTEMS 2022; 8:100063. [PMID: 36824373 PMCID: PMC9934498 DOI: 10.1016/j.bbiosy.2022.100063] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/08/2022] [Accepted: 08/30/2022] [Indexed: 10/14/2022] Open
Abstract
One of the major aims of bio-engineering tissue equivalents in vitro is to create physiologically relevant culture conditions to accurately recreate the cellular microenvironment. This often includes incorporation of factors such as the extracellular matrix, co-culture of multiple cell types and three-dimensional culture techniques. These advanced techniques can recapitulate some of the properties of tissue in vivo, however fluid flow is a key aspect that is often absent. Fluid flow can be introduced into cell and tissue culture using bioreactors, which are becoming increasingly common as we seek to produce increasingly accurate tissue models. Bespoke technology is continuously being developed to tailor systems for specific applications and to allow compatibility with a range of culture techniques. For effective perfusion of a tissue culture many parameters can be controlled, ranging from impacts of the fluid flow such as increased shear stress and mass transport, to potentially unwanted side effects such as temperature fluctuations. A thorough understanding of these properties and their implications on the culture model can aid with a more accurate interpretation of results. Improved and more complete characterisation of bioreactor properties will also lead to greater accuracy when reporting culture conditions in protocols, aiding experimental reproducibility, and allowing more precise comparison of results between different systems. In this review we provide an analysis of the different factors involved in the development of benchtop flow bioreactors and their potential biological impacts across a range of applications.
Collapse
Key Words
- 3D, three-dimensional
- ABS, acrylonitrile butadiene styrene
- ALI, air-liquid interface
- Bioreactors
- CFD, computational fluid dynamics
- Cell culture
- ECM, extracellular matrix
- FDM, fused deposition modelling
- Fluid flow
- PC, polycarbonate
- PET, polyethylene terephthalate
- PLA, polylactic acid
- PTFE, polytetrafluoroethylene
- SLA, stereolithography
- Tissue engineering
- UL, unstirred layer
- UV, ultraviolet light
Collapse
Affiliation(s)
- H.W. Hoyle
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK
| | - C.M.L. Stenger
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK
| | - S.A. Przyborski
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK,NETPark Incubator, Reprocell Europe Ltd., Thomas Wright Way, Sedgefield TS21 3FD, UK,Corresponding author at: Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK.
| |
Collapse
|
50
|
Agulló V, Moreno DA, Domínguez‐Perles R, García‐Viguera C. Contribution of the diverse experimental models to unravelling the biological scope of dietary (poly)phenols. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:3940-3951. [PMID: 35285937 PMCID: PMC9321600 DOI: 10.1002/jsfa.11865] [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] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/07/2022] [Accepted: 03/14/2022] [Indexed: 06/14/2023]
Abstract
The health benefits associated with (poly)phenols need to be supported by robust and insightful information on their biological effects. The use of in vitro, ex vivo, and in vivo models is crucial to demonstrate functionalities in specific targets. In this regard, bioaccessibility, bioavailability, and tissue/organ distribution need to be fully understood and established. In addition, the structure-function relationships, concerning both descriptive and mechanistic information, between specific compounds and therapeutic objectives, need to be supported by results obtained from in vivo studies. Nevertheless, these studies are not always possible or have some limitations, particularly concerning the mechanistic information explaining the health benefits provided that should be covered with complementary experimental models. Based on these premises, this review aims to overview the contribution of the separate experimental approaches to gain insights into the bioaccessibility, bioavailability, and bioactivity of (poly)phenols. To achieve this objective, recent evidence available on the linkage of healthy/functional foods with the incidence of non-communicable pathologies is presented. The different experimental approaches provide complementary information that allows advances to be applied to the knowledge gained on the functional properties and mechanistic facts responsible for the health attributions of polyphenols. © 2022 The Authors. Journal of The Science of Food and Agriculture published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
Affiliation(s)
- Vicente Agulló
- Phytochemistry and Healthy Food Lab (LabFAS)Department of Food Science and Technology, CEBAS‐CSICMurcia30100Spain
| | - Diego A Moreno
- Phytochemistry and Healthy Food Lab (LabFAS)Department of Food Science and Technology, CEBAS‐CSICMurcia30100Spain
| | - Raúl Domínguez‐Perles
- Phytochemistry and Healthy Food Lab (LabFAS)Department of Food Science and Technology, CEBAS‐CSICMurcia30100Spain
| | - Cristina García‐Viguera
- Phytochemistry and Healthy Food Lab (LabFAS)Department of Food Science and Technology, CEBAS‐CSICMurcia30100Spain
| |
Collapse
|