1
|
Hu Y, Geng Q, Wang L, Wang Y, Huang C, Fan Z, Kong D. Research progress and application of liver organoids for disease modeling and regenerative therapy. J Mol Med (Berl) 2024; 102:859-874. [PMID: 38802517 PMCID: PMC11213763 DOI: 10.1007/s00109-024-02455-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 04/19/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024]
Abstract
The liver is a major metabolic organ of the human body and has a high incidence of diseases. In recent years, the annual incidence of liver disease has increased, seriously endangering human life and health. The study of the occurrence and development mechanism of liver diseases, discovery of new therapeutic targets, and establishment of new methods of medical treatment are major issues related to the national economy and people's livelihood. The development of stable and effective research models is expected to provide new insights into the pathogenesis of liver diseases and the search for more effective treatment options. Organoid technology is a new in vitro culture system, and organoids constructed by human cells can simulate the morphological structure, gene expression, and glucose and lipid metabolism of organs in vivo, providing a new model for related research on liver diseases. This paper reviews the latest research progress on liver organoids from the establishment of cell sources and application of liver organoids and discusses their application potential in the field of liver disease research.
Collapse
Affiliation(s)
- Yang Hu
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, China
| | - Qiao Geng
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
| | - Lu Wang
- Department of Angioenterology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, 157 Daming Avenue, Nanjing, 210022, Jiangsu, China
| | - Yi Wang
- Department of Angioenterology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, 157 Daming Avenue, Nanjing, 210022, Jiangsu, China
| | - Chuyue Huang
- Department of Angioenterology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, 157 Daming Avenue, Nanjing, 210022, Jiangsu, China
| | - Zhimin Fan
- Department of Angioenterology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, 157 Daming Avenue, Nanjing, 210022, Jiangsu, China.
| | - Desong Kong
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, China.
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China.
| |
Collapse
|
2
|
Sun B, Liang Z, Wang Y, Yu Y, Zhou X, Geng X, Li B. A 3D spheroid model of quadruple cell co-culture with improved liver functions for hepatotoxicity prediction. Toxicology 2024; 505:153829. [PMID: 38740170 DOI: 10.1016/j.tox.2024.153829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/04/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
Drug-induced liver injury (DILI) is one of the major concerns during drug development. Wide acceptance of the 3 R principles and the innovation of in-vitro techniques have introduced various novel model options, among which the three-dimensional (3D) cell spheroid cultures have shown a promising prospect in DILI prediction. The present study developed a 3D quadruple cell co-culture liver spheroid model for DILI prediction via self-assembly. Induction by phorbol 12-myristate 13-acetate at the concentration of 15.42 ng/mL for 48 hours with a following 24-hour rest period was used for THP-1 cell differentiation, resulting in credible macrophagic phenotypes. HepG2 cells, PUMC-HUVEC-T1 cells, THP-1-originated macrophages, and human hepatic stellate cells were selected as the components, which exhibited adaptability in the designated spheroid culture conditions. Following establishment, the characterization demonstrated the competence of the model in long-term stability reflected by the maintenance of morphology, viability, cellular integration, and cell-cell junctions for at least six days, as well as the reliable liver-specific functions including superior albumin and urea secretion, improved drug metabolic enzyme expression and CYP3A4 activity, and the expression of MRP2, BSEP, and P-GP accompanied by the bile acid efflux transport function. In the comparative testing using 22 DILI-positive and 5 DILI-negative compounds among the novel 3D co-culture model, 3D HepG2 spheroids, and 2D HepG2 monolayers, the 3D culture method significantly enhanced the model sensitivity to compound cytotoxicity compared to the 2D form. The novel co-culture liver spheroid model exhibited higher overall predictive power with margin of safety as the classifying tool. In addition, the non-parenchymal cell components could amplify the toxicity of isoniazid in the 3D model, suggesting their potential mediating role in immune-mediated toxicity. The proof-of-concept experiments demonstrated the capability of the model in replicating drug-induced lipid dysregulation, bile acid efflux inhibition, and α-SMA upregulation, which are the key features of liver steatosis and phospholipidosis, cholestasis, and fibrosis, respectively. Overall, the novel 3D quadruple cell co-culture spheroid model is a reliable and readily available option for DILI prediction.
Collapse
Affiliation(s)
- Baiyang Sun
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Nonclinical Safety Evaluation of Drugs, Beijing 100176, China
| | - Zihe Liang
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Nonclinical Safety Evaluation of Drugs, Beijing 100176, China
| | - Yupeng Wang
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Nonclinical Safety Evaluation of Drugs, Beijing 100176, China
| | - Yue Yu
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Nonclinical Safety Evaluation of Drugs, Beijing 100176, China
| | - Xiaobing Zhou
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Nonclinical Safety Evaluation of Drugs, Beijing 100176, China
| | - Xingchao Geng
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Nonclinical Safety Evaluation of Drugs, Beijing 100176, China.
| | - Bo Li
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; National Institutes for Food and Drug Control, Beijing 102629, China.
| |
Collapse
|
3
|
Seo JE, Le Y, Revollo J, Miranda-Colon J, Xu H, McKinzie P, Mei N, Chen T, Heflich RH, Zhou T, Robison T, Bonzo JA, Guo X. Evaluating the mutagenicity of N-nitrosodimethylamine in 2D and 3D HepaRG cell cultures using error-corrected next generation sequencing. Arch Toxicol 2024; 98:1919-1935. [PMID: 38584193 PMCID: PMC11106104 DOI: 10.1007/s00204-024-03731-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/07/2024] [Indexed: 04/09/2024]
Abstract
Human liver-derived metabolically competent HepaRG cells have been successfully employed in both two-dimensional (2D) and 3D spheroid formats for performing the comet assay and micronucleus (MN) assay. In the present study, we have investigated expanding the genotoxicity endpoints evaluated in HepaRG cells by detecting mutagenesis using two error-corrected next generation sequencing (ecNGS) technologies, Duplex Sequencing (DS) and High-Fidelity (HiFi) Sequencing. Both HepaRG 2D cells and 3D spheroids were exposed for 72 h to N-nitrosodimethylamine (NDMA), followed by an additional incubation for the fixation of induced mutations. NDMA-induced DNA damage, chromosomal damage, and mutagenesis were determined using the comet assay, MN assay, and ecNGS, respectively. The 72-h treatment with NDMA resulted in concentration-dependent increases in cytotoxicity, DNA damage, MN formation, and mutation frequency in both 2D and 3D cultures, with greater responses observed in the 3D spheroids compared to 2D cells. The mutational spectrum analysis showed that NDMA induced predominantly A:T → G:C transitions, along with a lower frequency of G:C → A:T transitions, and exhibited a different trinucleotide signature relative to the negative control. These results demonstrate that the HepaRG 2D cells and 3D spheroid models can be used for mutagenesis assessment using both DS and HiFi Sequencing, with the caveat that severe cytotoxic concentrations should be avoided when conducting DS. With further validation, the HepaRG 2D/3D system may become a powerful human-based metabolically competent platform for genotoxicity testing.
Collapse
Affiliation(s)
- Ji-Eun Seo
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Yuan Le
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Javier Revollo
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Jaime Miranda-Colon
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Hannah Xu
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Page McKinzie
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Nan Mei
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Tao Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Robert H Heflich
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Tong Zhou
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Rockville, MD, 20855, USA
| | - Timothy Robison
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Jessica A Bonzo
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Xiaoqing Guo
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA.
| |
Collapse
|
4
|
Ortega-Vallbona R, Méndez R, Tolosa L, Escher SE, Castell JV, Gozalbes R, Serrano-Candelas E. Uncovering the toxicity mechanisms of a series of carboxylic acids in liver cells through computational and experimental approaches. Toxicology 2024; 504:153764. [PMID: 38428665 DOI: 10.1016/j.tox.2024.153764] [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: 12/22/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024]
Abstract
Hepatotoxicity poses a significant concern in drug design due to the potential liver damage that can be caused by new drugs. Among common manifestations of hepatotoxic damage is lipid accumulation in hepatic tissue, resulting in liver steatosis or phospholipidosis. Carboxylic derivatives are prone to interfere with fatty acid metabolism and cause lipid accumulation in hepatocytes. This study investigates the toxic behaviour of 24 structurally related carboxylic acids in hepatocytes, specifically their ability to cause accumulation of fatty acids and phospholipids. Using high-content screening (HCS) assays, we identified two distinct lipid accumulation patterns. Subsequently, we developed structure-activity relationship (SAR) and quantitative structure-activity relationship (QSAR) models to determine relevant molecular substructures and descriptors contributing to these adverse effects. Additionally, we calculated physicochemical properties associated with lipid accumulation in hepatocytes and examined their correlation with our chemical structure characteristics. To assess the applicability of our findings to a wide range of chemical compounds, we employed two external datasets to evaluate the distribution of our QSAR descriptors. Our study highlights the significance of subtle molecular structural variations in triggering hepatotoxicity, such as the presence of nitrogen or the specific arrangement of substitutions within the carbon chain. By employing our comprehensive approach, we pinpointed specific molecules and elucidated their mechanisms of toxicity, thus offering valuable insights to guide future toxicology investigations.
Collapse
Affiliation(s)
- Rita Ortega-Vallbona
- ProtoQSAR SL., Centro Europeo de Empresas e Innovación (CEEI), Parque Tecnológico de Valencia, Av. Benjamín Franklin, 12, Valencia, Paterna 46980, Spain
| | - Rebeca Méndez
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, Valencia 46026, Spain
| | - Laia Tolosa
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, Valencia 46026, Spain; Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), ISCIII, C/ Monforte de Lemos, Madrid 28029, Spain
| | - Sylvia E Escher
- Fraunhofer ITEM, Chemical Safety and Toxicology, Nikolai-Fuchs-Straße 1, Hannover 30625, Germany
| | - José V Castell
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av Fernando Abril Martorell 106, Valencia 46026, Spain; Departamento de Bioquímica y Biología Molecular. Facultad de Medicina, Universidad de Valencia, Av. de Blasco Ibáñez, 15, Valencia 46010, Spain; CIBEREHD, ISCIII, C/ Monforte de Lemos, Madrid 28029, Spain.
| | - Rafael Gozalbes
- ProtoQSAR SL., Centro Europeo de Empresas e Innovación (CEEI), Parque Tecnológico de Valencia, Av. Benjamín Franklin, 12, Valencia, Paterna 46980, Spain; Moldrug AI Systems SL, c/Olimpia Arozena Torres 45, Valencia 46018, Spain
| | - Eva Serrano-Candelas
- ProtoQSAR SL., Centro Europeo de Empresas e Innovación (CEEI), Parque Tecnológico de Valencia, Av. Benjamín Franklin, 12, Valencia, Paterna 46980, Spain
| |
Collapse
|
5
|
Haller K, Doß S, Sauer M. In Vitro Hepatotoxicity of Routinely Used Opioids and Sedative Drugs. Curr Issues Mol Biol 2024; 46:3022-3038. [PMID: 38666919 PMCID: PMC11049542 DOI: 10.3390/cimb46040189] [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: 01/15/2024] [Revised: 03/17/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
A hepatocyte cell line was used to determine the hepatotoxicity of sedatives and opioids, as the hepatotoxicity of these drugs has not yet been well characterized. This might pose a threat, especially to critically ill patients, as they often receive high cumulative doses for daily analgosedation and often already have impaired liver function due to an underlying disease or complications during treatment. A well-established biosensor based on HepG2/C3A cells was used for the determination of the hepatotoxicity of commonly used sedatives and opioids in the intensive care setting (midazolam, propofol, s-ketamin, thiopental, fentanyl, remifentanil, and sufentanil). The incubation time was 2 × 3 days with clinically relevant (Cmax) and higher concentrations (C5× and C10×) of each drug in cell culture medium or human plasma. Afterward, we measured the cell count, vitality, lactate dehydrogenase (LDH), mitochondrial dehydrogenase activity, cytochrome P 450 1A2 (CYP1A2), and albumin synthesis. All tested substances reduced the viability of hepatocyte cells, but sufentanil and remifentanil showed more pronounced effects. The cell count was diminished by sufentanil in both the medium and plasma and by remifentanil only in plasma. Sufentanil and remifentanil also led to higher values of LDH in the cell culture supernatant. A reduction of mitochondrial dehydrogenase activity was seen with the use of midazolam and s-ketamine. Microalbumin synthesis was reduced in plasma after its incubation with higher concentrations of sufentanil and remifentanil. Remifentanil and s-ketamine reduced CYP1A2 activity, while propofol and thiopental increased it. Our findings suggest that none of the tested sedatives and opioids have pronounced hepatotoxicity. Sufentanil, remifentanil, and s-ketamine showed moderate hepatotoxic effects in vitro. These drugs should be given with caution to patients vulnerable to hepatotoxic drugs, e.g., patients with pre-existing liver disease or liver impairment as part of their underlying disease (e.g., hypoxic hepatitis or cholestatic liver dysfunction in sepsis). Further studies are indicated for this topic, which may use more complex cell culture models and global pharmacovigilance reports, addressing the limitation of the used cell model: HepG2/C3A cells have a lower metabolic capacity due to their low levels of CYP enzymes compared to primary hepatocytes. However, while the test model is suitable for parental substances, it is not for toxicity testing of metabolites.
Collapse
Affiliation(s)
- Katharina Haller
- Department of Anesthesiology and Intensive Care Medicine, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany;
| | - Sandra Doß
- Department Extracorporeal Therapy Systems (EXTHER), Fraunhofer Institute for Cell Therapy and Immunology, Schillingallee 68, 18057 Rostock, Germany;
| | - Martin Sauer
- Department Extracorporeal Therapy Systems (EXTHER), Fraunhofer Institute for Cell Therapy and Immunology, 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
|
6
|
Kamat V, Grumbine MK, Bao K, Mokate K, Khalil G, Cook D, Clearwater B, Hirst R, Harman J, Boeck M, Fu Z, Smith LEH, Goswami M, Wubben TJ, Walker EM, Zhu J, Soleimanpour SA, Scarlett JM, Robbings BM, Hass D, Hurley JB, Sweet IR. A versatile pumpless multi-channel fluidics system for maintenance and real-time functional assessment of tissue and cells. CELL REPORTS METHODS 2023; 3:100642. [PMID: 37963464 PMCID: PMC10694526 DOI: 10.1016/j.crmeth.2023.100642] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/24/2023] [Accepted: 10/20/2023] [Indexed: 11/16/2023]
Abstract
To address the needs of the life sciences community and the pharmaceutical industry in pre-clinical drug development to both maintain and continuously assess tissue metabolism and function with simple and rapid systems, we improved on the initial BaroFuse to develop it into a fully functional, pumpless, scalable multi-channel fluidics instrument that continuously measures changes in oxygen consumption and other endpoints in response to test compounds. We and several other laboratories assessed it with a wide range of tissue types including retina, pancreatic islets, liver, and hypothalamus with both aqueous and gaseous test compounds. The setup time was less than an hour for all collaborating groups, and there was close agreement between data obtained from the different laboratories. This easy-to-use system reliably generates real-time metabolic and functional data from tissue and cells in response to test compounds that will address a critical need in basic and applied research.
Collapse
Affiliation(s)
- Varun Kamat
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA
| | | | - Khang Bao
- EnTox Sciences, Inc., Mercer Island, WA 98040, USA
| | - Kedar Mokate
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA
| | - Gamal Khalil
- EnTox Sciences, Inc., Mercer Island, WA 98040, USA
| | - Daniel Cook
- EnTox Sciences, Inc., Mercer Island, WA 98040, USA
| | | | - Richard Hirst
- Technical Assembly Service Corporation, Seattle, WA 98109, USA
| | - Jarrod Harman
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Myriam Boeck
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Moloy Goswami
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Thomas J Wubben
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Emily M Walker
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 98195, USA
| | - Jie Zhu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 98195, USA
| | - Scott A Soleimanpour
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 98195, USA
| | - Jarrad M Scarlett
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA 98145, USA
| | - Brian M Robbings
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Biochemistry, University of Washington, Seattle, WA 98109, USA
| | - Daniel Hass
- Department of Biochemistry, University of Washington, Seattle, WA 98109, USA
| | - James B Hurley
- Department of Biochemistry, University of Washington, Seattle, WA 98109, USA
| | - Ian R Sweet
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA 98109, USA; EnTox Sciences, Inc., Mercer Island, WA 98040, USA.
| |
Collapse
|
7
|
Roser LA, Luckhardt S, Ziegler N, Thomas D, Wagner PV, Damm G, Scheffschick A, Hewitt P, Parnham MJ, Schiffmann S. Immuno-inflammatory in vitro hepatotoxicity models to assess side effects of biologicals exemplified by aldesleukin. Front Immunol 2023; 14:1275368. [PMID: 38045689 PMCID: PMC10693457 DOI: 10.3389/fimmu.2023.1275368] [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: 08/09/2023] [Accepted: 10/27/2023] [Indexed: 12/05/2023] Open
Abstract
Introduction Hepatotoxicity induced by immunotherapeutics is an appearing cause for immune-mediated drug-induced liver injury. Such immuno-toxic mechanisms are difficult to assess using current preclinical models and the incidence is too low to detect in clinical trials. As hepatotoxicity is a frequent reason for post-authorisation drug withdrawal, there is an urgent need for immuno-inflammatory in vitro models to assess the hepatotoxic potential of immuno-modulatory drug candidates. We developed several immuno-inflammatory hepatotoxicity test systems based on recombinant human interleukin-2 (aldesleukin). Methods Co-culture models of primary human CD8+ T cells or NK cells with the hepatocyte cell line HepaRG were established and validated with primary human hepatocytes (PHHs). Subsequently, the HepaRG model was refined by increasing complexity by inclusion of monocyte-derived macrophages (MdMs). The main readouts were cytotoxicity, inflammatory mediator release, surface marker expression and specific hepatocyte functions. Results We identified CD8+ T cells as possible mediators of aldesleukin-mediated hepatotoxicity, with MdMs being implicated in increased aldesleukin-induced inflammatory effects. In co-cultures of CD8+ T cells with MdMs and HepaRG cells, cytotoxicity was induced at intermediate/high aldesleukin concentrations and perforin was upregulated. A pro-inflammatory milieu was created measured by interleukin-6 (IL-6), c-reactive protein (CRP), interferon gamma (IFN-γ), and monocyte chemoattractant protein-1 (MCP-1) increase. NK cells responded to aldesleukin, however, only minor aldesleukin-induced cytotoxic effects were measured in co-cultures. Results obtained with HepaRG cells and with PHHs were comparable, especially regarding cytotoxicity, but high inter-donor variations limited meaningfulness of the PHH model. Discussion The in vitro test systems developed contribute to the understanding of potential key mechanisms in aldesleukin-mediated hepatotoxicity. In addition, they may aid assessment of immune-mediated hepatotoxicity during the development of novel immunotherapeutics.
Collapse
Affiliation(s)
- Luise A. Roser
- Department of Preclinical Research, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
| | - Sonja Luckhardt
- Department of Preclinical Research, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
| | - Nicole Ziegler
- Department of Preclinical Research, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
| | - Dominique Thomas
- Department of Preclinical Research, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
- pharmazentrum frankfurt/ZAFES, Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Pia Viktoria Wagner
- Department of Preclinical Research, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Leipzig, Germany
| | - Andrea Scheffschick
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Leipzig, Germany
| | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany
| | - Michael J. Parnham
- Department of Preclinical Research, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
| | - Susanne Schiffmann
- Department of Preclinical Research, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
- pharmazentrum frankfurt/ZAFES, Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Frankfurt am Main, Germany
| |
Collapse
|
8
|
Dager WE, Trujillo TC, Gilbert BW. Approaches to Precision-based Anticoagulation management in the critically Ill. Pharmacotherapy 2023; 43:1221-1236. [PMID: 37604646 DOI: 10.1002/phar.2868] [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: 01/30/2023] [Revised: 05/31/2023] [Accepted: 06/07/2023] [Indexed: 08/23/2023]
Abstract
Anticoagulant therapy is commonly associated with a high incidence of avoidable adverse events, especially in the acute care setting. This has led to several initiatives by key national health care stakeholders, including specific attention to The Joint Commission's National Patient Safety Goals, to improve anticoagulation management. The subject of special populations has long been identified as challenging by clinicians with the use of anticoagulants. This is driven in part by numerous variables that can contribute to hard outcomes such as bleeding, thrombosis, length of stay, hospital re-admission, morbidity, and mortality. Despite the notable effort to improve the use of anticoagulants with numerous clinical trials, guidelines, guidance statements, and other sources of published evidence, notable difficulties continue to challenge practitioners in managing this class of medications. This is especially the case with very diverse critically ill populations where countless variables exist, many of which were never explored in trials or have historically been frequently excluded. Trials evaluating anticoagulation therapy often can only account for small portions of variables that may affect thrombosis and hemostasis, and study methods often do not reflect the constantly changing dynamic conditions seen in unique critically ill patients. Clinicians providing care to the numerous critically ill populations are faced with conditions that lead to relatively small therapeutic windows, which makes designing safe optimal anticoagulation management plans difficult when dealing with complex patients and mechanical support devices. The approach to crafting a successful management plan for anticoagulant therapy must incorporate the numerous variables that are continuously assessed and revised during the patient's time in the intensive care unit. We explore considerations and approaches when developing, assessing, and implementing an individualized or precision-based management plan that involves the use of anticoagulants in the critically ill. The skills and thought process provided will assist clinicians in managing this unique, variable, and challenging population.
Collapse
Affiliation(s)
- William E Dager
- University of California, Davis Medical Center, Sacramento, California, USA
- University of California San Francisco School of Pharmacy, San Francisco, California, USA
- University of California School of Medicine, Sacramento, California, USA
| | - Toby C Trujillo
- University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado, USA
- Anticoagulation/Cardiology, University of Colorado Hospital, Aurora, Colorado, USA
| | | |
Collapse
|
9
|
Mahapatra D, Maronpot R. Translational Relevance of Rodent Models to Predict Human Liver Disease. Toxicol Pathol 2023; 51:482-486. [PMID: 38494947 DOI: 10.1177/01926233241230543] [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: 03/19/2024]
Abstract
Animals models are essential to understand the complex pathobiology of human diseases. George Box's aphorism based on statistics "All models are wrong, but some are useful" certainly applies to animal models of disease. In this session, the translational relevance of various animal models applicable to human liver disease was explored starting with a historic overview of the rodent cancer bioassay with emphasis on hepatocarcinogenesis from early work at the National Cancer Institute, refinement by the National Toxicology Program and contemporary efforts to identify potential mechanisms and their relevance to human cancer risk. Subsequently, recently elucidated understanding of the molecular drivers and signaling mechanisms of liver pathophysiology and liver cancer, including factors associated with liver regeneration, metabolic hepatocellular zonation, and the role of macrophages and their crosstalk with stellate cells in understanding human liver disease was discussed. Next, our contemporary understanding of the role of nuclear receptors in hepatic homeostasis and drug response highlighting nuclear receptor activation and crosstalk in modulating biological responses associated with liver damage and neoplastic response were discussed. Finally, an overview and translational relevance of different drug-induced liver injury (DILI) rodent model systems focused on pathology and mechanisms with commentary on current relevant Food and Drug Administration (FDA) perspective were summarized with closing remarks.
Collapse
|
10
|
Gould S, Templin MV. Off target toxicities and links with physicochemical properties of medicinal products, including antibiotics, oligonucleotides, lipid nanoparticles (with cationic and/or anionic charges). Data review suggests an emerging pattern. Toxicol Lett 2023; 384:14-29. [PMID: 37454775 DOI: 10.1016/j.toxlet.2023.07.011] [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: 01/10/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Toxicology is an essential part of any drug development plan. Circumnavigating the risk of failure because of a toxicity issue can be a challenge, and failure in late development is extremely costly. To identify potential risks, it requires more than just understanding the biological target. The toxicologist needs to consider a compound's structure, it's physicochemical properties (including the impact of the overall formulation), as well as the biological target (e.g., receptor interactions). Understanding the impact of the physicochemical properties can be used to predict potential toxicities in advance by incorporating key endpoints in early screening strategies and/or used to compare toxicity profiles across lead candidates. This review discussed the risks of off-target and/or non-specific toxicities that may be associated with the physicochemical properties of compounds, especially those carrying dominant positive or negative charges, including amphiphilic small molecules, peptides, oligonucleotides and lipids/liposomes/lipid nanoparticles. The latter of which are being seen more and more in drug development, including the recent Covid pandemic, where mRNA and lipid nanoparticle technology is playing more of a role in vaccine development. The translation between non-clinical and clinical data is also considered, questioning how a physicochemical driven toxicity may be more universal across species, which means that such toxicity may be reassuringly translatable between species and as such, this information may also be considered as a support to the 3 R's, particularly in the early screening stages of a drug development plan.
Collapse
|
11
|
Martinez-Lopez S, Angel-Gomis E, Sanchez-Ardid E, Pastor-Campos A, Picó J, Gomez-Hurtado I. The 3Rs in Experimental Liver Disease. Animals (Basel) 2023; 13:2357. [PMID: 37508134 PMCID: PMC10376896 DOI: 10.3390/ani13142357] [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: 06/14/2023] [Revised: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Patients with cirrhosis present multiple physiological and immunological alterations that play a very important role in the development of clinically relevant secondary complications to the disease. Experimentation in animal models is essential to understand the pathogenesis of human diseases and, considering the high prevalence of liver disease worldwide, to understand the pathophysiology of disease progression and the molecular pathways involved, due to the complexity of the liver as an organ and its relationship with the rest of the organism. However, today there is a growing awareness about the sensitivity and suffering of animals, causing opposition to animal research among a minority in society and some scientists, but also about the attention to the welfare of laboratory animals since this has been built into regulations in most nations that conduct animal research. In 1959, Russell and Burch published the book "The Principles of Humane Experimental Technique", proposing that in those experiments where animals were necessary, everything possible should be done to try to replace them with non-sentient alternatives, to reduce to a minimum their number, and to refine experiments that are essential so that they caused the least amount of pain and distress. In this review, a comprehensive summary of the most widely used techniques to replace, reduce, and refine in experimental liver research is offered, to assess the advantages and weaknesses of available experimental liver disease models for researchers who are planning to perform animal studies in the near future.
Collapse
Affiliation(s)
- Sebastian Martinez-Lopez
- Instituto ISABIAL, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain
- Departamento de Medicina Clínica, Universidad Miguel Hernández, 03550 Sant Joan, Spain
| | - Enrique Angel-Gomis
- Instituto ISABIAL, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain
- Departamento de Medicina Clínica, Universidad Miguel Hernández, 03550 Sant Joan, Spain
| | - Elisabet Sanchez-Ardid
- CIBERehd, Instituto de Salud Carlos III, 28220 Madrid, Spain
- Servicio de Patología Digestiva, Institut de Recerca IIB-Sant Pau, Hospital de Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Alberto Pastor-Campos
- Oficina de Investigación Responsable, Universidad Miguel Hernández, 03202 Elche, Spain
| | - Joanna Picó
- Instituto ISABIAL, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain
| | - Isabel Gomez-Hurtado
- Instituto ISABIAL, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain
- Departamento de Medicina Clínica, Universidad Miguel Hernández, 03550 Sant Joan, Spain
- CIBERehd, Instituto de Salud Carlos III, 28220 Madrid, Spain
| |
Collapse
|
12
|
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
|
13
|
Carpentier N, Urbani L, Dubruel P, Van Vlierberghe S. The native liver as inspiration to create superior in vitro hepatic models. Biomater Sci 2023; 11:1091-1115. [PMID: 36594602 DOI: 10.1039/d2bm01646j] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Drug induced liver injury (DILI) is one of the major reasons of drug withdrawal during the different phases of drug development. The later in the drug development a drug is discovered to be toxic, the higher the economical as well as the ethical impact will be. In vitro models for early detection of drug liver toxicity are under constant development, however to date a superior model of the liver is still lacking. Ideally, a highly reliable model should be established to maintain the different hepatic cell functionalities to the greatest extent possible, during a period of time long enough to allow for tracking of the toxicity of compounds. In the case of DILI, toxicity can appear even after months of exposure. To reach this goal, an in vitro model should be developed that mimics the in vivo liver environment, function and response to external stimuli. The different approaches for the development of liver models currently used in the field of tissue engineering will be described in this review. Combining different technologies, leading to optimal materials, cells and 3D-constructs will ultimately lead to an ideal superior model that fully recapitulates the liver.
Collapse
Affiliation(s)
- Nathan Carpentier
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| | - Luca Urbani
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK.,Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Peter Dubruel
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| |
Collapse
|
14
|
Molecular Mechanisms of Hepatotoxicity. Int J Mol Sci 2023; 24:ijms24043791. [PMID: 36835204 PMCID: PMC9961832 DOI: 10.3390/ijms24043791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Drug-induced liver injury, also known as drug-induced hepatotoxicity (DILI), is a major cause of medicine withdrawal (prescription or over-the-counter) from the market [...].
Collapse
|
15
|
Lin J, Li M, Mak W, Shi Y, Zhu X, Tang Z, He Q, Xiang X. Applications of In Silico Models to Predict Drug-Induced Liver Injury. TOXICS 2022; 10:788. [PMID: 36548621 PMCID: PMC9785299 DOI: 10.3390/toxics10120788] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 06/17/2023]
Abstract
Drug-induced liver injury (DILI) is a major cause of the withdrawal of pre-marketed drugs, typically attributed to oxidative stress, mitochondrial damage, disrupted bile acid homeostasis, and innate immune-related inflammation. DILI can be divided into intrinsic and idiosyncratic DILI with cholestatic liver injury as an important manifestation. The diagnosis of DILI remains a challenge today and relies on clinical judgment and knowledge of the insulting agent. Early prediction of hepatotoxicity is an important but still unfulfilled component of drug development. In response, in silico modeling has shown good potential to fill the missing puzzle. Computer algorithms, with machine learning and artificial intelligence as a representative, can be established to initiate a reaction on the given condition to predict DILI. DILIsym is a mechanistic approach that integrates physiologically based pharmacokinetic modeling with the mechanisms of hepatoxicity and has gained increasing popularity for DILI prediction. This article reviews existing in silico approaches utilized to predict DILI risks in clinical medication and provides an overview of the underlying principles and related practical applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qingfeng He
- Correspondence: (Q.H.); (X.X.); Tel.: +86-21-51980024 (X.X.)
| | - Xiaoqiang Xiang
- Correspondence: (Q.H.); (X.X.); Tel.: +86-21-51980024 (X.X.)
| |
Collapse
|