1
|
Uetrecht J. DILI prediction in drug development: present and future. Expert Opin Drug Metab Toxicol 2025; 21:665-676. [PMID: 40253704 DOI: 10.1080/17425255.2025.2495955] [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: 01/22/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
INTRODUCTION Idiosyncratic drug-induced liver injury (iDILI) results in significant patient morbidity and significantly increases the risk of drug development. The current methods to screen for iDILI risk are inadequate. AREAS COVERED The general mechanism of iDILI and the current methods to screen for iDILI are reviewed. Then the potential for new biomarkers is explored. EXPERT OPINION Better biomarkers of iDILI risk should be based on the mechanism of iDILI. In general, it is an adaptive immune response, specifically CD8+ cytotoxic T cells, that is responsible for hepatocyte cell death, not direct toxicity of the drug. Therefore, in vitro cytotoxicity assays represent an artifact not the mechanism of iDILI. Activation of the adaptive immune response leading to iDILI requires an innate immune response, in particular activation of antigen presenting cells. The innate immune response is immediate and unlikely to be idiosyncratic. For example, studies have found that incubation of hepatocytes with drugs causes the release of molecules that activate THP-1-derived macrophages. The response of hepatocytes, the release of damage-associated molecular pattern molecules (DAMPs), especially in extracellular vesicles, and the response of antigen presenting cells (APCs) are likely to provide better biomarkers of iDILI risk.
Collapse
Affiliation(s)
- Jack Uetrecht
- Faculty of Pharmacy, University of Toronto, Toronto, ON, USA
| |
Collapse
|
2
|
Dassarma B, Mahapatra SK, Nandi DK, Gangopadhyay S, Samanta S. Protective role of butylated hydroxyanisole (BHA) and hydroxytoluene (BHT) against oxidative stress-induced inflammatory response in carbon tetrachloride-induced acute hepatorenal toxicity. Arch Physiol Biochem 2025:1-8. [PMID: 40272386 DOI: 10.1080/13813455.2025.2493105] [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: 10/01/2024] [Revised: 02/11/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Any toxicity initially damages the hepatic system, followed by renal dysfunction. Previously, it was established that carbon tetrachloride (CCl4) intoxication severely damaged hepatocytes. Moreover, CCl4-mediated toxicity significantly impacted immune functions and influenced the inflammatory response, with mitochondrial dysfunction. The present study focused on the levels of inflammatory markers and mitochondrial dysfunction, as well as the protective role of BHA and BHT. METHODS In the present study, hepatorenal dysfunction was developed in experimental rats by applying a subcutaneous injection of CCl4 with a dose of 230 mg/kg bwt/rat/day. The level of immune toxicity was determined by measuring C-reactive protein (CRP), IL-6, 12, TNF-α, IL-10, and TGF-β in CCl4 intoxicated group and pretreated BHA and BHT groups. ROS generation and MMP were also measured in hepatic and renal cells using flow cytometric technique. RESULTS The level of toxicity was determined by a significant increase of CRP (407.29%), IL-6 (525.65%), IL-12 (1026.54%), and TNF-α (1007.33%) in CCl4 intoxicated group, while IL-10 and TGF-β were significantly decreased 84.65% and 66.36%, respectively. CCl4 intoxication caused decreased mitochondrial membrane potential and high levels of intracellular ROS generation. Pretreatment with BHA (0.5 mg/kg/bwt) and BHT (0.8 mg/kg/bwt) significantly (p<0.001, p<0.05) reduced inflammatory markers in the CCl4-treated group, restored mitochondrial membrane potential and decreased intracellular ROS levels. CONCLUSION BHA and BHT treatment could restrict the higher concentration of pro-inflammatory markers by scavenging ROS. Therefore, the study suggested that supplementation of BHA and BHT could be an alternative treatment for preventing hepatorenal dysfunctions.
Collapse
Affiliation(s)
- Barsha Dassarma
- Department of Physiology, Midnapore College, Midnapore, India
- Department of Physiology, Raja N.L. Khan Women's College (Autonomous), Midnapore, India
- Department of Physiology and Allied Sciences, Amity Institute of Health and Allied Sciences, Amity University Uttar Pradesh, Noida, India
| | - Santanu Kar Mahapatra
- Department of Paramedical and Allied Health Sciences, Midnapore City College, Midnapore, India
| | - Dilip Kumar Nandi
- Department of Physiology, Raja N.L. Khan Women's College (Autonomous), Midnapore, India
| | - Somnath Gangopadhyay
- Department of Physiology, Occupational Ergonomics Laboratory, University College of Science and Technology, University of Calcutta, Kolkata, India
| | | |
Collapse
|
3
|
Shinozawa T, Miyamoto K, Baker KS, Faber SC, Flores R, Uetrecht J, von Hehn C, Yukawa T, Tohyama K, Kadali H, von Grotthuss M, Sudo Y, Smith EN, Diogo D, Zhu AZX, Dragan Y, Cebers G, Wagoner MP. TAK-994 mechanistic investigation into drug-induced liver injury. Toxicol Sci 2025; 204:143-153. [PMID: 39786842 PMCID: PMC11939078 DOI: 10.1093/toxsci/kfaf003] [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: 01/12/2025] Open
Abstract
The frequency of drug-induced liver injury (DILI) in clinical trials remains a challenge for drug developers despite advances in human hepatotoxicity models and improvements in reducing liver-related attrition in preclinical species. TAK-994, an oral orexin receptor 2 agonist, was withdrawn from phase II clinical trials due to the appearance of severe DILI. Here, we investigate the likely mechanism of TAK-994 DILI in hepatic cell culture systems examined cytotoxicity, mitochondrial toxicity, impact on drug transporter proteins, and covalent binding. Hepatic liabilities were absent in rat and nonhuman primate safety studies, however, murine studies initiated during clinical trials revealed hepatic single-cell necrosis following cytochrome P450 induction at clinically relevant doses. Hepatic cell culture experiments uncovered wide margins to known mechanisms of intrinsic DILI, including cytotoxicity (>100× Cmax/IC50), mitochondrial toxicity (>100× Cmax/IC50), and bile salt efflux pump inhibition (>20× Css, avg/IC50). A potential covalent binding liability was uncovered with TAK-994 following hepatic metabolism consistent with idiosyncratic DILI and the delayed-onset clinical toxicity. Although idiosyncratic DILI is challenging to detect preclinically, reductions in total daily dose and covalent binding can reduce the covalent body binding burden and, subsequently, the clinical incidence of idiosyncratic DILI.
Collapse
Affiliation(s)
| | - Kazumasa Miyamoto
- Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Kevin S Baker
- Takeda Development Center Americas, Inc, Cambridge, MA 02139, United States
| | - Samantha C Faber
- Takeda Development Center Americas, Inc, San Diego, CA 92121, United States
| | | | - Jack Uetrecht
- Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Christian von Hehn
- Takeda Development Center Americas, Inc, Cambridge, MA 02139, United States
| | - Tomoya Yukawa
- Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Kimio Tohyama
- Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Harisha Kadali
- Takeda Development Center Americas, Inc, Cambridge, MA 02139, United States
| | | | - Yusuke Sudo
- Takeda Pharmaceutical Company Ltd, Fujisawa, Kanagawa 251-8555, Japan
| | - Erin N Smith
- Takeda Development Center Americas, Inc, San Diego, CA 92121, United States
| | - Dorothée Diogo
- Takeda Development Center Americas, Inc, Cambridge, MA 02139, United States
| | - Andy Z X Zhu
- Takeda Development Center Americas, Inc, Cambridge, MA 02139, United States
| | - Yvonne Dragan
- Takeda Development Center Americas, Inc, Cambridge, MA 02139, United States
| | - Gvido Cebers
- Takeda Development Center Americas, Inc, Cambridge, MA 02139, United States
| | - Matthew P Wagoner
- Takeda Development Center Americas, Inc, Cambridge, MA 02139, United States
| |
Collapse
|
4
|
Hou Y, Gao Z, Tu P, Ai X, Jiang Y. Establishment of an Idiosyncratic Drug-Induced Liver Injury Model on a Stacked Array Chip for Identification of CCL5-Mediated Paracrine Dynamics. Anal Chem 2024; 96:19370-19377. [PMID: 39602550 DOI: 10.1021/acs.analchem.4c03552] [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: 11/29/2024]
Abstract
Idiosyncratic drug-induced liver injury (iDILI) poses significant challenges in both drug development and clinical practice due to its unpredictable nature and poorly understood mechanism. The current in vitro iDILI models are limited in their ability to replicate dynamic paracrine signaling in the inflammatory microenvironment. Here, we develop an iDILI model on a stacked array chip, allowing ease of assembly and disassembly for precise temporal manipulation of 3D liver microtissue and macrophages. First, the iDILI model is constructed and optimized on the chip to effectively distinguish drugs inducing idiosyncratic versus intrinsic liver injuries. Next, the iDILI mechanism is investigated using nimesulide (NIM) as a case study. Our organ-on-a-chip model successfully recapitulates iDILI, offering a platform to distinguish drugs between intrinsic and idiosyncratic liver injury. Our findings revealed that NIM-induced iDILI triggered inflammation-induced injury in the liver microtissues through activating the TNF pathways. Moreover, NIM-induced iDILI promotes the M1 polarization of macrophages through CCL5-mediated paracrine dynamics, influenced by the interactions between hepatocytes and macrophages. Leveraging the flexibility of the chip, we observe a dynamic equilibrium between preactivation of inflammation and the pretreatment of NIM during iDILI process. Therefore, our developed iDILI model on a stacked array chip provides a valuable tool for identifying iDILI drugs and understanding the importance of temporal specificity in intercellular signaling in iDILI.
Collapse
Affiliation(s)
- Yu Hou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhao Gao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiaoni Ai
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yong Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
5
|
Lim AY, Kato Y, Sakolish C, Valdiviezo A, Han G, Bajaj P, Stanko J, Ferguson SS, Villenave R, Hewitt P, Hardwick RN, Rusyn I. Reproducibility and Robustness of a Liver Microphysiological System PhysioMimix LC12 under Varying Culture Conditions and Cell Type Combinations. Bioengineering (Basel) 2023; 10:1195. [PMID: 37892925 PMCID: PMC10603899 DOI: 10.3390/bioengineering10101195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/04/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
The liver is one of the key organs for exogenous and endogenous metabolism and is often a target for drug- and chemical-driven toxicity. A wide range of experimental approaches has been established to model and characterize the mechanisms of drug- and chemical-induced hepatotoxicity. A number of microfluidics-enabled in vitro models of the liver have been developed, but the unclear translatability of these platforms has hindered their adoption by the pharmaceutical industry; to achieve wide use for drug and chemical safety evaluation, demonstration of reproducibility and robustness under various contexts of use is required. One of these commercially available platforms is the PhysioMimix LC12, a microfluidic device where cells are seeded into a 3D scaffold that is continuously perfused with recirculating cell culture media to mimic liver sinusoids. Previous studies demonstrated this model's functionality and potential applicability to preclinical drug development. However, to gain confidence in PhysioMimix LC12's robustness and reproducibility, supplementary characterization steps are needed, including the assessment of various human hepatocyte sources, contribution of non-parenchymal cells (NPCs), and comparison to other models. In this study, we performed replicate studies averaging 14 days with either primary human hepatocytes (PHHs) or induced pluripotent stem cell (iPSC)-derived hepatocytes, with and without NPCs. Albumin and urea secretion, lactate dehydrogenase, CYP3A4 activity, and metabolism were evaluated to assess basal function and metabolic capacity. Model performance was characterized by different cell combinations under intra- and inter-experimental replication and compared to multi-well plates and other liver platforms. PhysioMimix LC12 demonstrated the highest metabolic function with PHHs, with or without THP-1 or Kupffer cells, for up to 10-14 days. iPSC-derived hepatocytes and PHHs co-cultured with additional NPCs demonstrated sub-optimal performance. Power analyses based on replicate experiments and different contexts of use will inform future study designs due to the limited throughput and high cell demand. Overall, this study describes a workflow for independent testing of a complex microphysiological system for specific contexts of use, which may increase end-user adoption in drug development.
Collapse
Affiliation(s)
- Alicia Y. Lim
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Yuki Kato
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
- Laboratory for Drug Discovery and Development, Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Courtney Sakolish
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Alan Valdiviezo
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Gang Han
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, TX 77843, USA
| | - Piyush Bajaj
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA 02141, USA
| | - Jason Stanko
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Stephen S. Ferguson
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Remi Villenave
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, 64293 Darmstadt, Germany
| | - Rhiannon N. Hardwick
- Discovery Toxicology, Pharmaceutical Candidate Optimization, Bristol Myers Squibb, San Diego, CA 92121, USA
| | - Ivan Rusyn
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
6
|
Kaden T, Graf K, Rennert K, Li R, Mosig AS, Raasch M. Evaluation of drug-induced liver toxicity of trovafloxacin and levofloxacin in a human microphysiological liver model. Sci Rep 2023; 13:13338. [PMID: 37587168 PMCID: PMC10432496 DOI: 10.1038/s41598-023-40004-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 08/03/2023] [Indexed: 08/18/2023] Open
Abstract
Drug-induced liver injury induced by already approved substances is a major threat to human patients, potentially resulting in drug withdrawal and substantial loss of financial resources in the pharmaceutical industry. Trovafloxacin, a broad-spectrum fluoroquinolone, was found to have unexpected side effects of severe hepatotoxicity, which was not detected by preclinical testing. To address the limitations of current drug testing strategies mainly involving 2D cell cultures and animal testing, a three-dimensional microphysiological model of the human liver containing expandable human liver sinusoidal endothelial cells, monocyte-derived macrophages and differentiated HepaRG cells was utilized to investigate the toxicity of trovafloxacin and compared it to the structurally-related non-toxic drug levofloxacin. In the model, trovafloxacin elicited vascular and hepatocellular toxicity associated with pro-inflammatory cytokine release already at clinically relevant concentrations, whereas levofloxacin did not provoke tissue injury. Similar to in vivo, cytokine secretion was dependent on a multicellular immune response, highlighting the potential of the complex microphysiological liver model for reliably detecting drug-related cytotoxicity in preclinical testing. Moreover, hepatic glutathione depletion and mitochondrial ROS formation were elucidated as intrinsic toxicity mechanisms contributing to trovafloxacin toxicity.
Collapse
Affiliation(s)
- Tim Kaden
- Dynamic42 GmbH, Jena, Germany
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | | | | | - Ruoya Li
- Biopredic International, St Gregoire, France
| | - Alexander S Mosig
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | | |
Collapse
|
7
|
Villanueva-Badenas E, Donato MT, Tolosa L. Mechanistic Understanding of Idiosyncratic Drug-Induced Hepatotoxicity Using Co-Cultures of Hepatocytes and Macrophages. Antioxidants (Basel) 2023; 12:1315. [PMID: 37507855 PMCID: PMC10376129 DOI: 10.3390/antiox12071315] [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: 06/01/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
Hepatotoxicity or drug-induced liver injury (DILI) is a major safety issue in drug development as a primary reason for drug failure in clinical trials and the main cause for post-marketing regulatory measures like drug withdrawal. Idiosyncratic DILI (iDILI) is a patient-specific, multifactorial, and multicellular process that cannot be recapitulated in current in vitro models; thus, our major goal is to develop and fully characterize a co-culture system and to evaluate its suitability for predicting iDILI. For this purpose, we used human hepatoma HepG2 cells and macrophages differentiated from a monocyte cell line (THP-1) and established the appropriate co-culture conditions for mimicking an inflammatory environment. Then, mono-cultures and co-cultures were treated with model iDILI compounds (trovafloxacin, troglitazone) and their parent non-iDILI compounds (levofloxacin, rosiglitazone), and the effects on viability and the mechanisms implicated (i.e., oxidative stress induction) were analyzed. Our results show that co-culture systems including hepatocytes (HepG2) and other cell types (THP-1-derived macrophages) help to enhance the mechanistic understanding of iDILI, providing better hepatotoxicity predictions.
Collapse
Affiliation(s)
- Estela Villanueva-Badenas
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Odontología, Universidad de Valencia, 46010 Valencia, Spain
| | - M Teresa Donato
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Odontología, Universidad de Valencia, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Laia Tolosa
- Unidad de Hepatología Experimental, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
8
|
Kato Y, Lim AY, Sakolish C, Valdiviezo A, Moyer HL, Hewitt P, Bajaj P, Han G, Rusyn I. Analysis of reproducibility and robustness of OrganoPlate® 2-lane 96, a liver microphysiological system for studies of pharmacokinetics and toxicological assessment of drugs. Toxicol In Vitro 2022; 85:105464. [PMID: 36057418 PMCID: PMC10015056 DOI: 10.1016/j.tiv.2022.105464] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/26/2022] [Accepted: 08/26/2022] [Indexed: 02/06/2023]
Abstract
Establishing the functionality, reproducibility, robustness, and reliability of microphysiological systems is a critical need for adoption of these technologies. A high throughput microphysiological system for liver studies was recently proposed in which induced pluripotent stem cell-derived hepatocytes (iHeps) and non-parenchymal cells (endothelial cells and THP-1 cells differentiated with phorbol 12-myristate 13-acetate into macrophage-like cells) were co-cultured in OrganoPlate® 2-lane 96 devices. The goal of this study was to evaluate this platform using additional cell types and conditions and characterize its utility and reproducibility. Primary human hepatocytes or iHeps, with and without non-parenchymal cells, were cultured for up to 17 days. Image-based cell viability, albumin and urea secretion into culture media, CYP3A4 activity and drug metabolism were assessed. The iHeps co-cultured with non-parenchymal cells demonstrated stable cell viability and function up to 17 days; however, variability was appreciable both within and among studies. The iHeps in monoculture did not form clusters and lost viability and function over time. The primary human hepatocytes in monoculture also exhibited low cell viability and hepatic function. Metabolism of various drugs was most efficient when iHeps were co-cultured with non-parenchymal cells. Overall, we found that the OrganoPlate® 2-lane 96 device, when used with iHeps and non-parenchymal cells, is a functional liver microphysiological model; however, the high-throughput nature of this model is somewhat dampened by the need for replicates to compensate for high variability.
Collapse
Affiliation(s)
- Yuki Kato
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; Laboratory for Drug Discovery and Development, Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Alicia Y Lim
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Courtney Sakolish
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Alan Valdiviezo
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Haley L Moyer
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, 64293 Darmstadt, Germany
| | - Piyush Bajaj
- Global Investigative Toxicology, Preclinical Safety, Sanofi USA, MA 01701, USA
| | - Gang Han
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, TX 77843, USA
| | - Ivan Rusyn
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
9
|
Li D, Lyu Y, Song Q, Lai YS, Zuo Z. Idiosyncratic liver injury induced by bolus combination treatment with emodin and 2,3,5,4′-tetrahydroxystilbene-2-O-β-D-glucopyranoside in rats. Front Pharmacol 2022; 13:1017741. [PMID: 36225587 PMCID: PMC9549410 DOI: 10.3389/fphar.2022.1017741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Polygoni Multiflori Radix (PMR) is a commonly used traditional Chinese medicine in clinical practice, while adverse effects of hepatotoxicity related to PMR have been frequently reported. The clinical case reports indicated that PMR hepatotoxicity could occur under both overdose medication/long-term exposure and low doses with short-duration (idiosyncratic) conditions. The combination treatment with emodin and 2,3,5,4′-tetrahydroxystilbene-2-O-β-D-glucopyranoside (TSG), two major PMR components, was reported to contribute to PMR hepatotoxicity after long-term treatment. However, the role of the combination treatment of these two components in PMR-induced idiosyncratic liver injury has not been clearly clarified. In this study, the LPS-mediated inflammatory stress model rats were adopted to explore the idiosyncratic liver injury induced by the bolus combination treatment with emodin and TSG. After a bolus oral administration with TSG (165 mg/kg), emodin (5 mg/kg) or their combination in both normal and LPS-mediated inflammatory stress model rats, the systemic/hepatic concentrations of emodin, emodin glucuronides and bile acids were determined; the hepatotoxicity assessments were conducted via monitoring histopathological changes and liver injury biomarkers (ALT and AST). Moreover, the protein expressions of bile acid homeostasis- and apoptosis-related proteins were examined. No liver damage was observed in the normal rats after a bolus dose with the individual or combination treatment, while the bolus combination treatment with emodin and TSG induced liver injury in the LPS-mediated inflammatory stress model rats, evidenced by the elevated plasma levels of alanine aminotransferase (∼66%) and aspartate aminotransferase (∼72%) accompanied by severe inflammatory cell infiltration and apoptotic hepatocytes in liver tissue. Moreover, such combination treatment at a bolus dose in the LPS-mediated inflammatory stress model rats could significantly elevate the hepatic TBA levels by about 45% via up-regulating the hepatic protein expression levels of bile acid synthesis enzymes and inhibiting that of bile acid efflux transporters and the expression levels of apoptosis-related proteins. Our study for the first time proved the major contribution of the combination treatment with emodin and TSG in PMR-induced idiosyncratic liver injury.
Collapse
|
10
|
Qin T, Hasnat M, Zhou Y, Yuan Z, Zhang W. Macrophage malfunction in Triptolide-induced indirect hepatotoxicity. Front Pharmacol 2022; 13:981996. [PMID: 36225585 PMCID: PMC9548637 DOI: 10.3389/fphar.2022.981996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/23/2022] [Indexed: 12/05/2022] Open
Abstract
Background and Objective: Indirect hepatotoxicity is a new type of drug-induced hepatotoxicity in which the character of a drug that may induce its occurrence and the underlying mechanism remains elusive. Previously, we proved that Triptolide (TP) induced indirect hepatotoxicity upon LPS stimulation resulting from the deficiency of cytoprotective protein of hepatocyte. However, whether immune cells participated in TP-induced indirect hepatotoxicity and the way immune cells change the liver hypersensitivity to LPS still need to be deeply investigated. In this study, we tried to explore whether and how macrophages are involved in TP-induced indirect hepatotoxicity. Method: Firstly, TP (500 μg/kg) and LPS (0.1 mg/kg) were administrated into female C57BL/6 mice as previously reported. Serum biochemical indicators, morphological changes, hepatic macrophage markers, as well as macrophage M1/M2 markers were detected. Secondly, macrophage scavenger clodronate liposomes were injected to prove whether macrophages participated in TP-induced indirect hepatotoxicity. Also, the ability of macrophages to secrete inflammatory factors and macrophage phagocytosis were detected. Lastly, reverse docking was used to find the target of TP on macrophage and the possible target was verified in vivo and in RAW264.7 cells. Results: TP pretreatment increased the liver hypersensitization to LPS accompanied by the recruitment of macrophages to the liver and promoted the transformation of macrophages to M1 type. Depletion of hepatic macrophages almost completely alleviated the liver injury induced by TP/LPS. TP pretreatment increased the secretion of pro-inflammatory factors and weakened the phagocytic function of macrophages upon LPS exposure. Reverse docking results revealed that MerTK might be the real target of TP. Conclusion: TP disrupts inflammatory cytokines profile and phagocytic function of hepatic macrophages, resulting in the production of massive inflammatory factors and the accumulation of endotoxin in the liver, ultimately leading to the indirect hepatotoxicity of TP. MerTK might be the target of TP on the macrophage, while the binding of TP to MerTK should be investigated in vivo and in vitro.
Collapse
Affiliation(s)
- Tingting Qin
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center for Tumor Precision Medicine and Comprehensive Evaluation, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou University, Zhengzhou, China
- Henan Provincial Key Laboratory of Anticancer Drug Research, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou University, Zhengzhou, China
| | - Muhammad Hasnat
- Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Yang Zhou
- Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou University, Zhengzhou, China
| | - Ziqiao Yuan
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- *Correspondence: Wenzhou Zhang, , Ziqiao Yuan, ,
| | - Wenzhou Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center for Tumor Precision Medicine and Comprehensive Evaluation, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou University, Zhengzhou, China
- Henan Provincial Key Laboratory of Anticancer Drug Research, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou University, Zhengzhou, China
- *Correspondence: Wenzhou Zhang, , Ziqiao Yuan, ,
| |
Collapse
|
11
|
Gu R, Liang A, Liao G, To I, Shehu A, Ma X. Roles of Cofactors in Drug-Induced Liver Injury: Drug Metabolism and Beyond. Drug Metab Dispos 2022; 50:646-654. [PMID: 35221288 PMCID: PMC9132098 DOI: 10.1124/dmd.121.000457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 02/22/2022] [Indexed: 11/22/2022] Open
Abstract
Drug-induced liver injury (DILI) remains one of the major concerns for healthcare providers and patients. Unfortunately, it is difficult to predict and prevent DILI in the clinic because detailed mechanisms of DILI are largely unknown. Many risk factors have been identified for both "intrinsic" and "idiosyncratic" DILI, suggesting that cofactors are an important aspect in understanding DILI. This review outlines the cofactors that potentiate DILI and categorizes them into two types: (1) the specific cofactors that target metabolic enzymes, transporters, antioxidation defense, immune response, and liver regeneration; and (2) the general cofactors that include inflammation, age, gender, comorbidity, gut microbiota, and lifestyle. The underlying mechanisms by which cofactors potentiate DILI are also discussed. SIGNIFICANCE STATEMENT: This review summarizes the risk factors for DILI, which can be used to predict and prevent DILI in the clinic. This work also highlights the gaps in the DILI field and provides future perspectives on the roles of cofactors in DILI.
Collapse
Affiliation(s)
- Ruizhi Gu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences (R.G., A.S., X.M.) and School of Pharmacy (A.L., G.L., I.T.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alina Liang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences (R.G., A.S., X.M.) and School of Pharmacy (A.L., G.L., I.T.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Grace Liao
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences (R.G., A.S., X.M.) and School of Pharmacy (A.L., G.L., I.T.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Isabelle To
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences (R.G., A.S., X.M.) and School of Pharmacy (A.L., G.L., I.T.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Amina Shehu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences (R.G., A.S., X.M.) and School of Pharmacy (A.L., G.L., I.T.), University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xiaochao Ma
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences (R.G., A.S., X.M.) and School of Pharmacy (A.L., G.L., I.T.), University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
12
|
Tasnim F, Huang X, Lee CZW, Ginhoux F, Yu H. Recent Advances in Models of Immune-Mediated Drug-Induced Liver Injury. FRONTIERS IN TOXICOLOGY 2022; 3:605392. [PMID: 35295156 PMCID: PMC8915912 DOI: 10.3389/ftox.2021.605392] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatic inflammation is a key feature of a variety of liver diseases including drug-induced liver injury (DILI), orchestrated by the innate immune response (Kupffer cells, monocytes, neutrophils, dendritic cells) and the adaptive immune system (T cells and natural killer T cells). In contrast to acute DILI, prediction of immune-mediated DILI (im-DILI) has been more challenging due to complex disease pathogenesis, lack of reliable models and limited knowledge of underlying mechanisms. This review summarizes in vivo and in vitro systems that have been used to model im-DILI. In particular, the review focuses on state-of-the-art in vitro human-based multicellular models which have been developed to supplement the use of in vivo models due to interspecies variation and increasing ethical concerns regarding animal use. Advantages of the co-cultures in maintaining hepatocyte functions and importantly, introducing heterotypic cell-cell interactions to mimic inflammatory hepatic microenvironment are discussed. Challenges regarding cell source and incorporation of different cells with physical cell-cell contact are outlined and potential solutions are proposed. It is likely that better understanding of the interplay of immune cells in liver models will allow for the development of more accurate systems to better predict hepatotoxicity and stratification of drugs that can cause immune-mediated effects.
Collapse
Affiliation(s)
- Farah Tasnim
- Innovations in Food & Chemical Safety Programme, ASTAR, Singapore, Singapore.,Institute of Bioengineering and Nanotechnology, The Nanos, Singapore, Singapore
| | - Xiaozhong Huang
- Innovations in Food & Chemical Safety Programme, ASTAR, Singapore, Singapore.,Institute of Bioengineering and Nanotechnology, The Nanos, Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christopher Zhe Wei Lee
- Innovations in Food & Chemical Safety Programme, ASTAR, Singapore, Singapore.,Singapore Immunology Network, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Florent Ginhoux
- Innovations in Food & Chemical Safety Programme, ASTAR, Singapore, Singapore.,Singapore Immunology Network, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Hanry Yu
- Innovations in Food & Chemical Safety Programme, ASTAR, Singapore, Singapore.,Institute of Bioengineering and Nanotechnology, The Nanos, Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,National University of Singapore (NUS) Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences, Singapore, Singapore.,T-Labs, Mechanobiology Institute, Singapore, Singapore.,Critical Analytics for Manufacturing Personalised-Medicine Interdisciplinary Research Groups (CAMP-IRG), Singapore-Massachusetts Institute of Technology Alliance for Research and Technology, Singapore, Singapore
| |
Collapse
|
13
|
Lin L, Chen Y, Li Q, Xu G, Ding K, Ren L, Shi W, Wang Y, Li Z, Dai W, Wei Z, Yang Y, Bai Z, Xiao X. Isoxanthohumol, a component of Sophora flavescens, promotes the activation of the NLRP3 inflammasome and induces idiosyncratic hepatotoxicity. JOURNAL OF ETHNOPHARMACOLOGY 2022; 285:114796. [PMID: 34740771 DOI: 10.1016/j.jep.2021.114796] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/15/2021] [Accepted: 10/30/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sophora flavescens is a traditional Chinese medicine commonly used in clinical practice, which has the effects of clearing away heat and dampness. Unfortunately, it has been reported that Sophora flavescens and its preparation may cause liver damage to a certain extent, but the exact mechanism is not clear. AIM OF THE STUDY To assess the safety and risk of Sophora flavescens and to elucidate the relationship between Idiosyncratic drug-induced liver injury (IDILI) and the NOD-like receptor family protein 3 (NLRP3) inflammasome. MATERIALS AND METHODS Western blot, Caspase-Glo® 1 Inflammasome Assay, ELISA kits, Flow cytometry and FLIPRT Tetra system were used to study the effect of isoxanthohumol (IXN) on the activation of NLRP3 inflammasome and its mechanism. Combined with the lipopolysaccharide-mediated susceptibility IDILI model in mice to evaluate the hepatotoxicity of IXN. RESULTS IXN facilitates the activation of caspase-1 and secretion of interleukin (IL)-1β triggered by adenosine triphosphate (ATP), nigericin but not those induced by silicon dioxide and poly (I:C). Furthermore, the activation of NLR-family CARD-containing protein 4 (NLRC4) and the absent in melanoma 2 (AIM2) was not affected by IXN. Mechanistically, IXN promotes NLRP3-dependent apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC) oligomerization and the generation of mitochondrial reactive oxygen species (mtROS) triggered by ATP. The in vivo data showed that non-hepatotoxic doses of IXN resulted in increased levels of glutamate-pyruvate transaminase, glutamate-oxaloacetate transaminase, tumor necrosis factor and IL-1β in the serum and showed increased liver inflammation in the susceptible IDILI model mediated by lipopolysaccharide. CONCLUSIONS These results show that IXN enhances NLRP3 inflammasome activation by promoting the accumulation of ATP-induced mtROS and ASC oligomerization to cause IDILI, indicating that IXN may be a risk factor for liver injury caused by the clinical use of Sophora flavescens.
Collapse
Affiliation(s)
- Li Lin
- School of Pharmacy, Dali University, Dali, 671000, China; Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Yuanyuan Chen
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qiang Li
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China; School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Guang Xu
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Kaixin Ding
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Lutong Ren
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Wei Shi
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Yan Wang
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Zhiyong Li
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Wenzhang Dai
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Ziying Wei
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Yan Yang
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Zhaofang Bai
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China.
| | - Xiaohe Xiao
- School of Pharmacy, Dali University, Dali, 671000, China; Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China; China Military Institute of Chinese Materia, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
14
|
Hefler J, Marfil-Garza BA, Pawlick RL, Freed DH, Karvellas CJ, Bigam DL, Shapiro AMJ. Preclinical models of acute liver failure: a comprehensive review. PeerJ 2021; 9:e12579. [PMID: 34966588 PMCID: PMC8667744 DOI: 10.7717/peerj.12579] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/10/2021] [Indexed: 12/14/2022] Open
Abstract
Acute liver failure is marked by the rapid deterioration of liver function in a previously well patient over period of days to weeks. Though relatively rare, it is associated with high morbidity and mortality. This makes it a challenging disease to study clinically, necessitating reliance on preclinical models as means to explore pathophysiology and novel therapies. Preclinical models of acute liver failure are artificial by nature, and generally fall into one of three categories: surgical, pharmacologic or immunogenic. This article reviews preclinical models of acute liver failure and considers their relevance in modeling clinical disease.
Collapse
Affiliation(s)
- Joshua Hefler
- Division of General Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Braulio A Marfil-Garza
- Division of General Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada.,National Institutes of Medical Sciences & Nutrition Salvador Zubiran, Mexico City, Mexico.,CHRISTUS-LatAm Hub Excellence & Innovation Center, Monterrey, Mexico
| | - Rena L Pawlick
- Division of General Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Darren H Freed
- Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Constantine J Karvellas
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Department of Critical Care Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - David L Bigam
- Division of General Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - A M James Shapiro
- Division of General Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
15
|
Segovia-Zafra A, Di Zeo-Sánchez DE, López-Gómez C, Pérez-Valdés Z, García-Fuentes E, Andrade RJ, Lucena MI, Villanueva-Paz M. Preclinical models of idiosyncratic drug-induced liver injury (iDILI): Moving towards prediction. Acta Pharm Sin B 2021; 11:3685-3726. [PMID: 35024301 PMCID: PMC8727925 DOI: 10.1016/j.apsb.2021.11.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
Idiosyncratic drug-induced liver injury (iDILI) encompasses the unexpected harms that prescription and non-prescription drugs, herbal and dietary supplements can cause to the liver. iDILI remains a major public health problem and a major cause of drug attrition. Given the lack of biomarkers for iDILI prediction, diagnosis and prognosis, searching new models to predict and study mechanisms of iDILI is necessary. One of the major limitations of iDILI preclinical assessment has been the lack of correlation between the markers of hepatotoxicity in animal toxicological studies and clinically significant iDILI. Thus, major advances in the understanding of iDILI susceptibility and pathogenesis have come from the study of well-phenotyped iDILI patients. However, there are many gaps for explaining all the complexity of iDILI susceptibility and mechanisms. Therefore, there is a need to optimize preclinical human in vitro models to reduce the risk of iDILI during drug development. Here, the current experimental models and the future directions in iDILI modelling are thoroughly discussed, focusing on the human cellular models available to study the pathophysiological mechanisms of the disease and the most used in vivo animal iDILI models. We also comment about in silico approaches and the increasing relevance of patient-derived cellular models.
Collapse
Affiliation(s)
- Antonio Segovia-Zafra
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
| | - Daniel E. Di Zeo-Sánchez
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| | - Carlos López-Gómez
- Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga 29010, Spain
| | - Zeus Pérez-Valdés
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| | - Eduardo García-Fuentes
- Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga 29010, Spain
| | - Raúl J. Andrade
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
| | - M. Isabel Lucena
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
- Platform ISCIII de Ensayos Clínicos, UICEC-IBIMA, Málaga 29071, Spain
| | - Marina Villanueva-Paz
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| |
Collapse
|
16
|
Zhai XR, Zou ZS, Wang JB, Xiao XH. Herb-Induced Liver Injury Related to Reynoutria multiflora (Thunb.) Moldenke: Risk Factors, Molecular and Mechanistic Specifics. Front Pharmacol 2021; 12:738577. [PMID: 34539416 PMCID: PMC8443768 DOI: 10.3389/fphar.2021.738577] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
Herbal medicine is widely used in Asia as well as the west. Hepatotoxicity is one of the most severe side effects of herbal medicine which is an increasing concern around the world. Reynoutria multiflora (Thunb.) Moldenke (Polygonum multiflorum Thunb., PM) is the most common herb that can cause herb-induced liver injury (HILI). The recent scientific and technological advancements in clinical and basic research are paving the way for a better understanding of the molecular aspects of PM-related HILI (PM-HILI). This review provides an updated overview of the clinical characteristics, predisposing factors, hepatotoxic components, and molecular mechanisms of PM-HILI. It can also aid in a better understanding of HILI and help in further research on the same.
Collapse
Affiliation(s)
- Xing-Ran Zhai
- Peking University 302 Clinical Medical School, Beijing, China
| | - Zheng-Sheng Zou
- Peking University 302 Clinical Medical School, Beijing, China
- Medical School of Chinese PLA, Beijing, China
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Jia-Bo Wang
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, China
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xiao-He Xiao
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, China
- China Military Institute of Chinese Medicine, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
17
|
Qin N, Xu G, Wang Y, Zhan X, Gao Y, Wang Z, Fu S, Shi W, Hou X, Wang C, Li R, Liu Y, Wang J, Zhao H, Xiao X, Bai Z. Bavachin enhances NLRP3 inflammasome activation induced by ATP or nigericin and causes idiosyncratic hepatotoxicity. Front Med 2021; 15:594-607. [PMID: 33909257 DOI: 10.1007/s11684-020-0809-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 06/03/2020] [Indexed: 12/31/2022]
Abstract
Psoraleae Fructus (PF) is a well-known traditional herbal medicine in China, and it is widely used for osteoporosis, vitiligo, and other diseases in clinical settings. However, liver injury caused by PF and its preparations has been frequently reported in recent years. Our previous studies have demonstrated that PF could cause idiosyncratic drug-induced liver injury (IDILI), but the mechanism underlying its hepatotoxicity remains unclear. This paper reports that bavachin isolated from PF enhances the specific stimuli-induced activation of the NLRP3 inflammasome and leads to hepatotoxicity. Bavachin boosts the secretion of IL-1β and caspase-1 caused by ATP or nigericin but not those induced by poly(I:C), monosodium urate crystal, or intracellular lipopolysaccharide. Bavachin does not affect AIM2 or NLRC4 inflammasome activation. Mechanistically, bavachin specifically increases the production of nigericin-induced mitochondrial reactive oxygen species among the most important upstream events in the activation of the NLRP3 inflammasome. Bavachin increases the levels of aspartate transaminase and alanine aminotransferase in serum and hepatocyte injury accompanied by the secretion of IL-1β via a mouse model of lipopolysaccharide-mediated susceptibility to IDILI. These results suggest that bavachin specifically enhances the ATP- or nigericin-induced activation of the NLRP3 inflammasome. Bavachin also potentially contributes to PF-induced idiosyncratic hepatotoxicity. Moreover, bavachin and PF should be evaded among patients with diseases linked to the ATP- or nigericin-mediated activation of the NLRP3 inflammasome, which may be a dangerous factor for liver injury.
Collapse
Affiliation(s)
- Nan Qin
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China.,Department of Pharmacy, Nantong Hospital of Traditional Chinese Medicine, Nantong, 226300, China
| | - Guang Xu
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Yan Wang
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China.,School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Xiaoyan Zhan
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Yuan Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Zhilei Wang
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Shubin Fu
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Wei Shi
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Xiaorong Hou
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Chunyu Wang
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Ruisheng Li
- Research Center for Clinical and Translational Medicine, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Yan Liu
- Research Center for Clinical and Translational Medicine, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Jiabo Wang
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Haiping Zhao
- School of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China.
| | - Xiaohe Xiao
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China. .,Integrative Medical Centre, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China.
| | - Zhaofang Bai
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
18
|
Gerussi A, Natalini A, Antonangeli F, Mancuso C, Agostinetto E, Barisani D, Di Rosa F, Andrade R, Invernizzi P. Immune-Mediated Drug-Induced Liver Injury: Immunogenetics and Experimental Models. Int J Mol Sci 2021; 22:4557. [PMID: 33925355 PMCID: PMC8123708 DOI: 10.3390/ijms22094557] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023] Open
Abstract
Drug-induced liver injury (DILI) is a challenging clinical event in medicine, particularly because of its ability to present with a variety of phenotypes including that of autoimmune hepatitis or other immune mediated liver injuries. Limited diagnostic and therapeutic tools are available, mostly because its pathogenesis has remained poorly understood for decades. The recent scientific and technological advancements in genomics and immunology are paving the way for a better understanding of the molecular aspects of DILI. This review provides an updated overview of the genetic predisposition and immunological mechanisms behind the pathogenesis of DILI and presents the state-of-the-art experimental models to study DILI at the pre-clinical level.
Collapse
Affiliation(s)
- Alessio Gerussi
- Centre for Autoimmune Liver Diseases, Division of Gastroenterology, Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (C.M.); (D.B.); (P.I.)
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, 20900 Monza, Italy
| | - Ambra Natalini
- Institute of Molecular Biology and Pathology (IBPM), National Research Council of Italy (CNR), 00185 Rome, Italy; (A.N.); (F.A.); (F.D.R.)
| | - Fabrizio Antonangeli
- Institute of Molecular Biology and Pathology (IBPM), National Research Council of Italy (CNR), 00185 Rome, Italy; (A.N.); (F.A.); (F.D.R.)
| | - Clara Mancuso
- Centre for Autoimmune Liver Diseases, Division of Gastroenterology, Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (C.M.); (D.B.); (P.I.)
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, 20900 Monza, Italy
| | - Elisa Agostinetto
- Academic Trials Promoting Team, Institut Jules Bordet, L’Universite’ Libre de Bruxelles (ULB), 1050 Brussels, Belgium;
- Medical Oncology and Hematology Unit, Humanitas Clinical and Research Center—IRCCS, Humanitas Cancer Center, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy
| | - Donatella Barisani
- Centre for Autoimmune Liver Diseases, Division of Gastroenterology, Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (C.M.); (D.B.); (P.I.)
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, 20900 Monza, Italy
| | - Francesca Di Rosa
- Institute of Molecular Biology and Pathology (IBPM), National Research Council of Italy (CNR), 00185 Rome, Italy; (A.N.); (F.A.); (F.D.R.)
| | - Raul Andrade
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), UGC Aparato Digestivo, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, 29016 Málaga, Spain;
| | - Pietro Invernizzi
- Centre for Autoimmune Liver Diseases, Division of Gastroenterology, Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (C.M.); (D.B.); (P.I.)
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, 20900 Monza, Italy
| |
Collapse
|
19
|
Ahn JH, Jegal H, Choi MS, Kim S, Park SM, Ahn J, Han HY, Cho HS, Yoon S, Oh JH. TNFα enhances trovafloxacin-induced in vitro hepatotoxicity by inhibiting protective autophagy. Toxicol Lett 2021; 342:73-84. [PMID: 33609687 DOI: 10.1016/j.toxlet.2021.02.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 02/09/2021] [Accepted: 02/12/2021] [Indexed: 12/12/2022]
Abstract
Trovafloxacin (TVX) is associated with idiosyncratic drug-induced liver injury (iDILI) and inflammation-mediated hepatotoxicity. However, the inflammatory stress-regulated mechanisms in iDILI remain unclear. Herein, we elucidated the novel role of tumor-necrosis factor alpha (TNFα), an inflammatory stress factor, in TVX-induced in vitro hepatotoxicity and synergistic toxicity. TVX specifically induced synergistic toxicity in HepG2 cells with TNFα, which inhibits autophagy. TVX-treated HepG2 cells induced protective autophagy by inhibiting the expression of mTOR signaling proteins, while ATG5 knockdown in HepG2 cells, responsible for the impairment of autophagy, enhanced TVX-induced toxicity due to the increase in cytochrome C release and JNK pathway activation. Interestingly, the expression of mTOR signal proteins, which were suppressed by TVX, disrupted the negative feedback of the PI3K/AKT pathway and TNFα rebounded p70S6K phosphorylation. Co-treatment with TVX and TNFα inhibited protective autophagy by maintaining p70S6K activity, which enhanced TVX-induced cytotoxicity. Phosphorylation of p70S6K was inhibited by siRNA knockdown and rapamycin to restore TNFα-inhibited autophagy, which prevented the synergistic effect on TVX-induced cytotoxicity. These results indicate that TVX activates protective autophagy in HepG2 cells exposed to toxicity and an imbalance in negative feedback regulation of autophagy by TNFα synergistically enhanced the toxicity. The finding from this study may contribute to a better understanding of the mechanisms underlying iDILI associated with inflammatory stress.
Collapse
Affiliation(s)
- Jun-Ho Ahn
- Department of Predictive Toxicology, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea; Bio Medical Research Center, Bio Medical & Health Division, Korea Testing Laboratory (KTL), Seoul, 08389, Republic of Korea
| | - Hyun Jegal
- Department of Predictive Toxicology, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea; Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, 34113, Republic of Korea
| | - Mi-Sun Choi
- Department of Predictive Toxicology, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Soojin Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Se-Myo Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Jaehwan Ahn
- Department of Predictive Toxicology, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Hyoung-Yun Han
- Department of Predictive Toxicology, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Hyun-Soo Cho
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Seokjoo Yoon
- Department of Predictive Toxicology, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea; Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, 34113, Republic of Korea.
| | - Jung-Hwa Oh
- Department of Predictive Toxicology, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea; Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
20
|
Hastings KL, Green MD, Gao B, Ganey PE, Roth RA, Burleson GR. Beyond Metabolism: Role of the Immune System in Hepatic Toxicity. Int J Toxicol 2021; 39:151-164. [PMID: 32174281 DOI: 10.1177/1091581819898399] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The liver is primarily thought of as a metabolic organ; however, the liver is also an important mediator of immunological functions. Key perspectives on this emerging topic were presented in a symposium at the 2018 annual meeting of the American College of Toxicology entitled "Beyond metabolism: Role of the immune system in hepatic toxicity." Viral hepatitis is an important disease of the liver for which insufficient preventive vaccines exist. Host immune responses inadequately clear these viruses and often potentiate immunological inflammation that damages the liver. In addition, the liver is a key innate immune organ against bacterial infection. Hepatocytes and immune cells cooperatively control systemic and local bacterial infections. Conversely, bacterial infection can activate multiple types of immune cells and pathways to cause hepatocyte damage and liver injury. Finally, the immune system and specifically cytokines and drugs can interact in idiosyncratic drug-induced liver injury. This rare disease can result in a disease spectrum that ranges from mild to acute liver failure. The immune system plays a role in this disease spectrum.
Collapse
Affiliation(s)
| | | | - Bin Gao
- Laboratory of Liver Diseases, NIH, Bethesda, MD, USA
| | - Patricia E Ganey
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Robert A Roth
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Gary R Burleson
- BRT-Burleson Research Technologies, Inc, Morrisville, NC, USA
| |
Collapse
|
21
|
Rao T, Liu YT, Zeng XC, Li CP, Ou-Yang DS. The hepatotoxicity of Polygonum multiflorum: The emerging role of the immune-mediated liver injury. Acta Pharmacol Sin 2021; 42:27-35. [PMID: 32123300 PMCID: PMC7921551 DOI: 10.1038/s41401-020-0360-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/02/2020] [Indexed: 02/06/2023]
Abstract
Herbal and dietary supplements (HDS)-induced liver injury has been a great concern all over the world. Polygonum multiflorum Thunb., a well-known Chinese herbal medicine, is recently drawn increasing attention because of its hepatotoxicity. According to the clinical and experimental studies, P. multiflorum-induced liver injury (PM-DILI) is considered to be immune-mediated idiosyncratic liver injury, but the role of immune response and the underlying mechanisms are not completely elucidated. Previous studies focused on the direct toxicity of PM-DILI by using animal models with intrinsic drug-induced liver injury (DILI). However, most epidemiological and clinical evidence demonstrate that PM-DILI is immune-mediated idiosyncratic liver injury. The aim of this review is to assess current epidemiological, clinical and experimental evidence about the possible role of innate and adaptive immunity in the idiosyncratic hepatotoxicity of P. multiflorum. The potential effects of factors associated with immune tolerance, including immune checkpoint molecules and regulatory immune cells on the individual's susceptibility to PM-DILI are also discussed. We conclude by giving our hypothesis of possible immune mechanisms of PM-DILI and providing suggestions for future studies on valuable biomarkers identification and proper immune models establishment.
Collapse
Affiliation(s)
- Tai Rao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410008, China.
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China.
| | - Ya-Ting Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410008, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Xiang-Chang Zeng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410008, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Chao-Peng Li
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, 410205, China
| | - Dong-Sheng Ou-Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410008, China.
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China.
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, 410205, China.
| |
Collapse
|
22
|
Roth RA, Ganey PE. What have we learned from animal models of idiosyncratic, drug-induced liver injury? Expert Opin Drug Metab Toxicol 2020; 16:475-491. [PMID: 32324077 DOI: 10.1080/17425255.2020.1760246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Idiosyncratic, drug-induced liver injury (IDILI) continues to plague patients and restrict the use of drugs that are pharmacologically effective. Mechanisms of IDILI are incompletely understood, and a better understanding would reduce speculation and could help to identify safer drug candidates preclinically. Animal models have the potential to enhance knowledge of mechanisms of IDILI. AREAS COVERED Numerous hypotheses have emerged to explain IDILI pathogenesis, many of which center on the roles of the innate and/or adaptive immune systems. Animal models based on these hypotheses are reviewed in the context of their contributions to understanding of IDILI and their limitations. EXPERT OPINION Animal models of IDILI based on an activated adaptive immune system have to date failed to reproduce major liver injury that is of most concern clinically. The only models that have so far resulted in pronounced liver injury are based on the multiple determinant hypothesis or the inflammatory stress hypothesis. The liver pathogenesis in IDILI animal models involves various leukocytes and immune mediators such as cytokines. Insights from animal models are changing the way we view IDILI pathogenesis and are leading to better approaches to preclinical prediction of IDILI potential of new drug candidates.
Collapse
Affiliation(s)
- Robert A Roth
- Department of Pharmacology and Toxicology and Institute for Integrative Toxicology, Michigan State University , East Lansing, MI, USA
| | - Patricia E Ganey
- Department of Pharmacology and Toxicology and Institute for Integrative Toxicology, Michigan State University , East Lansing, MI, USA
| |
Collapse
|
23
|
Yuan Z, Yuan Z, Hasnat M, Zhang H, Liang P, Sun L, Jiang Z, Zhang L. A new perspective of triptolide-associated hepatotoxicity: the relevance of NF- κ B and NF- κ B-mediated cellular FLICE-inhibitory protein. Acta Pharm Sin B 2020; 10:861-877. [PMID: 32528833 PMCID: PMC7280150 DOI: 10.1016/j.apsb.2020.02.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/05/2019] [Accepted: 01/02/2020] [Indexed: 02/07/2023] Open
Abstract
Previously, we proposed a new perspective of triptolide (TP)-associated hepatotoxicity: liver hypersensitivity upon lipopolysaccharide (LPS) stimulation. However, the mechanisms for TP/LPS-induced hepatotoxicity remained elusive. The present study aimed to clarify the role of LPS in TP/LPS-induced hepatotoxicity and the mechanism by which TP induces liver hypersensitivity upon LPS stimulation. TNF-α inhibitor, etanercept, was injected intraperitoneally into mice to investigate whether induction of TNF-α by LPS participated in the liver injury induced by TP/LPS co-treatment. Mice and hepatocytes pretreated with TP were stimulated with recombinant TNF-α to assess the function of TNF-α in TP/LPS co-treatment. Additionally, time-dependent NF-κB activation and NF-κB-mediated pro-survival signals were measured in vivo and in vitro. Finally, overexpression of cellular FLICE-inhibitory protein (FLIP), the most potent NF-κB-mediated pro-survival protein, was measured in vivo and in vitro to assess its function in TP/LPS-induced hepatotoxicity. Etanercept counteracted the toxic reactions induced by TP/LPS. TP-treatment sensitized mice and hepatocytes to TNF-α, revealing the role of TNF-α in TP/LPS-induced hepatotoxicity. Mechanistic studies revealed that TP inhibited NF-κB dependent pro-survival signals, especially FLIP, induced by LPS/TNF-α. Moreover, overexpression of FLIP alleviated TP/LPS-induced hepatotoxicity in vivo and TP/TNF-α-induced apoptosis in vitro. Mice and hepatocytes treated with TP were sensitive to TNF-α, which was released from LPS-stimulated immune cells. These and other results show that the TP-induced inhibition of NF-κB-dependent transcriptional activity and FLIP production are responsible for liver hypersensitivity.
Collapse
Key Words
- CIAPs, cellular inhibitor of apoptosis proteins
- Etan, etanercept
- FADD, FAS-associated protein with death domain
- FLIP
- FLIP, cellular FLICE-inhibitory protein
- IκB-α, NF-κB inhibitor alpha
- LDH, lactate dehydrogenase
- LPS
- LPS, lipopolysaccharide
- MLKL, mixed lineage kinase domain like pseudokinase
- MPO, myeloperoxidase
- NF-κB
- PAS, periodic acid-schiff
- RIPK1/3, receptor-interacting protein kinase 1/3
- TNF-R1, tumor necrosis factor receptor type 1
- TNF-α
- TNFAIP3, TNF-α-induced protein 3
- TP, triptolide
- TRADD, TNF receptor-associated death domain
- TRAF2, TNF receptor-associated factor 2
- Triptolide
- XIAP, X-linked inhibitor of apoptosis protein
Collapse
Affiliation(s)
- Ziqiao Yuan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Zihang Yuan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Muhammad Hasnat
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
- Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Haoran Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Peishi Liang
- College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lixin Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing 21009, China
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
24
|
Giustarini G, Vrisekoop N, Kruijssen L, Wagenaar L, van Staveren S, van Roest M, Bleumink R, Bol-Schoenmakers M, Weaver RJ, Koenderman L, Smit J, Pieters R. Trovafloxacin-Induced Liver Injury: Lack in Regulation of Inflammation by Inhibition of Nucleotide Release and Neutrophil Movement. Toxicol Sci 2020; 167:385-396. [PMID: 30247740 DOI: 10.1093/toxsci/kfy244] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The fluoroquinolone trovafloxacin (TVX) is associated with a high risk of drug-induced liver injury (DILI). Although part of the liver damage by TVX+TNF relies on neutrophils, we have recently demonstrated that liver recruitment of monocytes and neutrophils is delayed by TVX. Here we show that the delayed leukocyte recruitment is caused by a combination of effects which are linked to the capacity of TVX to block the hemichannel pannexin 1. TVX inhibited find-me signal release in apoptotic HepG2 hepatocytes, decelerated freshly isolated human neutrophils toward IL-8 and f-MLF, and decreased the liver expression of ICAM-1. In blood of TVX+TNF-treated mice, we observed an accumulation of activated neutrophils despite an increased MIP-2 release by the liver. Depletion of monocytes and neutrophils caused increased serum concentrations of TNF, IL-6, and MIP-2 in TVX-treated mice as well as in mice treated with the fluoroquinolone levofloxacin, known to have a lower DILI-inducing profile. This supports the idea that early leukocyte recruitment regulates inflammation. In conclusion, disrupted regulation by leukocytes appears to constitute a fundamental step in the onset of TVX-induced liver injury, acting in concert with the capability of TVX to induce hepatocyte cell death. Interference of leukocyte-mediated regulation of inflammation represents a novel mechanism to explain the onset of DILI.
Collapse
Affiliation(s)
- Giulio Giustarini
- Immunotoxicology, Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences, Yalelaan 104, 3584CM, Utrecht University, Utrecht, The Netherlands
| | - Nienke Vrisekoop
- Department of Respiratory Medicine and Laboratory of Translational Immunology (LTI), University Medical Center Utrecht, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
| | - Laura Kruijssen
- Immunotoxicology, Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences, Yalelaan 104, 3584CM, Utrecht University, Utrecht, The Netherlands
| | - Laura Wagenaar
- Immunotoxicology, Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences, Yalelaan 104, 3584CM, Utrecht University, Utrecht, The Netherlands
| | - Selma van Staveren
- Department of Respiratory Medicine and Laboratory of Translational Immunology (LTI), University Medical Center Utrecht, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
| | - Manon van Roest
- Immunotoxicology, Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences, Yalelaan 104, 3584CM, Utrecht University, Utrecht, The Netherlands
| | - Rob Bleumink
- Immunotoxicology, Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences, Yalelaan 104, 3584CM, Utrecht University, Utrecht, The Netherlands
| | - Marianne Bol-Schoenmakers
- Immunotoxicology, Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences, Yalelaan 104, 3584CM, Utrecht University, Utrecht, The Netherlands
| | - Richard J Weaver
- Institut de Recherches Internationales Servier (I.R.I.S.), Suresnes 92284, France
| | - Leo Koenderman
- Department of Respiratory Medicine and Laboratory of Translational Immunology (LTI), University Medical Center Utrecht, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
| | - Joost Smit
- Immunotoxicology, Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences, Yalelaan 104, 3584CM, Utrecht University, Utrecht, The Netherlands
| | - Raymond Pieters
- Immunotoxicology, Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences, Yalelaan 104, 3584CM, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
25
|
Mir TM, Ma G, Ali Z, Khan IA, Ashfaq MK. Effect of Raspberry Ketone on Normal, Obese and Health-Compromised Obese Mice: A Preliminary Study. J Diet Suppl 2019; 18:1-16. [PMID: 31603036 DOI: 10.1080/19390211.2019.1674996] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Raspberry ketone (RK)-an aromatic compound found mostly in red raspberries (Rubus idaeus) is widely used as an over the counter product for weight loss. The present study was conducted to determine adverse effects associated with RK in obese and health-compromised obese mice. Two sets of experiments were conducted on normal obese and health-compromised obese mice treated with RK for a duration of 10 days. Obese conditions were induced by feeding mice a high fat diet for 10 weeks, while the health compromised obese mouse model was developed by a single intraperitoneal injection of a nontoxic dose of lipopolysaccharide (LPS) (6 mg/kg) to obese mice. Results showed that RK (165, 330, and 500 mg/kg) under obese as well as health-compromised condition retarded the gain in body weights as compared to the control groups. RK at doses 330 and 500 mg/kg resulted in 67.6 and 50% mortality, respectively in normal obese mice and 70% mortality was observed in health-compromised obese mice treated with RK at 500 mg/kg. At higher doses deaths were observed earlier than those given lower doses of RK. Significant elevations in blood alanine transaminase (ALT) were also observed with RK treatment in obese mice. Blood glucose levels were significantly elevated in all groups of mice treated with RK. This study suggests that higher doses of RK may cause adverse effects in health compromised conditions. Under these conditions, prolonged use of RK, especially in high doses, may pose a health hazard.
Collapse
Affiliation(s)
- Tahir Maqbool Mir
- National Center for Natural Product Research, School of Pharmacy, University of Mississippi, University, MS, USA
| | - Guoyi Ma
- Drug Discovery Division, Southern Research, Birmingham, AL, UK
| | - Zulfiqar Ali
- National Center for Natural Product Research, School of Pharmacy, University of Mississippi, University, MS, USA
| | - Ikhlas A Khan
- National Center for Natural Product Research, School of Pharmacy, University of Mississippi, University, MS, USA
| | - Mohammad K Ashfaq
- National Center for Natural Product Research, School of Pharmacy, University of Mississippi, University, MS, USA
| |
Collapse
|
26
|
Metabolic Comorbidities and Risk of Development and Severity of Drug-Induced Liver Injury. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8764093. [PMID: 31531370 PMCID: PMC6720367 DOI: 10.1155/2019/8764093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/09/2019] [Accepted: 07/25/2019] [Indexed: 12/15/2022]
Abstract
The incidence and rates of diagnosis of drug-induced liver injury (DILI) have been increasing in recent years as findings from basic research and the examination of clinical databases reveal information about the clinical course, etiology, and prognosis of this complex disease. The prevalence of metabolic comorbidities (e.g., diabetes mellitus, fatty liver, obesity, and metabolic syndrome (MetS)) has been increasing during the same period. The results of preclinical and clinical research studies indicate that characteristics of metabolic comorbidities are also factors that affect DILI phenotype and progression. The objective of this review is to present the evidence for DILI and hepatotoxicity mechanisms, incidence, and outcomes in patients with MetS and nonalcoholic fatty liver disease. Moreover, we also summarize the relationships between drugs used to treat metabolic comorbidities and DILI.
Collapse
|
27
|
Wu W, Wang T, Sun B, Liu D, Lin Z, Miao Y, Wang C, Geng X, Li B. Xian-Ling-Gu-Bao induced inflammatory stress rat liver injury: Inflammatory and oxidative stress playing important roles. JOURNAL OF ETHNOPHARMACOLOGY 2019; 239:111910. [PMID: 31026554 DOI: 10.1016/j.jep.2019.111910] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 03/31/2019] [Accepted: 04/23/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xian-Ling-Gu-Bao (XLGB) Fufang is an herbal formula that has been used in clinical settings to treat osteoporosis, osteoarthritis, aseptic bone necrosis, and climacteric syndrome. Despite its uses, XLGB treatment has been linked to potential liver injury. To date, there is a lack of clear demonstration of such toxicity in animal models. AIM OF THE STUDY As animal models fail to reproduce the XLGB hepatotoxicity reported in humans, because human hepatocytes are clearly more sensitive to XLGB, this study was designed to investigate a more reliable animal model of such toxicity. MATERIALS AND METHODS We randomized rats into five groups, as follows: CON (control), XLGB, lipopolysaccharide (LPS), L-XLGB/LPS (XLGB, 0.125 g/kg; LPS, 0.1 mg/kg), and XLGB/LPS (XLGB, 1.25 g/kg; LPS, 0.1 mg/kg). These groups were treated with 0.5% sodium carboxymethyl cellulose (CMC-Na), XLGB suspension, normal saline, or LPS. The first administration of XLGB [0.125 g/kg or 1.25 g/kg, by mouth (PO)] or its solvent (0.5% CMC-Na) was delivered, and then food was removed. Twelve hours after the first administration of XLGB, rats received LPS [0.1 mg/kg, intravenously (IV)] or saline control. After 30 min, a second administration of XLGB (0.125 g/kg or 1.25 g/kg, PO) or solvent was administered. The rats were anesthetized at 12 h or 24 h following the second administration of XLGB. Liver function was evaluated by measuring liver weight, liver microscopy, serum biochemistry and plasma microRNA-122 (miR-122). The plasma levels of 27 cytokines were measured to evaluate inflammation. Moreover, the expression of cytochrome P450 2E1 (CYP2E1), nicotinic adenine dinucleotide phosphate (NADPH) oxidase and inducible nitric oxide synthase (iNOS) at protein levels were observed; immunofluorescence and immunohistochemistry were used to confirmed the hepatotoxicity of XLGB. RESULTS Hepatotoxicity in male rats with moderate inflammation induced by XLGB was indicated by liver histopathology, serum biochemical analysis, serum miR-122 levels, and immunofluorescent assessments. We observed significant increases in liver weight and liver indexes in male rats with moderate inflammation in response to XLGB. Histopathological assessment further showed that extensive hepatocellular necrosis and inflammatory infiltration were evident in rats co-treated with XLGB/LPS. The levels of serum transaminases [alanine aminotransferase (ALT), aspartate aminotransferase (AST), gamma-glutamyl transferase (GGT)], total bilirubin (TBIL) and triglyceride (TG), which are markers of liver function, were also significantly increased by XLGB/LPS treatment. Similarly, miR-122 was significantly elevated in XLGB/LPS treated rats relative to other groups. An immunofluorescent assessment showed extensive apoptosis in hepatocytes from these co-treated rats. What is more, XLGB can dose-dependently induce liver injury in male rats with moderate inflammation. Hepatic CYP2E1, neutrophil chemotactic factor (NCF-1), iNOS, and NOX-2 (an NADPH oxidase subunit) levels were increased in response to XLGB treatment, and staining for DMPO nitrone adducts further showed elevated oxidative stress level in XLGB/LPS-treated rats relative to the other experimental groups. CONCLUSION LPS and XLGB co-treatment in rats led to marked hepatotoxicity. This toxicity was associated with disrupted lipid metabolism, extensive liver necrosis and inflammatory infiltration, apoptosis, and expression of oxidative stress-related proteins. These results demonstrate a valuable model for the study of iDILI in the context of XLGB treatment, and further provide insights into the potential mechanisms by which XLGB may induce hepatotoxicity in humans.
Collapse
Affiliation(s)
- Wenxiao Wu
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China; National Center for Safety Evaluation of Drugs, National Institute for Food and Drug Control, Key Laboratory of Beijing for Nonclinical Safety Evaluation of Drugs, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, China
| | - Ting Wang
- National Center for Safety Evaluation of Drugs, National Institute for Food and Drug Control, Key Laboratory of Beijing for Nonclinical Safety Evaluation of Drugs, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, China
| | - Bo Sun
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China; National Center for Safety Evaluation of Drugs, National Institute for Food and Drug Control, Key Laboratory of Beijing for Nonclinical Safety Evaluation of Drugs, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, China
| | - Dong Liu
- National Center for Safety Evaluation of Drugs, National Institute for Food and Drug Control, Key Laboratory of Beijing for Nonclinical Safety Evaluation of Drugs, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, China
| | - Zhi Lin
- National Center for Safety Evaluation of Drugs, National Institute for Food and Drug Control, Key Laboratory of Beijing for Nonclinical Safety Evaluation of Drugs, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, China
| | - Yufa Miao
- National Center for Safety Evaluation of Drugs, National Institute for Food and Drug Control, Key Laboratory of Beijing for Nonclinical Safety Evaluation of Drugs, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, China
| | - Chao Wang
- National Center for Safety Evaluation of Drugs, National Institute for Food and Drug Control, Key Laboratory of Beijing for Nonclinical Safety Evaluation of Drugs, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, China
| | - Xingchao Geng
- National Center for Safety Evaluation of Drugs, National Institute for Food and Drug Control, Key Laboratory of Beijing for Nonclinical Safety Evaluation of Drugs, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing, 100176, China.
| | - Bo Li
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China; National Institute for Food and Drug Control, 31 Hua Tuo Road, Daxing District, Beijing, 102629, China.
| |
Collapse
|
28
|
Research Progress on the Animal Models of Drug-Induced Liver Injury: Current Status and Further Perspectives. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1283824. [PMID: 31119149 PMCID: PMC6500714 DOI: 10.1155/2019/1283824] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023]
Abstract
Drug-induced liver injury (DILI) is a major concern in clinical studies as well as in postmarketing surveillance. It is necessary to establish an animal model of DILI for thorough investigation of mechanisms of DILI and searching for protective medications. This article reviews the current status and future perspective on establishment of DILI models based on different hepatotoxic drugs, as well as the underlying mechanisms of liver function damage induced by specific medicine. Therefore, information from this article can help researchers make a suitable selection of animal models for further study.
Collapse
|
29
|
McGill MR, Jaeschke H. Animal models of drug-induced liver injury. Biochim Biophys Acta Mol Basis Dis 2018; 1865:1031-1039. [PMID: 31007174 DOI: 10.1016/j.bbadis.2018.08.037] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/18/2018] [Accepted: 08/28/2018] [Indexed: 01/08/2023]
Abstract
Drug-induced liver injury (DILI) presents unique challenges for consumers, clinicians, and regulators. It is the most common cause of acute liver failure in the US. It is also one of the most common reasons for termination of new drugs during pre-clinical testing and withdrawal of new drugs post-marketing. DILI is generally divided into two forms: intrinsic and idiosyncratic. Many of the challenges with DILI are due in large part to poor understanding of the mechanisms of toxicity. Although useful models of intrinsic DILI are available, they are frequently misused. Modeling idiosyncratic DILI presents greater challenges, but promising new models have recently been developed. The purpose of this manuscript is to provide a critical review of the most popular animal models of DILI, and to discuss the future of DILI research.
Collapse
Affiliation(s)
- Mitchell R McGill
- Dept. of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Dept. of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Hartmut Jaeschke
- Dept. of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
30
|
Oppelt A, Kaschek D, Huppelschoten S, Sison-Young R, Zhang F, Buck-Wiese M, Herrmann F, Malkusch S, Krüger CL, Meub M, Merkt B, Zimmermann L, Schofield A, Jones RP, Malik H, Schilling M, Heilemann M, van de Water B, Goldring CE, Park BK, Timmer J, Klingmüller U. Model-based identification of TNFα-induced IKKβ-mediated and IκBα-mediated regulation of NFκB signal transduction as a tool to quantify the impact of drug-induced liver injury compounds. NPJ Syst Biol Appl 2018; 4:23. [PMID: 29900006 PMCID: PMC5995845 DOI: 10.1038/s41540-018-0058-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 04/16/2018] [Accepted: 05/04/2018] [Indexed: 02/07/2023] Open
Abstract
Drug-induced liver injury (DILI) has become a major problem for patients and for clinicians, academics and the pharmaceutical industry. To date, existing hepatotoxicity test systems are only poorly predictive and the underlying mechanisms are still unclear. One of the factors known to amplify hepatotoxicity is the tumor necrosis factor alpha (TNFα), especially due to its synergy with commonly used drugs such as diclofenac. However, the exact mechanism of how diclofenac in combination with TNFα induces liver injury remains elusive. Here, we combined time-resolved immunoblotting and live-cell imaging data of HepG2 cells and primary human hepatocytes (PHH) with dynamic pathway modeling using ordinary differential equations (ODEs) to describe the complex structure of TNFα-induced NFκB signal transduction and integrated the perturbations of the pathway caused by diclofenac. The resulting mathematical model was used to systematically identify parameters affected by diclofenac. These analyses showed that more than one regulatory module of TNFα-induced NFκB signal transduction is affected by diclofenac, suggesting that hepatotoxicity is the integrated consequence of multiple changes in hepatocytes and that multiple factors define toxicity thresholds. Applying our mathematical modeling approach to other DILI-causing compounds representing different putative DILI mechanism classes enabled us to quantify their impact on pathway activation, highlighting the potential of the dynamic pathway model as a quantitative tool for the analysis of DILI compounds.
Collapse
Affiliation(s)
- Angela Oppelt
- 1Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Kaschek
- 2Institute of Physics, University of Freiburg, Freiburg, Germany
| | - Suzanna Huppelschoten
- 3Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Rowena Sison-Young
- 4MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Fang Zhang
- 4MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Marie Buck-Wiese
- 1Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Franziska Herrmann
- 1Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Malkusch
- 5Institute of Physical and Theoretical Chemistry, Single Molecule Biophysics, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Carmen L Krüger
- 5Institute of Physical and Theoretical Chemistry, Single Molecule Biophysics, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Mara Meub
- 5Institute of Physical and Theoretical Chemistry, Single Molecule Biophysics, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Benjamin Merkt
- 2Institute of Physics, University of Freiburg, Freiburg, Germany
| | - Lea Zimmermann
- 1Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Amy Schofield
- 4MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Robert P Jones
- 4MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK.,6North Western Hepatobiliary Unit, Aintree University Hospital NHS Foundation Trust, Liverpool, UK
| | - Hassan Malik
- 6North Western Hepatobiliary Unit, Aintree University Hospital NHS Foundation Trust, Liverpool, UK
| | - Marcel Schilling
- 1Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mike Heilemann
- 5Institute of Physical and Theoretical Chemistry, Single Molecule Biophysics, Johann Wolfgang Goethe-University, Frankfurt, Germany.,7Bioquant, University of Heidelberg, Heidelberg, Germany
| | - Bob van de Water
- 3Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Christopher E Goldring
- 4MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - B Kevin Park
- 4MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Jens Timmer
- 2Institute of Physics, University of Freiburg, Freiburg, Germany.,8BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Ursula Klingmüller
- 1Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
31
|
Stephens C, Lucena MI, Andrade RJ. Host Risk Modifiers in Idiosyncratic Drug-Induced Liver Injury (DILI) and Its Interplay with Drug Properties. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/978-1-4939-7677-5_23] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
32
|
Garg C, Seo JH, Ramachandran J, Loh JM, Calderon F, Contreras JE. Trovafloxacin attenuates neuroinflammation and improves outcome after traumatic brain injury in mice. J Neuroinflammation 2018; 15:42. [PMID: 29439712 PMCID: PMC5812039 DOI: 10.1186/s12974-018-1069-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 01/17/2018] [Indexed: 12/20/2022] Open
Abstract
Background Trovafloxacin is a broad-spectrum antibiotic, recently identified as an inhibitor of pannexin-1 (Panx1) channels. Panx1 channels are important conduits for the adenosine triphosphate (ATP) release from live and dying cells that enhances the inflammatory response of immune cells. Elevated extracellular levels ATP released upon injury activate purinergic pathways in inflammatory cells that promote migration, proliferation, phagocytosis, and apoptotic signals. Here, we tested whether trovafloxacin administration attenuates the neuroinflammatory response and improves outcomes after brain trauma. Methods The murine controlled cortical impact (CCI) model was used to determine whether in vivo delivery of trovafloxacin has anti-inflammatory and neuroprotective actions after brain trauma. Locomotor deficit was assessed using the rotarod test. Levels of tissue damage markers and inflammation were measured using western blot, qPCR, and immunofluorescence. In vitro assays were used to evaluate whether trovafloxacin blocks ATP release and cell migration in a chemotactic-stimulated microglia cell line. Results Trovafloxacin treatment of CCI-injured mice significantly reduced tissue damage markers and improved locomotor deficits. In addition, trovafloxacin treatment significantly reduced mRNA levels of several pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α), which correlates with an overall reduction in the accumulation of inflammatory cell types (neutrophils, microglia/macrophages, and astroglia) at the injury zone. To determine whether trovafloxacin exerted these effects by direct action on immune cells, we evaluated its effect on ATP release and cell migration using a chemotactic-stimulated microglial cell line. We found that trovafloxacin significantly inhibited both ATP release and migration of these cells. Conclusion Our results show that trovafloxacin administration has pronounced anti-inflammatory and neuroprotective effects following brain injury. These findings lay the foundation for future studies to directly test a role for Panx1 channels in pathological inflammation following brain trauma. Electronic supplementary material The online version of this article (10.1186/s12974-018-1069-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Charu Garg
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Joon Ho Seo
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Jayalakshmi Ramachandran
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Ji Meng Loh
- Department of Mathematical Sciences, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| | - Frances Calderon
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, 185 South Orange Ave, Newark, NJ, 07103, USA.
| | - Jorge E Contreras
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, 185 South Orange Ave, Newark, NJ, 07103, USA.
| |
Collapse
|
33
|
Weaver RJ, Betts C, Blomme EAG, Gerets HHJ, Gjervig Jensen K, Hewitt PG, Juhila S, Labbe G, Liguori MJ, Mesens N, Ogese MO, Persson M, Snoeys J, Stevens JL, Walker T, Park BK. Test systems in drug discovery for hazard identification and risk assessment of human drug-induced liver injury. Expert Opin Drug Metab Toxicol 2017; 13:767-782. [PMID: 28604124 DOI: 10.1080/17425255.2017.1341489] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The liver is an important target for drug-induced toxicities. Early detection of hepatotoxic drugs requires use of well-characterized test systems, yet current knowledge, gaps and limitations of tests employed remains an important issue for drug development. Areas Covered: The current state of the science, understanding and application of test systems in use for the detection of drug-induced cytotoxicity, mitochondrial toxicity, cholestasis and inflammation is summarized. The test systems highlighted herein cover mostly in vitro and some in vivo models and endpoint measurements used in the assessment of small molecule toxic liabilities. Opportunities for research efforts in areas necessitating the development of specific tests and improved mechanistic understanding are highlighted. Expert Opinion: Use of in vitro test systems for safety optimization will remain a core activity in drug discovery. Substantial inroads have been made with a number of assays established for human Drug-induced Liver Injury. There nevertheless remain significant gaps with a need for improved in vitro tools and novel tests to address specific mechanisms of human Drug-Induced Liver Injury. Progress in these areas will necessitate not only models fit for application, but also mechanistic understanding of how chemical insult on the liver occurs in order to identify translational and quantifiable readouts for decision-making.
Collapse
Affiliation(s)
- Richard J Weaver
- a Research & Biopharmacy, Institut de Recherches Internationales Servier , Suresnes , France
| | - Catherine Betts
- b Pathology Sciences, Drug Safety and Metabolism , AstraZeneca R&D , Cambridge , UK
| | | | - Helga H J Gerets
- d Non Clinical Development, Chemin du Foriest , UCB BioPharma SPRL , Braine L'Alleud , Belgium
| | | | - Philip G Hewitt
- f Non-Clinical Development, Merck KGaA , Darmstadt , Germany
| | - Satu Juhila
- g In Vitro Biology , Orion Pharma , Espoo , Finland
| | - Gilles Labbe
- h Investigative Toxicology, Preclinical Safety , Sanofi R&D , Paris , France
| | | | - Natalie Mesens
- i Preclinical Development & Safety, Janssen (Pharmaceutical Companies of Johnson & Johnson) Turnhoutseweg 30 , Beerse , Belgium
| | - Monday O Ogese
- j Pathology Sciences, Drug Safety and Metabolism , AstraZeneca R&D , Cambridge , UK
| | - Mikael Persson
- k Innovative Medicines and Early Clinical Development, Drug Safety and Metabolism, Discovery Safety , AstraZeneca R&D , Mölndal , Sweden
| | - Jan Snoeys
- l Pharmacokinetics Dynamics & Metabolism, Janssen (Pharmaceutical Companies of Johnson & Johnson) Turnhoutseweg 30 , Beerse , Belgium
| | - James L Stevens
- m Dept of Toxicology , Lilly Research Laboratories, Eli Lilly and Company , Indianapolis , Indiana , USA
| | - Tracy Walker
- n Investigative Safety & Drug Metabolism , GlaxoSmithKline, David Jack Centre for Research and Development , Ware , Herts , Hertfordshire, UK
| | - B Kevin Park
- o Institute of Translational Medicine , University of Liverpool , Liverpool , UK
| |
Collapse
|
34
|
He L, Yin P, Meng Y, Tang J, He T, Niu M, Guo Y, Zhu Y, Jing J, Li C, Ma Z, Wang J, Bai Z, Xiao X. Immunological synergistic mechanisms of trans-/cis-stilbene glycosides in Heshouwu-related idiosyncratic liver injury. Sci Bull (Beijing) 2017; 62:748-751. [PMID: 36659266 DOI: 10.1016/j.scib.2017.04.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/26/2017] [Accepted: 04/11/2017] [Indexed: 01/21/2023]
Affiliation(s)
- Lanzhi He
- China Military Institute of Chinese Medicine, 302 Military Hospital, Beijing 100039, China; School of Pharmacy, Hunan University of Traditional Chinese Medicine, Changsha 410208, China
| | - Ping Yin
- China Military Institute of Chinese Medicine, 302 Military Hospital, Beijing 100039, China
| | - Yakun Meng
- China Military Institute of Chinese Medicine, 302 Military Hospital, Beijing 100039, China
| | - Jinfa Tang
- China Military Institute of Chinese Medicine, 302 Military Hospital, Beijing 100039, China
| | - Tingting He
- Integrative Medical Center, 302 Military Hospital, Beijing 100039, China
| | - Ming Niu
- China Military Institute of Chinese Medicine, 302 Military Hospital, Beijing 100039, China
| | - Yuming Guo
- China Military Institute of Chinese Medicine, 302 Military Hospital, Beijing 100039, China
| | - Yun Zhu
- China Military Institute of Chinese Medicine, 302 Military Hospital, Beijing 100039, China
| | - Jing Jing
- China Military Institute of Chinese Medicine, 302 Military Hospital, Beijing 100039, China
| | - Chunyu Li
- China Military Institute of Chinese Medicine, 302 Military Hospital, Beijing 100039, China
| | - Zhijie Ma
- China Military Institute of Chinese Medicine, 302 Military Hospital, Beijing 100039, China
| | - Jiabo Wang
- China Military Institute of Chinese Medicine, 302 Military Hospital, Beijing 100039, China.
| | - Zhaofang Bai
- China Military Institute of Chinese Medicine, 302 Military Hospital, Beijing 100039, China.
| | - Xiaohe Xiao
- Integrative Medical Center, 302 Military Hospital, Beijing 100039, China.
| |
Collapse
|
35
|
Granitzny A, Knebel J, Müller M, Braun A, Steinberg P, Dasenbrock C, Hansen T. Evaluation of a human in vitro hepatocyte-NPC co-culture model for the prediction of idiosyncratic drug-induced liver injury: A pilot study. Toxicol Rep 2017; 4:89-103. [PMID: 28959630 PMCID: PMC5615103 DOI: 10.1016/j.toxrep.2017.02.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 02/07/2017] [Indexed: 12/16/2022] Open
Abstract
Co-cultures of liver and immune cells can be used to detect iDILI compounds. Pro-inflammatory factors are involved in the development of iDILI. The co-exposure of a drug candidate with TNF might be sufficient to predict iDILI.
Interactions between hepatocytes and immune cells as well as inflammatory episodes are frequently discussed to play a critical role in the alteration of the individual susceptibility to idiosyncratic drug-induced liver injury (iDILI). To evaluate this hypothesis and to face the urgent need for predictive in vitro models, we established two co-culture systems based on two human cell lines in presence or absence of pro-inflammatory factors (LPS, TNF), i.e. hepatoma HepG2 cells co-cultured with monocytic or macrophage-like THP-1 cells. HepG2 monocultures served as control scenario. Mono- or co-cultures were treated with iDILI reference substances (Troglitazone [TGZ], Trovafloxacin [TVX], Diclofenac [DcL], Ketoconazole [KC]) or their non-iDILI partner compounds (Rosiglitazone, Levofloxacin, Acetylsalicylic Acid, Fluconazole). The liver cell viability was subsequently determined via WST-Assay. An enhanced cytotoxicity (synergy) or a hormetic response compared to the drug effect in the HepG2 monoculture was considered as iDILI positive. TGZ synergized in co-cultures with monocytes without an additional pro-inflammatory stimulus, while DcL and KC showed a hormetic response. All iDILI drugs synergized with TNF in the simple HepG2 monoculture, indicating its relevance as an initiator of iDILI. KC showed a synergy when co-exposed to both, monocytes and LPS, while TVX and DcL showed a synergy under the same conditions with macrophages. All described iDILI responses were not observed with the corresponding non-iDILI partner compounds. Our first results confirm that an inflammatory environment increases the sensitivity of liver cells towards iDILI compounds and point to an involvement of pro-inflammatory factors, especially TNF, in the development of iDILI.
Collapse
Key Words
- CD, cluster of differentiation
- Co-culture model
- DAMP, damage-associated molecular pattern
- Drug-induced liver injury
- EC, effective concentration
- EpCAM, epithelial cellular adhesion molecule
- HSP, heat shock protein
- Idiosyncratic
- Inflammation
- JNK, c-Jun N-terminal kinase
- LPS, bacterial lipopolysaccharide
- NF-κB, nuclear factor kappa B
- NPC, non-parenchymal cell
- NSAID, nonsteriodal anti-inflammatory drug
- PAMP, pathogen-associated molecular pattern
- Preclinical research
- SD, standard deviation
- TNF, tumor necrosis factor
- iDILI, idiosyncratic drug-induced liver injury
Collapse
Affiliation(s)
- Anne Granitzny
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
| | - Jan Knebel
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
| | - Meike Müller
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
| | - Armin Braun
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Member of the German Center for Lung Research (DZL), Biomedical Research in End stage and Obstructive Lung Disease (BREATH) research network, Member of the Cluster of Excellence Regenerative Biology to Reconstructive Therapy (REBIRTH), Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
| | - Pablo Steinberg
- University of Veterinary Medicine Hannover (TiHo), Institute for Food Toxicology and Analytical Chemistry, Bischofsholer Damm 15, 30173 Hannover, Germany
| | - Clemens Dasenbrock
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
| | - Tanja Hansen
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
- Corresponding author at: Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Department of In vitro and Mechanistic Toxicology, Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany.Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM)Nikolai-Fuchs-Straße 1Hannover30625Germany
| |
Collapse
|
36
|
Massart J, Begriche K, Moreau C, Fromenty B. Role of nonalcoholic fatty liver disease as risk factor for drug-induced hepatotoxicity. J Clin Transl Res 2017; 3:212-232. [PMID: 28691103 PMCID: PMC5500243 DOI: 10.18053/jctres.03.2017s1.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Obesity is often associated with nonalcoholic fatty liver disease (NAFLD), which refers to a large spectrum of hepatic lesions including fatty liver, nonalcoholic steatohepatitis (NASH) and cirrhosis. Different investigations showed or suggested that obesity and NAFLD are able to increase the risk of hepatotoxicity of different drugs. Some of these drugs could induce more frequently an acute hepatitis in obese individuals whereas others could worsen pre-existing NAFLD. AIM The main objective of the present review was to collect the available information regarding the role of NAFLD as risk factor for drug-induced hepatotoxicity. For this purpose, we performed a data-mining analysis using different queries including drug-induced liver injury (or DILI), drug-induced hepatotoxicity, fatty liver, nonalcoholic fatty liver disease (or NAFLD), steatosis and obesity. The main data from the collected articles are reported in this review and when available, some pathophysiological hypotheses are put forward. RELEVANCE FOR PATIENTS Drugs that could pose a potential risk in obese patients include compounds belonging to different pharmacological classes such as acetaminophen, halothane, methotrexate, rosiglitazone, stavudine and tamoxifen. For some of these drugs, experimental investigations in obese rodents confirmed the clinical observations and unveiled different pathophysiological mechanisms which could explain why these pharmaceuticals are particularly hepatotoxic in obesity and NAFLD. Other drugs such as pentoxifylline, phenobarbital and omeprazole might also pose a risk but more investigations are required to determine whether this risk is significant or not. Because obese people often take several drugs for the treatment of different obesity-related diseases such as type 2 diabetes, hyperlipidemia and coronary heart disease, it is urgent to identify the main pharmaceuticals that can cause acute hepatitis on a fatty liver background or induce NAFLD worsening.
Collapse
Affiliation(s)
- Julie Massart
- Department of Molecular Medicine and Surgery, Karolinska University Hospital, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | - Caroline Moreau
- INSERM, U991, Université de Rennes 1, Rennes, France.,Service de Biochimie et Toxicologie, CHU Pontchaillou, Rennes, France
| | | |
Collapse
|
37
|
Roth RA, Maiuri AR, Ganey PE. Idiosyncratic Drug-Induced Liver Injury: Is Drug-Cytokine Interaction the Linchpin? J Pharmacol Exp Ther 2017; 360:461-470. [PMID: 28104833 DOI: 10.1124/jpet.116.237578] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/11/2016] [Indexed: 12/11/2022] Open
Abstract
Idiosyncratic drug-induced liver injury continues to be a human health problem in part because drugs that cause these reactions are not identified in current preclinical testing and because progress in prevention is hampered by incomplete knowledge of mechanisms that underlie these adverse responses. Several hypotheses involving adaptive immune responses, inflammatory stress, inability to adapt to stress, and multiple, concurrent factors have been proposed. Yet much remains unknown about how drugs interact with the liver to effect death of hepatocytes. Evidence supporting hypotheses implicating adaptive or innate immune responses in afflicted patients has begun to emerge and is bolstered by results obtained in experimental animal models and in vitro systems. A commonality in adaptive and innate immunity is the production of cytokines, including interferon-γ (IFNγ). IFNγ initiates cell signaling pathways that culminate in cell death or inhibition of proliferative repair. Tumor necrosis factor-α, another cytokine prominent in immune responses, can also promote cell death. Furthermore, tumor necrosis factor-α interacts with IFNγ, leading to enhanced cellular responses to each cytokine. In this short review, we propose that the interaction of drugs with these cytokines contributes to idiosyncratic drug-induced liver injury, and mechanisms by which this could occur are discussed.
Collapse
Affiliation(s)
- Robert A Roth
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Ashley R Maiuri
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Patricia E Ganey
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| |
Collapse
|
38
|
Hirashima R, Itoh T, Tukey RH, Fujiwara R. Prediction of drug-induced liver injury using keratinocytes. J Appl Toxicol 2017; 37:863-872. [PMID: 28138970 DOI: 10.1002/jat.3435] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 12/06/2016] [Accepted: 12/12/2016] [Indexed: 01/07/2023]
Abstract
Drug-induced liver injury (DILI) is one of the most common adverse drug reactions. DILI is often accompanied by skin reactions, including rash and pruritus. However, it is still unknown whether DILI-associated genes such as S100 calcium-binding protein A and interleukin (IL)-1β are involved in drug-induced skin toxicity. In the present study, most of the tested hepatotoxic drugs such as pioglitazone and diclofenac induced DILI-associated genes in human and mouse keratinocytes. Keratinocytes of mice at higher risk for DILI exhibited an increased IL-1β basal expression. They also showed a higher inducibility of IL-1β when treated by pioglitazone. Mice at higher risk for DILI showed even higher sums of DILI-associated gene basal expression levels and induction rates in keratinocytes. Our data suggest that DILI-associated genes might be involved in the onset and progression of drug-induced skin toxicity. Furthermore, we might be able to identify individuals at higher risk of developing DILI less invasively by examining gene expression patterns in keratinocytes. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Rika Hirashima
- School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Tomoo Itoh
- School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Robert H Tukey
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Ryoichi Fujiwara
- School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| |
Collapse
|
39
|
Sistare FD, Mattes WB, LeCluyse EL. The Promise of New Technologies to Reduce, Refine, or Replace Animal Use while Reducing Risks of Drug Induced Liver Injury in Pharmaceutical Development. ILAR J 2017; 57:186-211. [DOI: 10.1093/ilar/ilw025] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 07/25/2016] [Accepted: 09/13/2016] [Indexed: 12/19/2022] Open
|
40
|
Rose KA, Holman NS, Green AM, Andersen ME, LeCluyse EL. Co-culture of Hepatocytes and Kupffer Cells as an In Vitro Model of Inflammation and Drug-Induced Hepatotoxicity. J Pharm Sci 2016; 105:950-964. [PMID: 26869439 DOI: 10.1016/s0022-3549(15)00192-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/10/2015] [Accepted: 11/17/2015] [Indexed: 12/17/2022]
Abstract
Immune-mediated drug-induced hepatotoxicity is often unrecognized as a potential mode of action due to the lack of appropriate in vitro models. We have established an in vitro rat donor-matched hepatocyte and Kupffer cell co-culture (HKCC) model to study immune-related responses to drug exposure. Optimal cell culture conditions were identified for the maintenance of co-cultures based on cell longevity, monolayer integrity, and cytokine response after lipopolysaccharide (LPS) exposure. Hepatocyte monocultures and HKCCs were then used to test a subset of compounds associated with hepatotoxic effects with or without LPS. Cytokine levels and metabolic activity (cytochrome P450 3A [Cyp3A]) were measured after a 48-h exposure to monitor endotoxin-induced changes in acute phase and functional end points. LPS-activated HKCCs, but not hepatocyte monocultures, treated with trovafloxacin or acetaminophen, compounds associated with immune-mediated hepatotoxicity, showed LPS-dependent decreases in interleukin-6 production with concomitant increases in Cyp3A activity. Differential endotoxin- and model-dependent alterations were observed in cytokine profiles and Cyp3A activity levels that corresponded to specific compounds. These results indicate the utility of the HKCC model system to discern compound-specific effects that may lead to enhanced or mitigate hepatocellular injury due to innate or adaptive immune responses.
Collapse
Affiliation(s)
- Kelly A Rose
- The Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709
| | - Natalie S Holman
- The Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709; The Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514
| | - Angela M Green
- The Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709
| | - Melvin E Andersen
- The Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709
| | - Edward L LeCluyse
- The Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709; The Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514.
| |
Collapse
|
41
|
Yao M, Gao W, Tao H, Yang J, Huang T. The regulation effects of danofloxacin on pig immune stress induced by LPS. Res Vet Sci 2016; 110:65-71. [PMID: 28159239 DOI: 10.1016/j.rvsc.2016.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 11/03/2016] [Accepted: 11/05/2016] [Indexed: 01/01/2023]
Abstract
Danofloxacin (DAN) is one of the Fluoroquinolone drugs (FQs) that has been widely used in the control and prevention of bacterial infectious disease in animal production. Most of the FQs have an obvious protective effect against lipopolysaccharide (LPS) induced Immune stress. However, the effect of DAN on the host immune system of animals remains unknown. In this study, a fever piglet model was built and a systematic survey of the response of inflammatory genes and mediators to DAN treatment and LPS induction was performed in the pig. The results indicated that the IL-1β, TNF-α, IL-6, NO (nitric oxide), and PGE2 (prostaglandin E2) levels were significantly suppressed in plasma and porcine alveolar macrophage 3D4/2 cells compared with the LPS treatment group. Interestingly, the IL-10 production was further stimulated by DAN treatment in the LPS challenged piglet. Our results suggested that DAN could relieve acute inflammation through inhibiting the activation of inflammatory genes introduced by LPS.
Collapse
Affiliation(s)
- Min Yao
- College of Animal Science, Yangtze University, Jingzhou, Hubei 434025, China
| | - Weihua Gao
- College of Animal Science, Yangtze University, Jingzhou, Hubei 434025, China
| | - Hengxun Tao
- College of Animal Science, Yangtze University, Jingzhou, Hubei 434025, China
| | - Jun Yang
- College of Animal Science, Yangtze University, Jingzhou, Hubei 434025, China
| | - Tinghua Huang
- College of Animal Science, Yangtze University, Jingzhou, Hubei 434025, China.
| |
Collapse
|
42
|
Mitsugi R, Itoh T, Fujiwara R. MicroRNA-877-5p is involved in the trovafloxacin-induced liver injury. Toxicol Lett 2016; 263:34-43. [PMID: 27713024 DOI: 10.1016/j.toxlet.2016.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/26/2016] [Accepted: 10/02/2016] [Indexed: 12/21/2022]
Abstract
Trovafloxacin develops severe hepatotoxicity; however, the underlying mechanism of the trovafloxacin-induced liver injury has not been cleared. It has been shown that microRNAs (miRNAs) can be involved in the development of drug-induced liver injuries. We performed a miRNA microarray analysis to identify hepatic miRNAs that were induced or reduced by trovafloxacin in mice. It was demonstrated that miR-877-5p was the most increased miRNA in the mouse liver 24h after the trovafloxacin administration. To investigate the role of miR-877-5p in the liver, we established miR-877-5p-overexpressed HepG2 cells. Microarray analysis detected altered expressions in 2077 (>2-fold) and 1547 (<0.5-fold) genes in the miR-877-5p overexpressing cells compared to the mock cells. Especially, SLCO4C1, PEPCK, MT1M, HIST1H2BM, LGI1, and PLA2G2A were markedly increased or decreased in the miR-877-5p overexpressing cells. We conducted a correlation analysis between the expression levels of miR-877-5p and the six genes in eight miR-877-5p stably-expressed clones. It was shown that the PEPCK expression levels were correlated with miR-877-5p expression levels. PEPCK is associated with development of apoptotic cell death; therefore, the increased miR- 877-5p-induced PEPCK can be a trigger that is involved in the development of trovafloxacin-induced liver injury.
Collapse
Affiliation(s)
- Ryo Mitsugi
- School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Tomoo Itoh
- School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Ryoichi Fujiwara
- School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan.
| |
Collapse
|
43
|
Role of Inflammatory and Oxidative Stress, Cytochrome P450 2E1, and Bile Acid Disturbance in Rat Liver Injury Induced by Isoniazid and Lipopolysaccharide Cotreatment. Antimicrob Agents Chemother 2016; 60:5285-93. [PMID: 27324775 DOI: 10.1128/aac.00854-16] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/11/2016] [Indexed: 12/22/2022] Open
Abstract
Isoniazid (INH) remains the core drug in tuberculosis management, but serious hepatotoxicity and potentially fatal liver injury continue to accompany INH consumption. Among numerous theories that have been established to explain INH-induced liver injury, an inflammatory stress theory has recently been widely used to explain the idiosyncrasy. Inflammatory stress usually sensitizes tissues to a drug's toxic consequences. Therefore, the present study was conducted to verify whether bacterial lipopolysaccharide (LPS)-induced inflammation may have a role in enhancing INH hepatotoxicity. While single INH or LPS administration showed no major toxicity signs, INH-LPS cotreatment intensified liver toxicity. Both blood biomarkers and histological evaluations clearly showed positive signs of severe liver damage accompanied by massive necrosis, inflammatory infiltration, and hepatic steatosis. Furthermore, elevated serum levels of bile acid associated with the repression of bile acid synthesis and transport regulatory parameters were observed. Moreover, the principal impact of cytochrome P450 2E1 (CYP2E1) on INH toxicity could be anticipated, as its protein expression showed enormous increases in INH-LPS-cotreated animals. Furthermore, the crucial role of CYP2E1 in the production of reactive oxygen species (ROS) was clearly obvious in the repression of hepatic antioxidant parameters. In summary, these results confirmed that this LPS-induced inflammation model might prove valuable in revealing the hepatotoxic mechanisms of INH and the crucial role played by CYP2E1 in the initiation and propagation of INH-induced liver damage, information which could be very useful to clinicians in understanding the pathogenesis of drug-induced liver injury.
Collapse
|
44
|
Nguyen DG, Funk J, Robbins JB, Crogan-Grundy C, Presnell SC, Singer T, Roth AB. Bioprinted 3D Primary Liver Tissues Allow Assessment of Organ-Level Response to Clinical Drug Induced Toxicity In Vitro. PLoS One 2016; 11:e0158674. [PMID: 27387377 PMCID: PMC4936711 DOI: 10.1371/journal.pone.0158674] [Citation(s) in RCA: 266] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 06/20/2016] [Indexed: 12/30/2022] Open
Abstract
Modeling clinically relevant tissue responses using cell models poses a significant challenge for drug development, in particular for drug induced liver injury (DILI). This is mainly because existing liver models lack longevity and tissue-level complexity which limits their utility in predictive toxicology. In this study, we established and characterized novel bioprinted human liver tissue mimetics comprised of patient-derived hepatocytes and non-parenchymal cells in a defined architecture. Scaffold-free assembly of different cell types in an in vivo-relevant architecture allowed for histologic analysis that revealed distinct intercellular hepatocyte junctions, CD31+ endothelial networks, and desmin positive, smooth muscle actin negative quiescent stellates. Unlike what was seen in 2D hepatocyte cultures, the tissues maintained levels of ATP, Albumin as well as expression and drug-induced enzyme activity of Cytochrome P450s over 4 weeks in culture. To assess the ability of the 3D liver cultures to model tissue-level DILI, dose responses of Trovafloxacin, a drug whose hepatotoxic potential could not be assessed by standard pre-clinical models, were compared to the structurally related non-toxic drug Levofloxacin. Trovafloxacin induced significant, dose-dependent toxicity at clinically relevant doses (≤ 4uM). Interestingly, Trovafloxacin toxicity was observed without lipopolysaccharide stimulation and in the absence of resident macrophages in contrast to earlier reports. Together, these results demonstrate that 3D bioprinted liver tissues can both effectively model DILI and distinguish between highly related compounds with differential profile. Thus, the combination of patient-derived primary cells with bioprinting technology here for the first time demonstrates superior performance in terms of mimicking human drug response in a known target organ at the tissue level.
Collapse
Affiliation(s)
- Deborah G. Nguyen
- Organovo Holdings Inc., San Diego, CA, United States of America
- * E-mail:
| | - Juergen Funk
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | | | | | | | - Thomas Singer
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Adrian B. Roth
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| |
Collapse
|
45
|
Chitohexaose protects against acetaminophen-induced hepatotoxicity in mice. Cell Death Dis 2016; 7:e2224. [PMID: 27171266 PMCID: PMC4917664 DOI: 10.1038/cddis.2016.131] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 04/05/2016] [Accepted: 04/07/2016] [Indexed: 12/11/2022]
Abstract
Acetaminophen (N-acetyl-para-aminophenol (APAP)) toxicity causes acute liver failure by inducing centrilobular hepatic damage as a consequence of mitochondrial oxidative stress. Sterile inflammation, triggered by hepatic damage, facilitates gut bacterial translocation leading to systemic inflammation; TLR4-mediated activation by LPS has been shown to have a critical role in APAP-mediated hepatotoxicity. In this study, we demonstrate significant protection mediated by chitohexaose (Chtx) in mice challenged with a lethal dose of APAP (400 mg/kg b.w.). Decreased mortality by Chtx was associated with reduced hepatic damage, increased peritoneal migration of neutrophils, decreased mRNA expression of IL-1β as well as inhibition of inflammasome activation in liver. Further, an alternate mouse model of co-administration of a sublethal doses of APAP (200 mg/kg b.w.) and LPS (5 mg/kg b.w.) operating synergistically and mediating complete mortality was developed. Overwhelming inflammation, characterized by increased inflammatory cytokines (TNF-α, IL-1β and so on) in liver as well as in circulation and mortality was demonstrable in this model. Also, Chtx administration mediated significant reversal of mortality in APAP+LPS co-administered mice, which was associated with reduced IL-1β in liver and plasma cytokines in this model. In conclusion, Chtx being a small molecular weight linear carbohydrate offers promise for clinical management of liver failure associated with APAP overdose.
Collapse
|
46
|
Bell CC, Hendriks DFG, Moro SML, Ellis E, Walsh J, Renblom A, Fredriksson Puigvert L, Dankers ACA, Jacobs F, Snoeys J, Sison-Young RL, Jenkins RE, Nordling Å, Mkrtchian S, Park BK, Kitteringham NR, Goldring CEP, Lauschke VM, Ingelman-Sundberg M. Characterization of primary human hepatocyte spheroids as a model system for drug-induced liver injury, liver function and disease. Sci Rep 2016; 6:25187. [PMID: 27143246 PMCID: PMC4855186 DOI: 10.1038/srep25187] [Citation(s) in RCA: 494] [Impact Index Per Article: 54.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 04/12/2016] [Indexed: 12/11/2022] Open
Abstract
Liver biology and function, drug-induced liver injury (DILI) and liver diseases are difficult to study using current in vitro models such as primary human hepatocyte (PHH) monolayer cultures, as their rapid de-differentiation restricts their usefulness substantially. Thus, we have developed and extensively characterized an easily scalable 3D PHH spheroid system in chemically-defined, serum-free conditions. Using whole proteome analyses, we found that PHH spheroids cultured this way were similar to the liver in vivo and even retained their inter-individual variability. Furthermore, PHH spheroids remained phenotypically stable and retained morphology, viability, and hepatocyte-specific functions for culture periods of at least 5 weeks. We show that under chronic exposure, the sensitivity of the hepatocytes drastically increased and toxicity of a set of hepatotoxins was detected at clinically relevant concentrations. An interesting example was the chronic toxicity of fialuridine for which hepatotoxicity was mimicked after repeated-dosing in the PHH spheroid model, not possible to detect using previous in vitro systems. Additionally, we provide proof-of-principle that PHH spheroids can reflect liver pathologies such as cholestasis, steatosis and viral hepatitis. Combined, our results demonstrate that the PHH spheroid system presented here constitutes a versatile and promising in vitro system to study liver function, liver diseases, drug targets and long-term DILI.
Collapse
Affiliation(s)
- Catherine C Bell
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm, Sweden
| | - Delilah F G Hendriks
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm, Sweden
| | - Sabrina M L Moro
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm, Sweden
| | - Ewa Ellis
- Department of Clinical Science, Intervention and Technology, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Joanne Walsh
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Sherrington Buildings, Ashton Street, University of Liverpool, UK
| | - Anna Renblom
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm, Sweden
| | - Lisa Fredriksson Puigvert
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm, Sweden
| | - Anita C A Dankers
- Janssen Pharmaceutical Companies of Johnson &Johnson, Department of Pharmacokinetics, Dynamics and Metabolism, Beerse, Belgium
| | - Frank Jacobs
- Janssen Pharmaceutical Companies of Johnson &Johnson, Department of Pharmacokinetics, Dynamics and Metabolism, Beerse, Belgium
| | - Jan Snoeys
- Janssen Pharmaceutical Companies of Johnson &Johnson, Department of Pharmacokinetics, Dynamics and Metabolism, Beerse, Belgium
| | - Rowena L Sison-Young
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Sherrington Buildings, Ashton Street, University of Liverpool, UK
| | - Rosalind E Jenkins
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Sherrington Buildings, Ashton Street, University of Liverpool, UK
| | - Åsa Nordling
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm, Sweden
| | - Souren Mkrtchian
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm, Sweden
| | - B Kevin Park
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Sherrington Buildings, Ashton Street, University of Liverpool, UK
| | - Neil R Kitteringham
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Sherrington Buildings, Ashton Street, University of Liverpool, UK
| | - Christopher E P Goldring
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Sherrington Buildings, Ashton Street, University of Liverpool, UK
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
47
|
Kwast L, Fiechter D, Kruijssen L, Bleumink R, Ludwig I, Bol-Schoenmakers M, Smit J, Pieters R. Oral exposure to immunostimulating drugs results in early changes in innate immune parameters in the spleen. J Immunotoxicol 2016; 13:535-47. [PMID: 27043250 DOI: 10.3109/1547691x.2016.1139643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The development of immune-dependent drug hypersensitivity reactions (IDHR) is likely to involve activation of the innate immune system to stimulate neo-antigen specific T-cells. Previously it has been shown that, upon oral exposure to several drugs with immune-adjuvant capacity, mice developed T-cell-dependent responses to TNP-OVA. These results were indicative of the adjuvant potential of these drugs. The present study set out to evaluate the nature of this adjuvant potential by focusing on early immune changes in the spleen, by testing several drugs in the same experimental model. Mice were exposed to one or multiple oral doses of previously-tested drugs: the non-steroidal-anti-inflammatory drug (NSAID) diclofenac (DF), the analgesic acetaminophen (APAP), the anti-epileptic drug carbamazepine (CMZ) or the antibiotic ofloxacin (OFLX). Within 24 h after the final dosing, early innate and also adaptive immune parameters in the spleen were examined. In addition, liver tissue was also evaluated for damage. Exposure to APAP resulted in severe liver damage, increased levels of serum alanine aminotransferase (ALT) and local MIP-2 expression. DF exposure did not cause visible liver damage, but did increase liver weight. DF also elicited clear effects on splenic innate and adaptive immune cells, i.e. increased levels of NK cells and memory T-cells. Furthermore, an increase in plasma MIP-2 levels combined with an influx of neutrophils into the spleen was observed. OFLX and CMZ exposure resulted in increased liver weights, MIP-2 expression and up-regulation of co-stimulatory molecules on antigen-presenting cells (APC). The data suggested that multiple immune parameters were altered upon exposure to drugs known to elicit immunosensitization and that broad evaluation of immune changes in straightforward short-term animal models is needed to determine whether a drug may harbor the hazard to induce IDHR. The oral exposure approach as used here may be applied in the future as an immunotoxicological research tool in this type of evaluation.
Collapse
Affiliation(s)
- Lydia Kwast
- a Division of Toxicology , Institute for Risk Assessment Sciences, Utrecht University , Utrecht , the Netherlands ;,b TI Pharma , Leiden , the Netherlands
| | - Daniëlle Fiechter
- a Division of Toxicology , Institute for Risk Assessment Sciences, Utrecht University , Utrecht , the Netherlands ;,b TI Pharma , Leiden , the Netherlands
| | - Laura Kruijssen
- a Division of Toxicology , Institute for Risk Assessment Sciences, Utrecht University , Utrecht , the Netherlands ;,b TI Pharma , Leiden , the Netherlands
| | - Rob Bleumink
- a Division of Toxicology , Institute for Risk Assessment Sciences, Utrecht University , Utrecht , the Netherlands
| | - Irene Ludwig
- a Division of Toxicology , Institute for Risk Assessment Sciences, Utrecht University , Utrecht , the Netherlands ;,b TI Pharma , Leiden , the Netherlands
| | - Marianne Bol-Schoenmakers
- a Division of Toxicology , Institute for Risk Assessment Sciences, Utrecht University , Utrecht , the Netherlands
| | - Joost Smit
- a Division of Toxicology , Institute for Risk Assessment Sciences, Utrecht University , Utrecht , the Netherlands
| | - Raymond Pieters
- a Division of Toxicology , Institute for Risk Assessment Sciences, Utrecht University , Utrecht , the Netherlands
| |
Collapse
|
48
|
Yan H, Endo Y, Shen Y, Rotstein D, Dokmanovic M, Mohan N, Mukhopadhyay P, Gao B, Pacher P, Wu WJ. Ado-Trastuzumab Emtansine Targets Hepatocytes Via Human Epidermal Growth Factor Receptor 2 to Induce Hepatotoxicity. Mol Cancer Ther 2016; 15:480-490. [PMID: 26712117 DOI: 10.1158/1535-7163.mct-15-0580] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 12/04/2015] [Indexed: 11/16/2022]
Abstract
Ado-trastuzumab emtansine (T-DM1) is an antibody-drug conjugate (ADC) approved for the treatment of HER2-positive metastatic breast cancer. It consists of trastuzumab, a humanized mAb directed against HER2, and a microtubule inhibitor, DM1, conjugated to trastuzumab via a thioether linker. Hepatotoxicity is one of the serious adverse events associated with T-DM1 therapy. Mechanisms underlying T-DM1-induced hepatotoxicity remain elusive. Here, we use hepatocytes and mouse models to investigate the mechanisms of T-DM1-induced hepatotoxicity. We show that T-DM1 is internalized upon binding to cell surface HER2 and is colocalized with LAMP1, resulting in DM1-associated cytotoxicity, including disorganized microtubules, nuclear fragmentation/multiple nuclei, and cell growth inhibition. We further demonstrate that T-DM1 treatment significantly increases the serum levels of aspartate aminotransferase, alanine aminotransferase, and lactate dehydrogenase in mice and induces inflammation and necrosis in liver tissues, and that T-DM1-induced hepatotoxicity is dose dependent. Moreover, the gene expression of TNFα in liver tissues is significantly increased in mice treated with T-DM1 as compared with those treated with trastuzumab or vehicle. We propose that T-DM1-induced upregulation of TNFα enhances the liver injury that may be initially caused by DM1-mediated intracellular damage. Our proposal is underscored by the fact that T-DM1 induces the outer mitochondrial membrane rupture, a typical morphologic change in the mitochondrial-dependent apoptosis, and mitochondrial membrane potential dysfunction. Our work provides mechanistic insights into T-DM1-induced hepatotoxicity, which may yield novel strategies to manage liver injury induced by T-DM1 or other ADCs.
Collapse
Affiliation(s)
- Haoheng Yan
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland. Interagency Oncology Task Force Fellowship: Oncology Product Research/Review Fellow, NCI, Bethesda, Maryland
| | - Yukinori Endo
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Yi Shen
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - David Rotstein
- Division of Compliance, Office of Surveillance and Compliance, Center for Veterinary Medicine, U.S. Food and Drug Administration, Derwood, Maryland
| | - Milos Dokmanovic
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Nishant Mohan
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Partha Mukhopadhyay
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
| | - Pal Pacher
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland
| | - Wen Jin Wu
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland.
| |
Collapse
|
49
|
Chen M, Suzuki A, Borlak J, Andrade RJ, Lucena MI. Drug-induced liver injury: Interactions between drug properties and host factors. J Hepatol 2015; 63:503-14. [PMID: 25912521 DOI: 10.1016/j.jhep.2015.04.016] [Citation(s) in RCA: 262] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/01/2015] [Accepted: 04/07/2015] [Indexed: 12/13/2022]
Abstract
Idiosyncratic drug-induced liver injury (DILI) is a common cause for drug withdrawal from the market and although infrequent, DILI can result in serious clinical outcomes including acute liver failure and the need for liver transplantation. Eliminating the iatrogenic "harm" caused by a therapeutic intent is a priority in patient care. However, identifying culprit drugs and individuals at risk for DILI remains challenging. Apart from genetic factors predisposing individuals at risk, the role of the drugs' physicochemical and toxicological properties and their interactions with host and environmental factors need to be considered. The influence of these factors on mechanisms involved in DILI is multi-layered. In this review, we summarize current knowledge on 1) drug properties associated with hepatotoxicity, 2) host factors considered to modify an individuals' risk for DILI and clinical phenotypes, and 3) drug-host interactions. We aim at clarifying knowledge gaps needed to be filled in as to improve risk stratification in patient care. We therefore broadly discuss relevant areas of future research. Emerging insight will stimulate new investigational approaches to facilitate the discovery of clinical DILI risk modifiers in the context of disease complexity and associated interactions with drug properties, and hence will be able to move towards safety personalized medicine.
Collapse
Affiliation(s)
- Minjun Chen
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, United States
| | - Ayako Suzuki
- Gastroenterology, Central Arkansas Veterans Healthcare System, Little Rock, AR, United States; Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Jürgen Borlak
- Center of Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany
| | - Raúl J Andrade
- Unidad de Gestión Clínica de Enfermedades Digestivas, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain.
| | - M Isabel Lucena
- Unidad de Gestión Clínica de Enfermedades Digestivas, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| |
Collapse
|
50
|
Vernetti LA, Senutovitch N, Boltz R, DeBiasio R, Shun TY, Gough A, Taylor DL. A human liver microphysiology platform for investigating physiology, drug safety, and disease models. Exp Biol Med (Maywood) 2015. [PMID: 26202373 DOI: 10.1177/1535370215592121] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
This paper describes the development and characterization of a microphysiology platform for drug safety and efficacy in liver models of disease that includes a human, 3D, microfluidic, four-cell, sequentially layered, self-assembly liver model (SQL-SAL); fluorescent protein biosensors for mechanistic readouts; as well as a microphysiology system database (MPS-Db) to manage, analyze, and model data. The goal of our approach is to create the simplest design in terms of cells, matrix materials, and microfluidic device parameters that will support a physiologically relevant liver model that is robust and reproducible for at least 28 days for stand-alone liver studies and microfluidic integration with other organs-on-chips. The current SQL-SAL uses primary human hepatocytes along with human endothelial (EA.hy926), immune (U937) and stellate (LX-2) cells in physiological ratios and is viable for at least 28 days under continuous flow. Approximately, 20% of primary hepatocytes and/or stellate cells contain fluorescent protein biosensors (called sentinel cells) to measure apoptosis, reactive oxygen species (ROS) and/or cell location by high content analysis (HCA). In addition, drugs, drug metabolites, albumin, urea and lactate dehydrogenase (LDH) are monitored in the efflux media. Exposure to 180 μM troglitazone or 210 μM nimesulide produced acute toxicity within 2-4 days, whereas 28 μM troglitazone produced a gradual and much delayed toxic response over 21 days, concordant with known mechanisms of toxicity, while 600 µM caffeine had no effect. Immune-mediated toxicity was demonstrated with trovafloxacin with lipopolysaccharide (LPS), but not levofloxacin with LPS. The SQL-SAL exhibited early fibrotic activation in response to 30 nM methotrexate, indicated by increased stellate cell migration, expression of alpha-smooth muscle actin and collagen, type 1, alpha 2. Data collected from the in vitro model can be integrated into a database with access to related chemical, bioactivity, preclinical and clinical information uploaded from external databases for constructing predictive models.
Collapse
Affiliation(s)
- Lawrence A Vernetti
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA University of Pittsburgh Dept. of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Nina Senutovitch
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA University of Pittsburgh Dept. of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Robert Boltz
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA University of Pittsburgh Dept. of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Richard DeBiasio
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Tong Ying Shun
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Albert Gough
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA University of Pittsburgh Dept. of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - D Lansing Taylor
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA University of Pittsburgh Dept. of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|