Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Expert Opin Drug Metab Toxicol ; 18(7-8): 469-481, 2022.
Article in English | MEDLINE | ID: mdl-36003040

ABSTRACT

INTRODUCTION: Idiosyncratic drug-induced liver injury (IDILI) causes morbidity and mortality in patients and leads to curtailed use of efficacious pharmaceuticals. Unlike intrinsically toxic reactions, which depend on dose, IDILI occurs in a minority of patients at therapeutic doses. Much remains unknown about causal links among drug exposure, a mode of action, and liver injury. Consequently, numerous hypotheses about IDILI pathogenesis have arisen. AREAS COVERED: Pharmacokinetic and toxicodynamic characteristics underlying current hypotheses of IDILI etiology are discussed and illustrated graphically. EXPERT OPINION: Hypotheses to explain IDILI etiology all involve alterations in pharmacokinetics, which lead to plasma drug concentrations that rise above a threshold for toxicity, or in toxicodynamics, which result in a lowering of the toxicity threshold. Altered pharmacokinetics arise, for example, from changes in drug metabolism or from transporter polymorphisms. A lowered toxicity threshold can arise from drug-induced mitochondrial injury, accumulation of toxic endogenous factors or harmful immune responses. Newly developed, interactive freeware (DemoTox-PK; https://bit.ly/DemoTox-PK) allows the user to visualize how such alterations might lead to a toxic reaction. The illustrations presented provide a framework for conceptualizing idiosyncratic reactions and could serve as a stimulus for future discussion, education, and research into modes of action of IDILI.


Subject(s)
Chemical and Drug Induced Liver Injury , Chemical and Drug Induced Liver Injury/etiology , Humans , Liver/metabolism
2.
Chem Biol Interact ; 345: 109521, 2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34052195

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (dioxin; TCDD) is an environmental contaminant that elicits a variety of toxic effects, many of which are mediated through activation of the aryl hydrocarbon receptor (AhR). Interaction between AhR and the peroxisome proliferator-activated receptor-alpha (PPAR-α), which regulates fatty acid metabolism, has been suggested. Furthermore, with recognition of the prevalence of inflammatory conditions, there is current interest in the potential for inflammatory stress to modulate the response to environmental agents. The aim of this work was to assess the interaction of TCDD with hepatic inflammation modulated by fenofibrate, a PPAR-α agonist. Female, C57BL/6 mice were treated orally with vehicle or fenofibrate (250 mg/kg) for 13 days, and then were given vehicle or 30 µg/kg TCDD. Four days later, the animals received an i.p. injection of lipopolysaccharide-galactosamine (LPS-GalN) (0.05x107 EU/kg and 500 mg/kg, respectively) to incite inflammation, or saline as vehicle control. After 4 h, the mice were euthanized, and blood and liver samples were collected for analysis. Livers of animals treated with TCDD with or without LPS-GalN had increased lipid deposition, and this effect was blocked by fenofibrate. In TCDD/LPS-GalN-treated mice, fenofibrate caused an increase in plasma activity of alanine aminotransferase, a marker of hepatocellular injury. TCDD reduced LPS-GalN-induced apoptosis, an effect that was prevented by fenofibrate pretreatment. LPS-GalN induced an increase in the concentration of interleukin-6 in plasma and accumulation of neutrophils in liver. TCDD exposure enhanced the former response and inhibited the latter one. These results suggest that fenofibrate counteracts the changes in lipid metabolism induced by TCDD but increases inflammation and liver injury in this model of inflammation-TCDD interaction.


Subject(s)
Dioxins/toxicity , Fatty Liver/drug therapy , Fenofibrate/pharmacology , PPAR alpha/agonists , Alanine Transaminase/blood , Animals , Apoptosis/drug effects , Disease Models, Animal , Drug Interactions , Fatty Liver/blood , Fatty Liver/metabolism , Fatty Liver/pathology , Female , Fenofibrate/therapeutic use , Interleukin-6/blood , Mice , Mice, Inbred C57BL
4.
Expert Opin Drug Metab Toxicol ; 16(6): 475-491, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32324077

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.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Adaptive Immunity/immunology , Animals , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/physiopathology , Cytokines/immunology , Humans , Immunity, Innate/immunology
5.
Int J Toxicol ; 39(2): 151-164, 2020.
Article in English | MEDLINE | ID: mdl-32174281

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.


Subject(s)
Bacterial Infections/immunology , Chemical and Drug Induced Liver Injury/immunology , Hepatitis, Viral, Human/immunology , Liver/immunology , Animals , Cytokines/immunology , Humans , Liver/microbiology , Liver/virology
6.
J Pharmacol Exp Ther ; 362(3): 459-473, 2017 09.
Article in English | MEDLINE | ID: mdl-28687704

ABSTRACT

Idiosyncratic drug-induced liver injury (IDILI) typically occurs in a small fraction of patients and has resulted in removal of otherwise efficacious drugs from the market. Current preclinical testing methods are ineffective in predicting which drug candidates have IDILI liability. Recent results suggest that immune mediators such as tumor necrosis factor-α (TNF) and interferon-γ (IFN) interact with drugs that cause IDILI to kill hepatocytes. This proof-of-concept study was designed to test the hypothesis that drugs can be classified according to their ability to cause IDILI in humans using classification modeling with covariates derived from concentration-response relationships that describe cytotoxic interaction with cytokines. Human hepatoma (HepG2) cells were treated with drugs associated with IDILI or with drugs lacking IDILI liability and cotreated with TNF and/or IFN. Detailed concentration-response relationships were determined for calculation of parameters such as the maximal cytotoxic effect, slope, and EC50 for use as covariates for classification modeling using logistic regression. These parameters were incorporated into multiple classification models to identify combinations of covariates that most accurately classified the drugs according to their association with human IDILI. Of 14 drugs associated with IDILI, almost all synergized with TNF to kill HepG2 cells and were successfully classified by statistical modeling. IFN enhanced the toxicity mediated by some IDILI-associated drugs in the presence of TNF. In contrast, of 10 drugs with little or no IDILI liability, none synergized with inflammatory cytokines to kill HepG2 cells and were classified accordingly. The resulting optimal model classified the drugs with extraordinary selectivity and specificity.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Cytokines/pharmacology , Pharmaceutical Preparations/classification , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Hep G2 Cells , Humans , Interferon-gamma/pharmacology , Logistic Models , ROC Curve , Tumor Necrosis Factor-alpha/pharmacology
7.
J Pharmacol Exp Ther ; 360(2): 461-470, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28104833

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.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Cytokines/metabolism , Pharmaceutical Preparations/metabolism , Animals , Cell Death/drug effects , Chemical and Drug Induced Liver Injury/pathology , Humans , Signal Transduction/drug effects
8.
Toxicol Sci ; 149(2): 372-84, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26609140

ABSTRACT

Diclofenac (DCLF) is a widely used non-steroidal anti-inflammatory drug that is associated with idiosyncratic, drug-induced liver injury (IDILI) in humans. The mechanisms of DCLF-induced liver injury are unknown; however, patients with certain inflammatory diseases have an increased risk of developing IDILI, which raises the possibility that immune mediators play a role in the pathogenesis. DCLF synergizes with the cytokines tumor necrosis factor-alpha (TNF) and interferon-gamma (IFN) to cause hepatocellular apoptosis in vitro by a mechanism that involves activation of the endoplasmic reticulum (ER) stress response pathway and of the mitogen-activated protein kinases, c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinase (ERK). DCLF also causes an increase in intracellular calcium (Ca(++)) in hepatocytes, but the role of this in the cytotoxic synergy between DCLF and cytokines is unknown. We tested the hypothesis that Ca(++) contributes to DCLF/cytokine-induced cytotoxic synergy. Treatment of HepG2 cells with DCLF led to an increase in intracellular Ca(++) at 6 and 12 h, and this response was augmented in the presence of TNF and IFN at 12 h. The intracellular Ca(++) chelator BAPTA/AM reduced cytotoxicity and caspase-3 activation caused by DCLF/cytokine cotreatment. BAPTA/AM also significantly reduced DCLF-induced activation of the ER stress sensor, protein kinase RNA-like ER kinase (PERK), as well as activation of JNK and ERK. Treatment of cells with an inositol trisphosphate receptor antagonist almost completely eliminated DCLF/cytokine-induced cytotoxicity and decreased DCLF-induced activation of PERK, JNK, and ERK. These findings indicate that Ca(++) contributes to DCLF/cytokine-induced cytotoxic synergy by promoting activation of the ER stress-response pathway and JNK and ERK.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Calcium/physiology , Chemical and Drug Induced Liver Injury/etiology , Cytokines/pharmacology , Diclofenac/toxicity , Endoplasmic Reticulum Stress , Extracellular Signal-Regulated MAP Kinases/metabolism , Hep G2 Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Inositol 1,4,5-Trisphosphate Receptors/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , STAT1 Transcription Factor/physiology
9.
Blood ; 126(15): 1835-43, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26179083

ABSTRACT

Acetaminophen (APAP)-induced liver injury in humans is associated with robust coagulation cascade activation and thrombocytopenia. However, it is not known whether coagulation-driven platelet activation participates in APAP hepatotoxicity. Here, we found that APAP overdose in mice caused liver damage accompanied by significant thrombocytopenia and accumulation of platelets in the liver. These changes were attenuated by administration of the direct thrombin inhibitor lepirudin. Platelet depletion with an anti-CD41 antibody also significantly reduced APAP-mediated liver injury and thrombin generation, indicated by the concentration of thrombin-antithrombin (TAT) complexes in plasma. Compared with APAP-treated wild-type mice, biomarkers of hepatocellular and endothelial damage, plasma TAT concentration, and hepatic platelet accumulation were reduced in mice lacking protease-activated receptor (PAR)-4, which mediates thrombin signaling in mouse platelets. However, selective hematopoietic cell PAR-4 deficiency did not affect APAP-induced liver injury or plasma TAT levels. These results suggest that interconnections between coagulation and hepatic platelet accumulation promote APAP-induced liver injury, independent of platelet PAR-4 signaling. Moreover, the results highlight a potential contribution of nonhematopoietic cell PAR-4 signaling to APAP hepatotoxicity.


Subject(s)
Acetaminophen/toxicity , Antithrombin III/metabolism , Blood Platelets/pathology , Chemical and Drug Induced Liver Injury/etiology , Hematopoietic Stem Cells/drug effects , Hepatocytes/drug effects , Peptide Hydrolases/metabolism , Receptors, Proteinase-Activated/physiology , Analgesics, Non-Narcotic/toxicity , Animals , Blood Coagulation/drug effects , Blood Platelets/drug effects , Blood Platelets/metabolism , Blotting, Western , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
J Pharmacol Exp Ther ; 354(2): 230-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26065700

ABSTRACT

Mouse hepatic parenchymal cells (HPCs) have become the most frequently used in vitro model to study mechanisms of acetaminophen (APAP)-induced hepatotoxicity. It is universally accepted that APAP hepatocellular injury requires bioactivation by cytochromes P450 (P450s), but this remains unproven in primary mouse HPCs in vitro, especially over the wide range of concentrations that have been employed in published reports. The aim of this work was to test the hypothesis that APAP-induced hepatocellular death in vitro depends solely on P450s. We evaluated APAP cytotoxicity and APAP-protein adducts (a biomarker of metabolic bioactivation by P450) using primary mouse HPCs in the presence and absence of a broad-spectrum inhibitor of P450s, 1-aminobenzotriazole (1-ABT). 1-ABT abolished formation of APAP-protein adducts at all concentrations of APAP (0-14 mM), but eliminated cytotoxicity only at small concentrations (≦5 mM), indicating the presence of a P450-independent mechanism at larger APAP concentrations. P450-independent cell death was delayed in onset relative to toxicity observed at smaller concentrations. p-Aminophenol was detected in primary mouse HPCs exposed to large concentrations of APAP, and a deacetylase inhibitor [bis (4-nitrophenyl) phosphate (BNPP)] significantly reduced cytotoxicity. In conclusion, APAP hepatocellular injury in vitro occurs by at least two mechanisms, a P450-dependent mechanism that operates at concentrations of APAP ≦ 5 mM and a P450-independent mechanism that predominates at larger concentrations and is slower in onset. p-Aminophenol most likely contributes to the latter mechanism. These findings should be considered in interpreting results from APAP cytotoxicity studies in vitro and in selecting APAP concentrations for use in such studies.


Subject(s)
Acetaminophen/metabolism , Acetaminophen/toxicity , Cytochrome P-450 Enzyme System , Hepatocytes/drug effects , Hepatocytes/metabolism , Analgesics, Non-Narcotic/metabolism , Analgesics, Non-Narcotic/toxicity , Animals , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL
11.
Toxicol Sci ; 146(2): 265-80, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25953702

ABSTRACT

Nonsteroidal anti-inflammatory drugs (NSAIDs) are among the most frequent causes of idiosyncratic, drug-induced liver injury (IDILI). Mechanisms of IDILI are unknown, but immune responses are suspected to underlie them. In animal models of IDILI, the cytokines tumor necrosis factor-alpha (TNFα) and interferon-gamma (IFNγ) are essential to the pathogenesis. Some drugs associated with IDILI interact with cytokines to kill hepatocytes in vitro, and mitogen-activated protein kinases (MAPKs) might play a role. We tested the hypothesis that caspases and MAPKs are involved in NSAID/cytokine-induced cytotoxicity. NSAIDs that are acetic acid (AA) derivatives and associated with IDILI synergized with TNFα in causing cytotoxicity in HepG2 cells, and IFNγ enhanced this interaction. NSAIDs that are propionic acid (PA) derivatives and cause IDILI that is of less clinical concern also synergized with TNFα, but IFNγ was without effect. Caspase inhibition prevented cytotoxicity from AA and PA derivative/cytokine treatment. Treatment with a representative AA or PA derivative induced activation of the MAPKs c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK), and p38. Inhibition of either JNK or ERK reduced cytotoxicity from cytokine interactions with AA derivatives. In contrast, an ERK inhibitor potentiated cytotoxicity from cytokine interactions with PA derivatives. An AA derivative but not a PA derivative enhanced IFNγ-mediated activation of STAT-1, and this enhancement was ERK-dependent. These findings raise the possibility that some IDILI reactions result from drug/cytokine synergy involving caspases and MAPKs and suggest that, even for drugs within the same pharmacologic class, synergy with cytokines occurs by different kinase signaling mechanisms.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Cytokines/toxicity , Liver/drug effects , Mitogen-Activated Protein Kinases/metabolism , Animals , Caspase 3/metabolism , Drug Synergism , Enzyme Activation , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Phosphorylation , STAT1 Transcription Factor/metabolism
12.
Toxicology ; 331: 35-46, 2015 May 04.
Article in English | MEDLINE | ID: mdl-25748550

ABSTRACT

Use of the fluoroquinolone antibiotic trovafloxacin (TVX) was restricted due to idiosyncratic, drug-induced liver injury (IDILI). Previous studies demonstrated that tumor necrosis factor-alpha (TNF) and TVX interact to cause death of hepatocytes in vitro that was associated with prolonged activation of c-Jun N-terminal kinase (JNK), activation of caspases 9 and 3, and DNA damage. The purpose of this study was to explore further the mechanism by which TVX interacts with TNF to cause cytotoxicity. Treatment with TVX caused cell cycle arrest, enhanced expression of p21 and impaired proliferation, but cell death only occurred after cotreatment with TVX and TNF. Cell death involved activation of extracellular signal-related kinase (ERK), which in turn activated caspase 3 and ataxia telangiectasia and Rad3-related (ATR), both of which contributed to cytotoxicity. Cotreatment of HepG2 cells with TVX and TNF caused double-strand breaks in DNA, and ERK contributed to this effect. Inhibition of caspase activity abolished the DNA strand breaks. The data suggest a complex interaction of TVX and TNF in which TVX causes replication stress, and the downstream effects are exacerbated by TNF, leading to hepatocellular death. These results raise the possibility that IDILI from TVX results from MAPK and ATR activation in hepatocytes initiated by interaction of cytokine signaling with drug-induced replication stress.


Subject(s)
Anti-Bacterial Agents/toxicity , Cell Proliferation/drug effects , Chemical and Drug Induced Liver Injury/etiology , DNA Replication/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluoroquinolones/toxicity , Hepatocytes/drug effects , Liver/drug effects , Naphthyridines/toxicity , Tumor Necrosis Factor-alpha/toxicity , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/metabolism , Caspase 3/metabolism , Cell Cycle Checkpoints/drug effects , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Breaks, Double-Stranded , Enzyme Activation , Hep G2 Cells , Hepatocytes/enzymology , Hepatocytes/pathology , Humans , Liver/enzymology , Liver/pathology , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Time Factors , Tumor Suppressor Protein p53/metabolism
13.
J Pharmacol Exp Ther ; 350(1): 164-70, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24817034

ABSTRACT

Trovafloxacin (TVX) is a drug that has caused idiosyncratic, drug-induced liver injury (IDILI) in humans. In a murine model of IDILI, otherwise nontoxic doses of TVX and the inflammagen lipopolysaccharide (LPS) interacted to produce pronounced hepatocellular injury. The liver injury depended on a TVX-induced, small but significant prolongation of tumor necrosis factor-α (TNF) appearance in the plasma. The enhancement of TNF expression by TVX was reproduced in vitro in RAW 264.7 murine macrophages (RAW cells) stimulated with LPS. The current study was designed to identify the molecular target of TVX responsible for this response in RAW cells. An in silico analysis suggested a favorable binding profile of TVX to eukaryotic topoisomerase II-α (TopIIα), and a cell-free assay revealed that TVX inhibited eukaryotic TopIIα activity. Topoisomerase inhibition is known to lead to DNA damage, and TVX increased the DNA damage marker phosphorylated histone 2A.X in RAW cells. Moreover, TVX induced activation of the DNA damage sensor kinases, ataxia telangiectasia mutated (ATM) and Rad3-related (ATR). The ATR inhibitor NU6027 [6-(cyclohexylmethoxy)-5-nitrosopyrimidine-2,4-diamine] prevented the TVX-mediated increases in LPS-induced TNF mRNA and protein release, whereas a selective ATM inhibitor [2-(4-morpholinyl)-6-(1-thianthrenyl)-4H-pyran-4-one (KU55933)] was without effect. TVX prolonged TNF mRNA stability, and this effect was largely attenuated by NU6027. These results suggest that TVX can inhibit eukaryotic topoisomerase, leading to activation of ATR and potentiation of TNF release by macrophages, at least in part through increased mRNA stability. This off-target effect might contribute to the ability of TVX to precipitate IDILI in humans.


Subject(s)
Antigens, Neoplasm/metabolism , DNA Damage/drug effects , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Fluoroquinolones/toxicity , Macrophages/metabolism , Naphthyridines/toxicity , Animals , Cells, Cultured , DNA-Binding Proteins/antagonists & inhibitors , Fluoroquinolones/antagonists & inhibitors , Humans , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Mice , Morpholines/pharmacology , Naphthyridines/antagonists & inhibitors , Nitroso Compounds/pharmacology , Pyrimidines/pharmacology , Pyrones/pharmacology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/biosynthesis
14.
J Pharmacol Exp Ther ; 349(2): 185-91, 2014 May.
Article in English | MEDLINE | ID: mdl-24525298

ABSTRACT

Trovafloxacin (TVX) is a fluoroquinolone antibiotic known to cause idiosyncratic, drug-induced liver injury (IDILI) in humans. The mechanism underlying this toxicity remains unknown. Previously, an animal model of IDILI in mice revealed that TVX synergizes with inflammatory stress from bacterial lipopolysaccharide (LPS) to produce a hepatotoxic interaction. The liver injury required prolongation of the appearance of tumor necrosis factor-α (TNF) in the plasma. The results presented here describe a model of TVX/LPS coexposure in RAW 264.7 cells acting as a surrogate for TNF-releasing cells in vivo. Pretreating cells with TVX for 2 hours before LPS addition led to increased TNF protein release into culture medium in a concentration- and time-dependent manner relative to cells treated with LPS or TVX alone. During the pretreatment period, TVX increased TNF mRNA, but this was less apparent when cells were exposed to TVX after LPS addition, suggesting that the pivotal signaling events that increase TNF expression occurred during the TVX pretreatment period. Indeed, TVX exposure increased activation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase. Inhibition of either ERK or JNK decreased the TVX-mediated increase in TNF mRNA and LPS-induced TNF protein release, but p38 inhibition did not. These results demonstrated that the increased TNF appearance from TVX-LPS interaction in vivo can be reproduced in vitro and occurs in an ERK- and JNK-dependent manner.


Subject(s)
Anti-Bacterial Agents/adverse effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluoroquinolones/adverse effects , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , Naphthyridines/adverse effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line , Dose-Response Relationship, Drug , Drug Synergism , Macrophages/drug effects , Macrophages/metabolism , Mice , Phosphorylation , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/genetics
15.
Toxicol Sci ; 137(1): 91-101, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24097668

ABSTRACT

Idiosyncratic drug-induced liver injury (IDILI) continues to be a significant human health problem. IDILI is characterized as occurring in a minority of individuals exposed to a drug, yet it accounts for as much as 17% of all cases of acute liver failure. Despite these concerns, the mechanisms underlying IDILI remain unknown. Trovafloxacin (TVX), which causes IDILI in humans, also causes hepatocellular death in vitro when combined with tumor necrosis factor-alpha (TNF) treatment. However, the molecular mechanisms involved in this toxicity are not fully characterized. The purpose of this study was to identify mechanisms by which TVX and TNF interact to cause hepatocellular death, with a focus on a human hepatocyte cell line. TVX and TNF interacted to cause cytotoxicity in HepG2 cells at drug concentrations similar to those in people undergoing TVX therapy. TVX/TNF treatment caused apoptosis and DNA damage in HepG2 cells that depended on caspase activation. Prolonged activation of JNK occurred in TVX/TNF-induced cytotoxicity, and treatment with the JNK selective inhibitor SP600125 attenuated cytotoxicity. TVX/TNF cotreatment also caused cytotoxicity in isolated primary murine hepatocytes that was dependent on caspase activation. These results increase understanding of molecular signaling pathways involved in hepatocellular death caused by a drug with idiosyncratic liability in the presence of TNF.


Subject(s)
Anti-Bacterial Agents/toxicity , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/etiology , Fluoroquinolones/toxicity , Hepatocytes/drug effects , Naphthyridines/toxicity , Tumor Necrosis Factor-alpha/metabolism , Animals , Caspases/metabolism , Cell Survival/drug effects , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Dose-Response Relationship, Drug , Enzyme Activation , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Kinase Inhibitors , Signal Transduction/drug effects , Time Factors
16.
Toxicol Sci ; 136(1): 72-85, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23970800

ABSTRACT

For many liver diseases, including viral and autoimmune hepatitis, immune cells play an important role in the development and progression of liver injury. Concanavalin A (Con A) administration to rodents has been used as a model of immune-mediated liver injury resembling human autoimmune hepatitis. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) has been demonstrated to alter the development of immune-mediated diseases. Mice pretreated with TCDD developed exacerbated liver injury in response to administration of a mild dose (6 mg/kg) of Con A. In the present study, we tested the hypothesis that TCDD pretreatment exacerbates Con A-induced liver injury by enhancing the activation and recruitment of accessory cell types including neutrophils, macrophages, and natural killer (NK) cells. Mice were treated with 0, 0.3, 3, or 30 µg/kg TCDD and 4 days later with Con A or saline. TCDD pretreatment with doses of 3 and 30 µg/kg significantly increased liver injury from Con A administration. The plasma concentrations of neutrophil chemokines were significantly increased in TCDD-pretreated mice after Con A administration. NKT cell-deficient (CD1d KO) mice were used to examine whether NKT cells were required for TCDD/Con A-induced liver injury. CD1d KO mice were completely protected from liver injury induced by treatment with Con A alone, whereas the injury from TCDD/Con A treatment was reduced but not eliminated. However, T-cell deficient (RAG1 KO) mice were protected from liver injury induced by Con A irrespective of pretreatment with TCDD. TCDD/Con A treatment increased the percentage of NK cells expressing the activation marker CD69. Depletion of NK cells prior to treatment resulted in significant reductions in plasma interferon-γ and liver injury from TCDD/Con A treatment. In summary, exposure to TCDD exacerbated the immune-mediated liver injury induced by Con A, and our findings suggest that NK cells play a critical role in this response.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Concanavalin A/toxicity , Liver/drug effects , Natural Killer T-Cells/drug effects , Polychlorinated Dibenzodioxins/toxicity , Animals , Antigens, CD/metabolism , Antigens, CD1d/genetics , Antigens, CD1d/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Chemokines/blood , Dose-Response Relationship, Drug , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Inflammation Mediators/blood , Lectins, C-Type/metabolism , Liver/immunology , Liver/metabolism , Liver/pathology , Lymphocyte Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Neutrophil Infiltration/drug effects , Time Factors
17.
Toxicol Sci ; 136(1): 51-62, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23912913

ABSTRACT

It has been demonstrated that co-treatment of rats with amiodarone (AMD) and bacterial lipopolysaccharide (LPS) produces idiosyncrasy-like liver injury. In this study, the hypothesis that the hemostatic system and neutrophils contribute to AMD/LPS-induced liver injury was explored. Rats were treated with AMD (400 mg/kg, ip) or vehicle and 16 h later with LPS (1.6×106 endotoxin units/kg, iv) or saline (Sal). AMD did not affect the hemostatic system by itself but significantly potentiated LPS-induced coagulation activation and fibrinolysis impairment. Increased hepatic fibrin deposition and subsequent hypoxia were observed only in AMD/LPS-treated animals, starting before the onset of liver injury. Administration of anticoagulant heparin abolished AMD/LPS-induced hepatic fibrin deposition and reduced AMD/LPS-induced liver damage. Polymorphonuclear neutrophils (PMNs) accumulated in liver after treatment with LPS or AMD/LPS, but PMN activation was only observed in AMD/LPS-treated rats. Rabbit anti-rat PMN serum, which reduced accumulation of PMNs in liver, prevented PMN activation and attenuated AMD/LPS-induced liver injury in rats. PMN depletion did not affect hepatic fibrin deposition. Anticoagulation prevented PMN activation without affecting PMN accumulation. In summary, both the hemostatic system alteration and PMN activation contributed to AMD/LPS-induced liver injury in rats, in which fibrin deposition was critical for the activation of PMNs.


Subject(s)
Amiodarone/toxicity , Chemical and Drug Induced Liver Injury/etiology , Hemostasis/drug effects , Lipopolysaccharides/toxicity , Liver/drug effects , Neutrophil Activation/drug effects , Neutrophils/drug effects , Animals , Anticoagulants/pharmacology , Blood Coagulation/drug effects , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/pathology , Fibrin/metabolism , Fibrinolysis/drug effects , Heparin/pharmacology , Hypoxia/blood , Hypoxia/chemically induced , Liver/metabolism , Liver/pathology , Male , Neutrophils/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
18.
Toxicol Sci ; 131(1): 164-78, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23042730

ABSTRACT

Amiodarone (AMD), a class III antiarrhythmic drug, causes idiosyncratic hepatotoxicity in human patients. We demonstrated previously that tumor necrosis factor-alpha (TNF-α) plays an important role in a rat model of AMD-induced hepatotoxicity under inflammatory stress. In this study, we developed a model in vitro to study the roles of caspase activation and oxidative stress in TNF potentiation of AMD cytotoxicity. AMD caused cell death in Hepa1c1c7 cells, and TNF cotreatment potentiated its toxicity. Activation of caspases 9 and 3/7 was observed in AMD/TNF-cotreated cells, and caspase inhibitors provided minor protection from cytotoxicity. Intracellular reactive oxygen species (ROS) generation and lipid peroxidation were observed after treatment with AMD and were further elevated by TNF cotreatment. Adding water-soluble antioxidants (trolox, N-acetylcysteine, glutathione, or ascorbate) produced only minor attenuation of AMD/TNF-induced cytotoxicity and did not influence the effect of AMD alone. On the other hand, α-tocopherol (TOCO), which reduced lipid peroxidation and ROS generation, prevented AMD toxicity and caused pronounced reduction in cytotoxicity from AMD/TNF cotreatment. α-TOCO plus a pancaspase inhibitor completely abolished AMD/TNF-induced cytotoxicity. In summary, activation of caspases and oxidative stress were observed after AMD/TNF cotreatment, and caspase inhibitors and a lipid-soluble free-radical scavenger attenuated AMD/TNF-induced cytotoxicity.


Subject(s)
Amiodarone/toxicity , Apoptosis/drug effects , Caspases/metabolism , Oxidative Stress/drug effects , Tumor Necrosis Factor-alpha/toxicity , Animals , Antioxidants/pharmacology , Caspase Inhibitors/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , DNA Breaks/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Activation , In Situ Nick-End Labeling , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred C57BL
19.
Toxicol Appl Pharmacol ; 266(2): 317-27, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23164664

ABSTRACT

Inflammation plays a major role in immune-mediated liver injury, and exposure to environmental pollutants such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) has been reported to alter the inflammatory response as well as affect immune cell activity. In this study, we tested the hypothesis that TCDD pretreatment exacerbates hepatotoxicity in a murine model of immune-mediated liver injury induced by concanavalin A (Con A) administration. Mice were pretreated with 30 µg/kg TCDD or vehicle control on day zero and then given either Con A or saline intravenously on day four. Mice treated with TCDD did not develop liver injury; however, TCDD pretreatment increased liver injury resulting from moderate doses of Con A (4-10 mg/kg). TCDD-pretreated mice had altered plasma concentrations of inflammatory cytokines, including interferon gamma (IFNγ), and TCDD/Con A-induced hepatotoxicity was attenuated in IFNγ knockout mice. At various times after treatment, intrahepatic immune cells were isolated, and expression of cell activation markers as well as cytolytic proteins was determined. TCDD pretreatment increased the proportion of activated natural killer T (NKT) cells and the percent of cells expressing Fas ligand (FasL) after Con A administration. In addition FasL knockout mice and mice treated with CD18 antiserum were both protected from TCDD/Con A-induced hepatotoxicity, suggesting a requirement for direct cell-cell interaction between effector immune cells and parenchymal cell targets in the development of liver injury from TCDD/Con A treatment. In summary, exposure to TCDD increased NKT cell activation and exacerbated immune-mediated liver injury induced by Con A through a mechanism involving IFNγ and FasL expression.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Concanavalin A/toxicity , Environmental Pollutants/toxicity , Inflammation/chemically induced , Polychlorinated Dibenzodioxins/toxicity , Animals , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/pathology , Concanavalin A/administration & dosage , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Environmental Pollutants/administration & dosage , Fas Ligand Protein/genetics , Gene Expression Regulation/drug effects , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/metabolism , Polychlorinated Dibenzodioxins/administration & dosage , Time Factors
20.
Chem Res Toxicol ; 25(11): 2412-22, 2012 Nov 19.
Article in English | MEDLINE | ID: mdl-23013248

ABSTRACT

An NMR-based metabonomic approach was applied to study the systems level metabolic effects of two closely related thiophene compounds, tienilic acid (TA) and tienilic acid isomer (TAI). The metabonomic data were anchored with traditional clinical chemistry and histopathologic analyses. TA was removed from the market as a result of suspected immune-mediated hepatotoxicity, whereas TAI is an intrinsic hepatotoxin. Equimolar doses of TA and TAI were administered to Sprague-Dawley rats, and sampling was conducted at 2, 6, and 24 h post-treatment. Histopathologic analyses revealed development of a significant hepatic lesion 24 h post-TAI treatment with a parallel increase in plasma alanine aminotransferase (ALT) activity. In contrast, TA was not associated with the development of a hepatic lesion or an increase in plasma ALT activity. High-resolution NMR spectral metabolic profiles were generated for liver extracts, plasma, and urine at multiple time points. Multivariate statistical tools were applied to model the metabolic profiles and identify discriminatory metabolites that reflected both the adaptation to TA administration and the onset and progression of TAI-induced hepatotoxicity. TAI was shown to induce marked metabolic effects on the metabolome at all time points, with dramatic metabolic perturbations at 24 h post-treatment correlating with the histopathologic and clinical chemistry evidence of a hepatic lesion. The TAI-induced metabolic perturbations provided evidence for the generation of electrophilic reactive metabolites and a significant impairment of bioenergetic metabolic pathways. TA induced early metabolic perturbations that were largely resolved by 24 h post-treatment, suggesting the reestablishment of metabolic homeostasis and the ability to adapt to the intervention, with hepatic hypotaurine potentially representing a means of assessment of hepatic adaptation. This comparative metabonomic approach enabled the discrimination of metabolic perturbations that were common to both treatments and were interpreted as nontoxic thiophene-induced perturbations. Importantly, this approach enabled the identification of temporal metabolic perturbations that were unique to TAI or TA treatment and hence were of relevance to the development of toxicity or the ability to adapt. This approach is applicable to the future study of pharmacologically and structurally similar compounds and represents a refined means of identification of biomarkers of toxicity.


Subject(s)
Ticrynafen/metabolism , Animals , Chemistry, Clinical , Endpoint Determination , Liver/chemistry , Liver/metabolism , Liver/pathology , Magnetic Resonance Spectroscopy , Male , Molecular Structure , Phenotype , Rats , Rats, Sprague-Dawley , Stereoisomerism , Ticrynafen/administration & dosage , Ticrynafen/chemistry , Toxicity Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...