Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Food Chem Toxicol ; 81: 62-70, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25865938

ABSTRACT

Overdose of acetaminophen (APAP) is a common cause of acute liver injury and liver failure. The mechanism involves formation of a reactive metabolite, protein binding, oxidative stress and activation of c-Jun N-terminal kinase (JNK), mitochondrial dysfunction, and nuclear DNA fragmentation caused by endonucleases released from damaged mitochondria. Previous work has shown that the natural product resveratrol (RSV) can protect against APAP hepatotoxicity in mice through prevention of lipid peroxidation and anti-inflammatory effects. However, these earlier studies did not take into consideration several fundamental aspects of the pathophysiology. To address this, we treated C57Bl/6 mice with 300 mg/kg APAP followed by 50 mg/kg RSV 1.5 h later. Our results confirmed that RSV reduced liver injury after APAP overdose in mice. Importantly, RSV did not inhibit reactive metabolite formation and protein bindings, nor did it reduce activation of JNK. However, RSV decreased protein nitration after APAP treatment, possibly through direct scavenging of peroxynitrite. Interestingly, RSV also inhibited release of apoptosis-inducing factor and endonuclease G from mitochondria independent of Bax pore formation and prevented the downstream nuclear DNA fragmentation. Our data show that RSV protects against APAP hepatotoxicity both through antioxidant effects and by preventing mitochondrial release of endonucleases and nuclear DNA damage.


Subject(s)
Acetaminophen/poisoning , Chemical and Drug Induced Liver Injury/drug therapy , Endonucleases/metabolism , Liver/drug effects , Mitochondria/drug effects , Stilbenes/pharmacology , Animals , Apoptosis Inducing Factor/antagonists & inhibitors , Apoptosis Inducing Factor/metabolism , DNA Fragmentation/drug effects , Drug Overdose/drug therapy , Endonucleases/antagonists & inhibitors , Hepatocytes/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mitochondria/enzymology , Oxidative Stress/drug effects , Peroxynitrous Acid/metabolism , Protein Binding , Resveratrol , Tyrosine/analogs & derivatives , Tyrosine/metabolism
2.
Xenobiotica ; 45(5): 442-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25423287

ABSTRACT

1. The mechanisms of furosemide (FS) hepatotoxicity were explored in mice. Specifically, C57Bl/6 J mice were treated with 500 mg FS/kg bodyweight, and c-Jun N-terminal kinase (JNK) activation and receptor-interacting protein kinase 3 (RIP3) expression were measured by western blotting. Co-treatment with FS and the JNK inhibitor SP600125 was also performed, and FS-induced liver injury was compared in wild-type and RIP3 knockout (KO) mice. 2. JNK phosphorylation and RIP3 expression were increased in livers from the FS-treated mice as early as 6 h after treatment and persisted until at least 24 h. JNK phosphorylation was also observed in primary mouse hepatocytes and human HepaRG cells treated with FS. 3. Phosphorylated JNK translocated into mitochondria in livers, but no evidence of mitochondrial damage was observed. 4. SP600125-treated mice, SP600125 co-treated primary mouse hepatocytes and RIP3 KO mice were not protected against FS hepatotoxicity. These data show that, although JNK activation and RIP3 expression are induced by FS, neither contributes to the liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury/enzymology , Furosemide/adverse effects , JNK Mitogen-Activated Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Anthracenes/pharmacology , Anthracenes/therapeutic use , Cell Line , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/pathology , DNA Damage , Enzyme Activation/drug effects , Glutathione Disulfide/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Time Factors
3.
Toxicol Appl Pharmacol ; 275(2): 122-33, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24440789

ABSTRACT

Following acetaminophen (APAP) overdose there is an inflammatory response triggered by the release of cellular contents from necrotic hepatocytes into the systemic circulation which initiates the recruitment of neutrophils into the liver. It has been demonstrated that neutrophils do not contribute to APAP-induced liver injury, but their role and the role of NADPH oxidase in injury resolution are controversial. C57BL/6 mice were subjected to APAP overdose and neutrophil activation status was determined during liver injury and liver regeneration. Additionally, human APAP overdose patients (ALT: >800 U/L) had serial blood draws during the injury and recovery phases for the determination of neutrophil activation. Neutrophils in the peripheral blood of mice showed an increasing activation status (CD11b expression and ROS priming) during and after the peak of injury but returned to baseline levels prior to complete injury resolution. Hepatic sequestered neutrophils showed an increased and sustained CD11b expression, but no ROS priming was observed. Confirming that NADPH oxidase is not critical to injury resolution, gp91(phox)⁻/⁻ mice following APAP overdose displayed no alteration in injury resolution. Peripheral blood from APAP overdose patients also showed increased neutrophil activation status after the peak of liver injury and remained elevated until discharge from the hospital. In mice and humans, markers of activation, like ROS priming, were increased and sustained well after active liver injury had subsided. The similar findings between surviving patients and mice indicate that neutrophil activation may be a critical event for host defense or injury resolution following APAP overdose, but not a contributing factor to APAP-induced injury.


Subject(s)
Acetaminophen/adverse effects , Liver Regeneration/drug effects , Liver/drug effects , Neutrophil Activation/drug effects , Adult , Aged , Animals , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Drug Overdose , Female , Granulocytes/drug effects , Granulocytes/metabolism , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , NADPH Oxidases/metabolism , Reactive Oxygen Species , Young Adult
4.
Toxicol Appl Pharmacol ; 274(3): 417-24, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24345528

ABSTRACT

Acetaminophen (APAP) overdose causes severe and occasionally fatal liver injury. Numerous drugs that attenuate APAP toxicity have been described. However these compounds frequently protect by cytochrome P450 inhibition, thereby preventing the initiating step of toxicity. We have previously shown that pretreatment with allopurinol can effectively protect against APAP toxicity, but the mechanism remains unclear. In the current study, C3HeB/FeJ mice were administered allopurinol 18h or 1h prior to an APAP overdose. Administration of allopurinol 18h prior to APAP overdose resulted in an 88% reduction in liver injury (serum ALT) 6h after APAP; however, 1h pretreatment offered no protection. APAP-cysteine adducts and glutathione depletion kinetics were similar with or without allopurinol pretreatment. The phosphorylation and mitochondrial translocation of c-jun-N-terminal-kinase (JNK) have been implicated in the progression of APAP toxicity. In our study we showed equivalent early JNK activation (2h) however late JNK activation (6h) was attenuated in allopurinol treated mice, which suggests that later JNK activation is more critical for the toxicity. Additional mice were administered oxypurinol (primary metabolite of allopurinol) 18h or 1h pre-APAP, but neither treatment protected. This finding implicated an aldehyde oxidase (AO)-mediated metabolism of allopurinol, so mice were treated with hydralazine to inhibit AO prior to allopurinol/APAP administration, which eliminated the protective effects of allopurinol. We evaluated potential targets of AO-mediated preconditioning and found increased hepatic metallothionein 18h post-allopurinol. These data show metabolism of allopurinol occurring independent of P450 isoenzymes preconditions the liver and renders the animal less susceptible to an APAP overdose.


Subject(s)
Acetaminophen/analogs & derivatives , Aldehyde Oxidase/metabolism , Allopurinol/pharmacology , Chemical and Drug Induced Liver Injury/drug therapy , Cysteine/analogs & derivatives , Liver/drug effects , Acetaminophen/administration & dosage , Acetaminophen/adverse effects , Aldehyde Oxidase/antagonists & inhibitors , Animals , Chemical and Drug Induced Liver Injury/pathology , Cysteine/administration & dosage , Cysteine/adverse effects , Drug Overdose , Glutathione/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/metabolism , Male , Metallothionein/metabolism , Mice , Mice, Inbred C3H , Mitochondria/drug effects , Mitochondria/metabolism , Oxypurinol/metabolism , Phosphorylation , Xanthine Oxidase/metabolism
5.
Toxicol Appl Pharmacol ; 273(3): 524-31, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24096036

ABSTRACT

Cholestasis is a pathological common component of numerous liver diseases that results in hepatotoxicity, inflammation, and cirrhosis when untreated. While the predominant hypothesis in cholestatic liver injury remains hepatocyte apoptosis due to direct toxicity of hydrophobic bile acid exposure, recent work suggests that the injury occurs through inflammatory necrosis. In order to resolve this controversy, we used novel plasma biomarkers to assess the mechanisms of cell death during early cholestatic liver injury. C57Bl/6 mice underwent bile duct ligation (BDL) for 6-72 h, or sham operation. Another group of mice were given d-galactosamine and endotoxin as a positive control for apoptosis and inflammatory necrosis. Plasma levels of full length cytokeratin-18 (FL-K18), microRNA-122 (miR-122) and high mobility group box-1 protein (HMGB1) increased progressively after BDL with peak levels observed after 48 h. These results indicate extensive cell necrosis after BDL, which is supported by the time course of plasma alanine aminotransferase activities and histology. In contrast, plasma caspase-3 activity, cleaved caspase-3 protein and caspase-cleaved cytokeratin-18 fragments (cK18) were not elevated at any time during BDL suggesting the absence of apoptosis. In contrast, all plasma biomarkers of necrosis and apoptosis were elevated 6 h after Gal/End treatment. In addition, acetylated HMGB1, a marker for macrophage and monocyte activation, was increased as early as 12 h but mainly at 48-72 h. However, progressive neutrophil accumulation in the area of necrosis started at 6h after BDL. In conclusion, these data indicate that early cholestatic liver injury in mice is an inflammatory event, and occurs through necrosis with little evidence for apoptosis.


Subject(s)
Apoptosis , Biomarkers/blood , Cholestasis/blood , Liver Diseases/blood , Alanine Transaminase/blood , Animals , Bile Acids and Salts/adverse effects , Bile Acids and Salts/metabolism , Bile Ducts/surgery , Caspase 3/blood , Cholestasis/pathology , Galactosamine/adverse effects , HMGB1 Protein/blood , Hepatocytes/pathology , Inflammation/pathology , Keratin-18/blood , Ligation , Liver/metabolism , Liver/pathology , Liver Diseases/pathology , Mice , Mice, Inbred C57BL , MicroRNAs/blood , Necrosis/blood , Necrosis/pathology , Neutrophils/pathology
6.
Toxicol Appl Pharmacol ; 269(3): 240-9, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23571099

ABSTRACT

At therapeutic doses, acetaminophen (APAP) is a safe and effective analgesic. However, overdose of APAP is the principal cause of acute liver failure in the West. Binding of the reactive metabolite of APAP (NAPQI) to proteins is thought to be the initiating event in the mechanism of hepatotoxicity. Early work suggested that APAP-protein binding could not occur without glutathione (GSH) depletion, and likely only at toxic doses. Moreover, it was found that protein-derived APAP-cysteine could only be detected in serum after the onset of liver injury. On this basis, it was recently proposed that serum APAP-cysteine could be used as diagnostic marker of APAP overdose. However, comprehensive dose-response and time course studies have not yet been done. Furthermore, the effects of co-morbidities on this parameter have not been investigated. We treated groups of mice with APAP at multiple doses and measured liver GSH and both liver and plasma APAP-protein adducts at various timepoints. Our results show that protein binding can occur without much loss of GSH. Importantly, the data confirm earlier work that showed that protein-derived APAP-cysteine can appear in plasma without liver injury. Experiments performed in vitro suggest that this may involve multiple mechanisms, including secretion of adducted proteins and diffusion of NAPQI directly into plasma. Induction of liver necrosis through ischemia-reperfusion significantly increased the plasma concentration of protein-derived APAP-cysteine after a subtoxic dose of APAP. While our data generally support the measurement of serum APAP-protein adducts in the clinic, caution is suggested in the interpretation of this parameter.


Subject(s)
Acetaminophen/metabolism , Analgesics, Non-Narcotic/metabolism , Liver/drug effects , Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Animals , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Dose-Response Relationship, Drug , Glutathione/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Reperfusion Injury/metabolism
7.
Basic Clin Pharmacol Toxicol ; 111(6): 417-25, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22900545

ABSTRACT

Acetaminophen (APAP) overdose is currently the most frequent cause of drug-induced liver failure in the United States. Recently, it was shown that lysosomal iron translocates to mitochondria where it contributes to the collapse of the mitochondrial membrane potential. Therefore, the purpose of this study was to investigate whether cathepsin B, a lysosomal protease, is involved in APAP-induced hepatotoxicity. Cathepsin B activity was measured in subcellular liver fractions of C57Bl/6 mice 3 hr after 300 mg/kg APAP treatment. There was a significant increase in cytoplasmic cathepsin activity, concurrent with a decrease in microsomal activity, indicative of lysosomal cathepsin B release. To investigate the effect of cathepsin B on hepatotoxicity, the cathepsin inhibitor AC-LVK-CHO was given 1 hr prior to 300 mg/kg APAP treatment along with vehicle control. There was no difference between groups in serum alanine aminotransferase (ALT) values, or by histological evaluation of necrosis, although cathepsin B activity was inhibited by 70-80% compared with controls. These findings were confirmed with a different inhibitor (z-FA-fmk) in vivo and in vitro. Hepatocytes were exposed to 5 mM acetaminophen. Lysotracker staining confirmed lysosomal instability and cathepsin B release, but there was no reduction in cell death after treatment with cathepsin B inhibitors. Finally, cathepsin B release was measured in clinical samples from patients with APAP-induced liver injury. Low levels of cathepsin B were released into plasma from overdose patients. APAP overdose causes lysosomal instability and release of cathepsin B into the cytosol but does not contribute to liver injury under these conditions.


Subject(s)
Acetaminophen/toxicity , Cathepsin B/metabolism , Chemical and Drug Induced Liver Injury/physiopathology , Hepatocytes/drug effects , Lysosomes/pathology , Adult , Animals , Cathepsin B/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Dose-Response Relationship, Drug , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Ketones/pharmacology , Liver/drug effects , Liver/metabolism , Liver/pathology , Lysosomes/drug effects , Male , Mice , Mice, Inbred C57BL , Middle Aged , Prospective Studies
8.
Toxicol Sci ; 122(2): 598-605, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21572097

ABSTRACT

Acetaminophen (APAP) overdose causes liver injury in humans and mice. DNA fragmentation is a hallmark of APAP-induced cell death, and nuclear translocation of apoptosis-inducing factor (AIF) correlates with DNA fragmentation after APAP overdose. To test the hypothesis that AIF may be a critical mediator of APAP-induced cell death, fasted male AIF-deficient Harlequin (Hq) mice and respective wild-type (WT) animals were treated with 200 mg/kg APAP. At 6 h after APAP, WT animals developed severe liver injury as indicated by the increase in plasma alanine aminotransferase (ALT) activities (8600 ± 1870 U/l) and 61 ± 8% necrosis. This injury was accompanied by massive DNA strand breaks in centrilobular hepatocytes (terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling [TUNEL] assay) and release of DNA fragments into the cytosol (anti-histone ELISA). In addition, there was formation of reactive oxygen (increase in liver glutathione disulfide (GSSG) levels and mitochondrial protein carbonyls) and peroxynitrite (nitrotyrosine [NT] staining) together with mitochondrial translocation of activated c-jun-N-terminal kinase (P-JNK) and release of AIF from the mitochondria. In contrast, Hq mice had significantly less liver injury (ALT: 330 ± 130 U/l; necrosis: 4 ± 2%), minimal nuclear DNA damage, and drastically reduced oxidant stress (based on all parameters) at 6 h. WT and Hq mice had the same baseline levels of cyp2E1 and of glutathione. The initial depletion of glutathione (20 min after APAP) was the same in both groups suggesting that there was no relevant difference in metabolic activation of APAP. Thus, AIF has a critical function in APAP hepatotoxicity by facilitating generation of reactive oxygen in mitochondria and, after nuclear translocation, AIF can be involved in DNA fragmentation.


Subject(s)
Acetaminophen/toxicity , Apoptosis Inducing Factor/metabolism , Chemical and Drug Induced Liver Injury/pathology , Hepatocytes/drug effects , Mitochondria, Liver/drug effects , Oxidative Stress/drug effects , Animals , Blotting, Western , DNA Fragmentation , Drug Overdose , Glutathione Disulfide/metabolism , Hepatocytes/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Liver/drug effects , Liver/pathology , Male , Mice , Mitochondria, Liver/metabolism , Necrosis/chemically induced , Necrosis/metabolism , Peroxynitrous Acid/metabolism , Reactive Oxygen Species/metabolism
9.
Toxicol Sci ; 117(2): 515-23, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20616211

ABSTRACT

Oxidative stress and mitochondrial dysfunction play an important role in acetaminophen (APAP)-induced hepatocyte cell death. However, exact mechanisms involved in the process are controversial, in part, because of the disparity in findings between in vitro and in vivo studies. A major difference in this context is the oxygen tension, with cells in culture being exposed to 21% oxygen, whereas those in the liver experience a gradient from 3 to 9% oxygen. To determine if oxygen tensions could modulate hepatocyte responses to APAP, primary mouse hepatocytes were treated with 5mM APAP for up to 15 h under various oxygen tensions and mitochondrial dysfunction (2,3-bis[2-methoxy-4-nitro-5-sulfophenyl]-2H-tetrazolium-5-carboxyanilide inner salt assay, 5,5',6,6'-tetrachloro-1,1,3,3-tetraethylbenzimidazolylcarbocyanine iodide [JC-1] fluorescence ratio) and cell death (lactate dehydrogenase release) was evaluated. Mitochondrial reactive oxygen and reactive nitrogen species were measured using Mitosox Red or dihydrorhodamine fluorescence and nitrotyrosine staining, respectively. Exposure of hepatocytes to 5mM APAP at 21% O(2) resulted in mitochondrial oxidant stress formation, deterioration of mitochondrial function, and loss of membrane potential as early as 6 h and massive cell death at 15 h. Culture of cells at 10% O(2) resulted in no increase in mitochondrial oxidant stress and better preserved mitochondrial function at 6 h and significant protection against cell death at 15 h. Furthermore, dihydrorhodamine fluorescence was significantly attenuated at 10% oxygen. Cells cultured at 5% oxygen were also protected but showed evidence of hypoxia (accumulation of lactate and nuclear translocation of hypoxia-inducing factor-1α). These results suggest that oxygen tension can modulate hepatocyte responses to APAP, with low physiological levels (10%) decreasing mitochondrial oxidant stress and delaying hepatocyte cell death.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Hepatocytes/drug effects , Mitochondria, Liver/drug effects , Oxidative Stress/drug effects , Oxygen/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , Hepatocytes/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Inbred C57BL , Mitochondria, Liver/metabolism , Oxygen/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism
10.
Liver Int ; 30(9): 1280-92, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20500806

ABSTRACT

BACKGROUND: Acetaminophen (APAP) hepatotoxicity is currently the most frequent cause of acute liver failure in the US and many European countries. Although intracellular signalling mechanisms are critical for hepatocellular injury, a contribution of inflammatory cells, especially neutrophils, has been suggested. However, conflicting results were obtained when using immunological intervention strategies. AIMS: The role of neutrophils was investigated using a CD18-deficient mouse model. RESULTS: Treatment of C57Bl/6 wild type mice with 300 mg/kg APAP resulted in severe liver cell necrosis at 12 and 24 h. This injury was accompanied by formation of cytokines and chemokines and accumulation of neutrophils in the liver. However, there was no difference in the inflammatory response or liver injury in CD18-deficient mice compared with wild-type animals. In contrast to treatment with endotoxin, no upregulation of CD11b or priming for reactive oxygen was observed on neutrophils isolated from the peripheral blood or the liver after APAP administration. Furthermore, animals treated with endotoxin 3 h after APAP experienced an exaggerated inflammatory response as indicated by substantially higher cytokine and chemokine formation and twice the number of neutrophils in the liver. However, liver injury in the two-hit model was the same as with APAP alone. CONCLUSIONS: Our data do not support the hypothesis that neutrophils contribute to APAP hepatotoxicity or that a neutrophil-mediated injury phase could be provoked by a second, pro-inflammatory hit. Thus, APAP-induced liver injury in mice is dominated by intracellular mechanisms of cell death rather than by neutrophilic inflammation.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , CD18 Antigens/genetics , Chemical and Drug Induced Liver Injury/etiology , Liver/drug effects , Neutrophils/metabolism , Animals , Biomarkers , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemokines/genetics , Chemokines/metabolism , Endotoxins/toxicity , Female , Gene Expression/drug effects , Hepatocytes/drug effects , Hepatocytes/pathology , Injections, Intraperitoneal , Liver/metabolism , Liver/pathology , Liver Function Tests , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis/chemically induced , Neutrophils/pathology , Oxidative Stress/drug effects , RNA, Messenger/metabolism
11.
Toxicol Sci ; 104(2): 419-27, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18469330

ABSTRACT

Deficiency in plasminogen activator inhibitor-1 (PAI-1) gene expression is known to promote growth factor activation and regeneration in a number of hepatotoxicity models. To evaluate if PAI-1 has similar effects in acetaminophen (APAP) hepatotoxicity, wild-type (WT) and PAI-1 gene knockout mice (PAI-KO) were treated with 200 mg/kg APAP and liver injury and its repair were assessed. In WT animals, plasma alanine aminotransferase (ALT) activities increased during the first 12 h and then returned to baseline within 48 h. The area of necrosis increased in parallel to the ALT values, peaked between 12 and 24 h and was completely resolved by 96 h. The regenerative response of cells outside the necrotic area, as indicated by proliferating cell nuclear antigen protein and cyclin D(1) gene expression, was observed within 24 h, peaked at 48 h and then declined but remained elevated until 96 h. Liver injury in response to APAP was similar in PAI-KO as in WT animals during the first 12 h. However, plasma ALT values and the area of necrosis further increased during the following 12 h with development of massive intrahepatic hemorrhage. Approximately, 50% of the PAI-KO animals did not survive. Although liver injury of the surviving animals was repaired, the regeneration process was delayed until 48 h. A potential reason for this delay may have been due to the more severe injury and/or the increased expression of the cell cycle inhibitor p21. Our data indicate that PAI activation limits liver injury and mortality during APAP hepatotoxicity by preventing excessive hemorrhage and thereby facilitating tissue repair.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Liver Regeneration/genetics , Serpins/physiology , Alanine Transaminase/blood , Animals , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , DNA Fragmentation/drug effects , Drug Overdose , Gene Expression Regulation/drug effects , Gene Silencing , Glutathione/metabolism , Hemorrhage/chemically induced , Hemorrhage/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Regeneration/drug effects , Longevity/drug effects , Mice , Mice, Inbred Strains , Mice, Knockout , Necrosis/chemically induced , Necrosis/pathology , RNA, Messenger/metabolism , Serpin E2 , Serpins/deficiency
12.
Toxicol Lett ; 177(3): 188-97, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18313239

ABSTRACT

Acetaminophen (APAP) overdose is the leading cause of drug related liver failure in many countries. N-acetyl-p-benzoquinone imine (NAPQI) is a reactive metabolite that is formed by the metabolism of APAP. NAPQI preferentially binds to glutathione and then cellular proteins. NAPQI binding is considered an upstream event in the pathophysiology, especially when binding to mitochondrial proteins and therefore leads to mitochondrial toxicity. APAP caused a significant increase in liver toxicity 3h post-APAP administration as measured by increased serum alanine aminotransferase (ALT) levels. Using high-resolution mitochondrial proteomics techniques to measure thiol and protein changes, no significant change in global thiol levels was observed. However, 3-hydroxy-3-methylglutaryl coenzyme A synthase 2 (HMG-CoA synthase) had significantly decreased levels of reduced thiols and activity after APAP treatment. HMG-CoA synthase is a key regulatory enzyme in ketogenesis and possesses a number of critical cysteines in the active site. Similarly, catalase, a key enzyme in hydrogen peroxide metabolism, also showed modification in protein thiol content. These data indicate post-translational modifications of a few selected proteins involved in mitochondrial and cellular regulation of metabolism during liver toxicity after APAP overdose. The pathophysiological relevance of these limited changes in protein thiols remains to be investigated.


Subject(s)
Acetaminophen/toxicity , Hydroxymethylglutaryl-CoA Synthase/metabolism , Liver/drug effects , Mitochondrial Proteins/analysis , Sulfhydryl Compounds/analysis , Alanine Transaminase/blood , Animals , Liver/metabolism , Male , Mice , Mice, Inbred C3H , Proteomics
13.
J Pharmacol Exp Ther ; 324(1): 8-14, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17906064

ABSTRACT

Mitochondria generate reactive oxygen and peroxynitrite and release endonucleases during acetaminophen (APAP) hepatotoxicity. Because mitochondrial translocation of Bax can initiate these events, we investigated the potential role of Bax in the pathophysiology of hepatic necrosis after 300 mg/kg APAP in fasted C57BL/6 mice. APAP overdose induced Bax translocation from the cytosol to the mitochondria as early as 1 h after APAP injection. At 6 h, there was extensive centrilobular nitrotyrosine staining (indicator for peroxynitrite formation) and nuclear DNA fragmentation. In addition, mitochondrial intermembrane proteins were released into the cytosol. Plasma alanine aminotransferase (ALT) activities of 5610 +/- 600 U/l indicated extensive necrotic cell death. Conversely, Bax gene knockout (Bax(-/-)) mice had 80% lower ALT activities, less DNA fragmentation, and less intermembrane protein release at 6 h. However, immunohistochemical staining for nitrotyrosine or APAP protein adducts did not show differences between wild-type and Bax(-/-) mice. In contrast to the early hepatoprotection in Bax(-/-) mice, plasma ALT activities (7605 +/- 480 U/l) and area of necrosis (53 +/- 6% hepatocytes) in wild-type animals was similar to values in Bax(-/-) mice at 12 h. In addition, there was no difference in DNA fragmentation or nitrotyrosine immunostaining. We concluded that the rapid mitochondrial Bax translocation after APAP overdose has no effect on peroxynitrite formation but that it contributes to the mitochondrial release of proteins, which cause nuclear DNA fragmentation. However, the persistent oxidant stress and peroxynitrite formation in mitochondria may eventually trigger the permeability transition pore opening and release intermembrane proteins independently of Bax.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Chemical and Drug Induced Liver Injury , DNA Fragmentation , Necrosis/chemically induced , bcl-2-Associated X Protein/metabolism , Animals , Disease Models, Animal , Liver Diseases/metabolism , Liver Diseases/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Necrosis/metabolism , Necrosis/pathology , Peroxynitrous Acid/metabolism , bcl-2-Associated X Protein/deficiency , bcl-2-Associated X Protein/genetics
14.
Toxicol Sci ; 94(1): 217-25, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16896059

ABSTRACT

Mitochondrial dysfunction and internucleosomal DNA fragmentation are well-recognized features of acetaminophen (AAP)-induced hepatocyte cell death. However, the endonucleases responsible for this effect have not been identified. Apoptosis-inducing factor (AIF) and endonuclease G are nucleases located in the intermembrane space of mitochondria. AIF is thought to trigger chromatin condensation and induce cleavage of DNA into high molecular weight fragments (50-300 kb), and endonuclease G can produce oligonucleosomal DNA fragments. Therefore, the objective of this investigation was to test the hypothesis that endonuclease G and AIF could be involved in AAP-induced nuclear DNA fragmentation. Using immunofluorescence microscopy, it was shown that in primary cultured mouse hepatocytes, endonuclease G and AIF translocated to the nucleus between 3 and 6 h after exposure to 5 mM AAP. In contrast, other mitochondrial intermembrane proteins such as cytochrome c or the second mitochondria-derived activator of caspases (Smac) did not accumulate in the nucleus. The translocation of AIF and endonuclease G correlated with mitochondrial dysfunction as indicated by the progressive loss of the mitochondrial membrane potential (measured with the JC-1 assay) and the appearance of nuclear DNA fragments in the cytosol (determined by an anti-histone ELISA). Pretreatment with 20mM N-acetylcysteine prevented mitochondrial dysfunction, the nuclear translocation of endonuclease G and AIF, and the nuclear DNA fragmentation. The data support the conclusion that endonuclease G and AIF translocate to the nucleus in response to AAP-induced mitochondrial dysfunction and may be responsible, at least in part, for the initial DNA fragmentation during AAP hepatotoxicity.


Subject(s)
Acetaminophen/toxicity , Apoptosis Inducing Factor/metabolism , Cell Nucleus/drug effects , Endodeoxyribonucleases/metabolism , Hepatocytes/drug effects , Acetylcysteine/pharmacology , Active Transport, Cell Nucleus , Animals , Apoptosis Regulatory Proteins , Benzimidazoles/metabolism , Carbocyanines/metabolism , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Cell Survival/drug effects , Cells, Cultured , Cytochromes c/metabolism , DNA Fragmentation/drug effects , Hepatocytes/cytology , Hepatocytes/metabolism , Immunohistochemistry/methods , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C3H , Microscopy, Fluorescence/methods , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Mitochondrial Proteins/metabolism
15.
Toxicol Appl Pharmacol ; 216(1): 98-107, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16781746

ABSTRACT

Neutrophils are recruited into the liver after acetaminophen (AAP) overdose but the pathophysiological relevance of this acute inflammatory response remains unclear. To address this question, we compared the time course of liver injury, hepatic neutrophil accumulation and inflammatory gene mRNA expression for up to 24 h after treatment with 300 mg/kg AAP in C3Heb/FeJ and C57BL/6 mice. Although there was no relevant difference in liver injury (assessed by the increase of plasma alanine aminotransferase activities and the areas of necrosis), the number of neutrophils and the expression of several pro-inflammatory genes (e.g., tumor necrosis factor-alpha, interleukin-1beta and macrophage inflammatory protein-2) was higher in C3Heb/FeJ than in C57BL/6 mice. In contrast, the expression of the anti-inflammatory genes interleukin-10 and heme oxygenase-1 was higher in C57BL/6 mice. Despite substantial hepatic neutrophil accumulation, none of the liver sections from both strains stained positive for hypochlorite-modified proteins, a specific marker for a neutrophil-induced oxidant stress. In addition, treatment with the NADPH oxidase inhibitors diphenyleneiodonium chloride or apocynin or the anti-neutrophil antibody Gr-1 did not protect against AAP hepatotoxicity. Furthermore, although intercellular adhesion molecule-1 (ICAM-1) was previously shown to be important for neutrophil extravasation and tissue injury in several models, ICAM-1-deficient mice were not protected against AAP-mediated liver injury. Together, these data do not support the hypothesis that neutrophils aggravate liver injury induced by AAP overdose.


Subject(s)
Acetaminophen/toxicity , Chemical and Drug Induced Liver Injury/physiopathology , Inflammation/physiopathology , Acute Disease , Alanine Transaminase/blood , Analgesics, Non-Narcotic/toxicity , Animals , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/etiology , Female , Gene Expression/drug effects , Inflammation/chemically induced , Inflammation/genetics , Intercellular Adhesion Molecule-1/genetics , Interleukin-1beta/genetics , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/drug effects , Poly (ADP-Ribose) Polymerase-1 , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity , Tumor Necrosis Factor-alpha/genetics
16.
Toxicol Sci ; 89(1): 31-41, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16177235

ABSTRACT

Acetaminophen hepatotoxicity is the leading cause of drug-induced liver failure. Despite substantial efforts in the past, the mechanisms of acetaminophen-induced liver cell injury are still incompletely understood. Recent advances suggest that reactive metabolite formation, glutathione depletion, and alkylation of proteins, especially mitochondrial proteins, are critical initiating events for the toxicity. Bcl-2 family members Bax and Bid then form pores in the outer mitochondrial membrane and release intermembrane proteins, e.g., apoptosis-inducing factor (AIF) and endonuclease G, which then translocate to the nucleus and initiate chromatin condensation and DNA fragmentation, respectively. Mitochondrial dysfunction, due to covalent binding, leads to formation of reactive oxygen and peroxynitrite, which trigger the membrane permeability transition and the collapse of the mitochondrial membrane potential. In addition to the diminishing capacity to synthesize ATP, endonuclease G and AIF are further released. Endonuclease G, together with an activated nuclear Ca2+,Mg2+-dependent endonuclease, cause DNA degradation, thereby preventing cell recovery and regeneration. Disruption of the Ca2+ homeostasis also leads to activation of intracellular proteases, e.g., calpains, which can proteolytically cleave structural proteins. Thus, multiple events including massive mitochondrial dysfunction and ATP depletion, extensive DNA fragmentation, and modification of intracellular proteins contribute to the development of oncotic necrotic cell death in the liver after acetaminophen overdose. Based on the recognition of the temporal sequence and interdependency of these mechanisms, it appears most promising to therapeutically target either the initiating event (metabolic activation) or the central propagating event (mitochondrial dysfunction and peroxynitrite formation) to prevent acetaminophen-induced liver cell death.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Hepatocytes/drug effects , Liver/drug effects , Signal Transduction/drug effects , Acetaminophen/metabolism , Analgesics, Non-Narcotic/metabolism , Apoptosis , Cell Death/drug effects , DNA Fragmentation , Hepatocytes/pathology , Liver/pathology , Necrosis , Peroxynitrous Acid/metabolism
17.
Life Sci ; 78(15): 1670-6, 2006 Mar 06.
Article in English | MEDLINE | ID: mdl-16226279

ABSTRACT

The mode of cell death after acetaminophen (AAP) overdose is controversially discussed. A recent study reported a protective effect of the pancaspase inhibitor Z-VAD-fmk against AAP toxicity in vivo but the mechanism of protection remained unclear. Therefore, the objective of this investigation was to assess if Z-VAD-fmk or the low doses of dimethyl sulfoxide (DMSO) used as solvent were responsible for the protection. Treatment with 10 mg/kg Z-VAD-fmk or diluted DMSO (0.25 ml/kg) for 15 min before but not 2.5 h after AAP prevented the oxidant stress (hepatic glutathione disulfide content; nitrotyrosine staining), DNA fragmentation (anti-histone ELISA, TUNEL assay) and liver injury (plasma ALT activities) at 6 h after administration of 300 mg/kg AAP. Even a lower dose (0.1 ml/kg) of DMSO was partially effective. DMSO pretreatment also attenuated the initial decline in hepatic glutathione levels. On the other hand, 10 microM Z-VAD-fmk was unable to prevent AAP-induced cell death in primary cultured mouse hepatocytes. We conclude that Z-VAD-fmk does not protect against AAP-induced liver injury and, therefore, caspases are not involved in the mechanism of AAP-induced liver injury. In contrast, the protection in vivo is caused by the diluted DMSO, which is used to solubilize the inhibitor Z-VAD-fmk. The results emphasize that even very low doses of DMSO, which are generally necessary to dissolve water-insoluble inhibitors, can have a profound impact on the toxicity of drugs and chemicals when metabolic activation is a critical aspect of the mechanism of cell injury.


Subject(s)
Acetaminophen/adverse effects , Analgesics, Non-Narcotic/adverse effects , Caspases/metabolism , Liver Diseases , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Caspase Inhibitors , Chemical and Drug Induced Liver Injury , Cysteine Proteinase Inhibitors/pharmacology , Dimethyl Sulfoxide/pharmacology , Dose-Response Relationship, Drug , Liver Diseases/enzymology , Liver Diseases/prevention & control , Male , Mice , Mice, Inbred Strains , Solvents/pharmacology
18.
J Pharmacol Exp Ther ; 315(2): 879-87, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16081675

ABSTRACT

Intracellular sources of peroxynitrite formation and potential targets for this powerful oxidant and nitrating agent have not been identified after acetaminophen (AAP) overdose. Therefore, we tested the hypothesis that peroxynitrite generated in mitochondria may be responsible for mitochondrial DNA (mtDNA) and nuclear DNA (nDNA) damage. C3Heb/FeJ mice were treated with 300 mg/kg AAP and monitored for up to 12 h. Loss of mtDNA (assayed by slot blot hybridization) and substantial nDNA fragmentation (evaluated by anti-histone enzyme-linked immunosorbent assay, terminal deoxynucleotidyl transferase dUTP nick-end labeling assay, and agarose gel electrophoresis) were observed as early as 3 h after AAP overdose. Analysis of nitrotyrosine protein adducts in subcellular fractions established that peroxynitrite was generated predominantly in mitochondria beginning at 1 h after AAP injection. Delayed treatment with a bolus dose of glutathione (GSH) accelerated the recovery of mitochondrial glutathione, which then effectively scavenged peroxynitrite. However, mtDNA loss was only partially prevented. Despite the absence of nitrotyrosine adducts in the nucleus after AAP overdose, nDNA damage was almost completely eliminated with GSH administration. A direct comparison of nDNA damage after AAP overdose with nDNA fragmentation during tumor necrosis factor receptor-mediated apoptosis showed similar DNA ladders on agarose gels but quantitatively different results in three other assays. We conclude that peroxynitrite may be partially responsible for mtDNA loss but is not directly involved in nDNA damage. In contrast, nDNA fragmentation after AAP overdose is not caused by caspase-activated DNase but most likely by other intracellular DNase(s), whose activation is dependent on the mitochondrial oxidant stress and peroxynitrite formation.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Chemical and Drug Induced Liver Injury/pathology , DNA Damage/drug effects , Endonucleases/metabolism , Mitochondria, Liver/drug effects , Peroxynitrous Acid/toxicity , Animals , Apoptosis/drug effects , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Chemical and Drug Induced Liver Injury/enzymology , DNA Fragmentation/drug effects , DNA, Mitochondrial/drug effects , In Situ Hybridization , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C3H , Mitochondria, Liver/enzymology , Peroxynitrous Acid/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Tumor Necrosis Factor-alpha/physiology
19.
Am J Physiol Gastrointest Liver Physiol ; 288(5): G880-6, 2005 May.
Article in English | MEDLINE | ID: mdl-15576625

ABSTRACT

The hypothesis that the neutrophil chemoattractant CXC chemokines KC and macrophage inflammatory protein-2 (MIP-2) are involved in neutrophil transmigration and liver injury was tested in C3Heb/FeJ mice treated with galactosamine (Gal, 700 mg/kg), endotoxin (ET, 100 microg/kg), or Gal + ET (Gal/ET). Hepatic KC and MIP-2 mRNA levels and plasma CXC chemokine concentrations were dramatically increased 1.5 h after Gal/ET or ET alone and gradually declined up to 7 h. Murine recombinant cytokines (TNF-alpha, IL-1 alpha, and IL-1 beta), but not Gal/ET, induced CXC chemokine formation in the ET-resistant C3H/HeJ strain. To assess the functional importance of KC and MIP-2, C3Heb/FeJ mice were treated with Gal/ET and control IgG or a combination of anti-KC and anti-MIP-2 antibodies. Anti-CXC chemokine antibodies did not attenuate hepatocellular apoptosis, sinusoidal neutrophil sequestration and extravasation, or liver injury at 7 h. Furthermore, there was no difference in liver injury between BALB/cJ wild-type and CXC receptor-2 gene knockout (CXCR2-/-) mice treated with Gal/ET. The higher neutrophil count in livers of CXCR2-/- than in wild-type mice after Gal/ET was caused by the elevated number of neutrophils located in sinusoids of untreated CXCR2-/- animals. The pancaspase inhibitor Z-Val-Ala-Asp-fluoromethylketone eliminated Gal/ET-induced apoptosis and neutrophil extravasation and injury but not CXC chemokine formation. Thus Gal/ET induced massive, cytokine-dependent CXC chemokine formation in the liver. However, neutrophil extravasation and injury occurred in response to apoptotic cell injury at 6-7 h and was independent of CXC chemokine formation.


Subject(s)
Chemokines, CXC/physiology , Endotoxemia/physiopathology , Liver/physiopathology , Neutrophils/physiology , Animals , Chemokine CXCL2 , Chemokines/physiology , Chemokines, CXC/biosynthesis , Endotoxins/physiology , Galactosamine/physiology , Interleukin-1/physiology , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Time Factors , Tumor Necrosis Factor-alpha/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...