Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Pharmacol Exp Ther ; 336(2): 551-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21078785

ABSTRACT

Amiodarone (AM) is a potent antidysrhythmic agent that can cause potentially life-threatening pulmonary fibrosis, and N-desethylamiodarone (DEA), an AM metabolite, may contribute to AM toxicity. Apoptotic cell death in nontransformed human peripheral lung epithelial 1A (HPL1A) cells was assessed by annexin V-fluorescein isothiocyanate (ann-V) staining and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL), and necrotic cell death was assessed by propidium iodide (PI) staining. The percentage of cells that were PI-positive increased more than six times with 20 µM AM and approximately doubled with 3.5 µM DEA, relative to control. The percentage of cells that were ann-V-positive decreased by more than 80% after 24-h exposure to 10 µM AM but more than doubled after 24-h incubation with 3.5 µM DEA. Incubation for 24 h with 5.0 µM DEA increased the percentage of cells that were TUNEL-positive more than six times. Incubation with AM (2.5 µM) or DEA (1-2 µM) for 24 h did not significantly alter angiotensinogen mRNA levels. Furthermore, angiotensin II (100 pM-1 µM) alone or in combination with AM or DEA did not alter cytotoxicity, and pretreatment with the angiotensin-converting enzyme inhibitor and antioxidant captopril (3-6 µM) did not protect against AM or DEA cytotoxicity. In conclusion, AM activates primarily necrotic pathways, whereas DEA activates both necrotic and apoptotic pathways, and the renin-angiotensin system does not seem to be involved in AM or DEA cytotoxicity in HPL1A cells.


Subject(s)
Amiodarone/analogs & derivatives , Amiodarone/toxicity , Anti-Arrhythmia Agents/toxicity , Lung/drug effects , Amiodarone/metabolism , Angiotensin II/toxicity , Angiotensinogen/genetics , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/pathology , Humans , In Situ Nick-End Labeling , Lung/pathology , Necrosis , RNA, Messenger/analysis
2.
Toxicol Appl Pharmacol ; 227(3): 370-9, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18191165

ABSTRACT

Amiodarone (AM), a drug used in the treatment of cardiac dysrrhythmias, can produce severe pulmonary adverse effects, including fibrosis. Although the pathogenesis of AM-induced pulmonary toxicity (AIPT) is not clearly understood, several hypotheses have been advanced, including increased inflammatory mediator release, mitochondrial dysfunction, and free-radical formation. The hypothesis that AM induces formation of reactive oxygen species (ROS) was tested in an in vitro model relevant for AIPT. Human peripheral lung epithelial HPL1A cells, as surrogates for target cells in AIPT, were susceptible to the toxicity of AM and N-desethylamiodarone (DEA), a major AM metabolite. Longer incubations (> or =6 h) of HPL1A cells with 100 microM AM significantly increased ROS formation. In contrast, shorter incubations (2 h) of HPL1A cells with AM resulted in mitochondrial dysfunction and cytoplasmic cytochrome c translocation. Preexposure of HPL1A cells to ubiquinone and alpha-tocopherol was more effective than that with Trolox C or 5,5-dimethylpyrolidine N-oxide (DMPO) at preventing AM cytotoxicity. These data suggest that mitochondrial dysfunction, rather than ROS overproduction, represents an early event in AM-induced toxicity in peripheral lung epithelial cells that may be relevant for triggering AIPT, and antioxidants that target mitochondria may potentially have beneficial effects in AIPT.


Subject(s)
Amiodarone/toxicity , Anti-Arrhythmia Agents/toxicity , Lung/drug effects , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Amiodarone/analogs & derivatives , Amiodarone/antagonists & inhibitors , Anti-Arrhythmia Agents/antagonists & inhibitors , Cell Line , Chromans/administration & dosage , Cyclic N-Oxides/administration & dosage , Cytochromes c/metabolism , Cytoplasm/enzymology , Epithelial Cells/drug effects , Epithelial Cells/ultrastructure , Humans , Lung/metabolism , Lung/ultrastructure , Mitochondria/metabolism , Ubiquinone/administration & dosage , alpha-Tocopherol/administration & dosage
3.
Toxicol Appl Pharmacol ; 220(1): 60-71, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17316728

ABSTRACT

Amiodarone (AM), an antidysrrhythmic drug, can produce serious adverse effects, including potentially fatal AM-induced pulmonary toxicity (AIPT). AM-induced cytotoxicity and pulmonary fibrosis are well recognized, but poorly understood mechanistically. The hypothesis of aryl radical involvement in AM toxicity was tested in non-biological and biological systems. Photolysis of anaerobic aqueous solutions of AM, or N-desethylamiodarone (DEA) resulted in the formation of an aryl radical, as determined by spin-trapping and electron paramagnetic resonance (EPR) spectroscopy experiments. The non-iodinated AM analogue, didesiodoamiodarone (DDIA), did not form aryl radicals under identical conditions. The toxic susceptibility of human lung epithelioid HPL1A cells to AM, DEA, and DDIA showed time- and concentration-dependence. DEA had a more rapid and potent toxic effect (LC(50)=8 microM) than AM (LC(50)=146 microM), whereas DDIA cytotoxicity was intermediate (LC(50)=26 microM) suggesting a minor contribution of the iodine atoms. Incubation of human lung epithelial cells with the spin-trapping nitrones alpha-phenyl-N-t-butylnitrone (PBN, 10 mM) or alpha-(4-pyridyl N-oxide)-N-t-butylnitrone (POBN, 5.0 mM) did not significantly protect against AM, DEA, or DDIA cytotoxicity. Intratracheal administration of AM to hamsters produced pulmonary fibrosis at day 21, which was not prevented by 4 days of treatment with 150 mg/kg/day PBN or 164 mg/kg/day POBN. However, the body weight loss in AM-treated animals was counteracted by PBN. These results suggest that, although AM can generate an aryl radical photochemically, its in vivo formation may not be a major contributor to AM toxicity, and that spin-trapping reagents do not halt the onset of AM toxicity.


Subject(s)
Amiodarone/toxicity , Anti-Arrhythmia Agents/toxicity , Cyclic N-Oxides/pharmacology , Lung/drug effects , Pyridines/pharmacology , Animals , Cricetinae , Electron Spin Resonance Spectroscopy , Free Radicals , Hydroxyproline/analysis , Lung/chemistry , Lung/pathology , Male , Mesocricetus , Photochemistry
4.
Toxicology ; 190(1-2): 3-14, 2003 Aug 21.
Article in English | MEDLINE | ID: mdl-12909394

ABSTRACT

This paper will provide an overview of the on-line resources available in toxicology in Canada. It will describe a brief history of The Society of Toxicology of Canada, with reference to other societies and also provide information on education, research and other resources related to toxicology. Toxicology in Canada emerged as a distinct and vibrant discipline following the thalidomide tragedy of the 1960s. In the pharmaceutical industry and government, toxicology was readily established as an essential component of drug development and safety, and as the need for toxicologists expanded, training programs were established, usually in collaboration with departments of pharmacology. In the last two to three decades other disciplines, environmental biology, analytical chemistry and epidemiology joined the ranks of toxicology. The on-line sources of toxicology information are rapidly expanding. This article describes those sources considered by the authors to be important from a national and international perspective. The majority of these sources are professional organizations and government agencies.


Subject(s)
Internet , Toxicology , Canada , Chemical Phenomena , Chemistry , Epidemiology , Government Agencies , Poison Control Centers , Research Support as Topic , Societies, Scientific
5.
Toxicol Sci ; 75(1): 169-80, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12832656

ABSTRACT

Pulmonary toxicity, including fibrosis, is a serious adverse effect associated with the antidysrhythmic drug amiodarone (AM). We tested the potential usefulness of pirfenidone against AM-induced pulmonary toxicity in the hamster model. Intratracheal AM administration resulted in pulmonary fibrosis 21 days posttreatment, as evidenced by an increased hydroxyproline content and histological damage. Dietary pirfenidone administration (0.5% w/w in chow), for 3 days prior to and continuously after AM, prevented fibrosis and suppressed elevation of pulmonary transforming growth factor (TGF)-beta1 mRNA content at 7 and 21 days post-AM. Protection against AM-induced lung damage was not observed when supplementation with pirfenidone was delayed until 7 days following AM administration, suggesting that alteration of early events in AM lung toxicity is necessary for the protective effect of pirfenidone. Both AM and bleomycin, another pulmonary fibrogen, caused inflammation 24 h after intratracheal dosing, measured as increased lactate dehydrogenase activity, protein content, and cellular alterations in bronchoalveolar lavage fluid, with the response to AM markedly greater than that to bleomycin. Administration of AM, but not bleomycin, also caused whole lung mitochondrial dysfunction, alveolar macrophage death, and an influx of eosinophils into the lung, of which pirfenidone was able to decrease only the latter. We conclude that: (1) AM induces alveolar macrophage death and severe, acute pulmonary inflammation with associated eosinophilia following intratracheal administration; (2) mitochondrial dysfunction may play an early role in AM pulmonary injury; and (3) pirfenidone decreases AM-induced pulmonary fibrosis in the hamster, probably through suppression of TGF-beta1 gene expression.


Subject(s)
Amiodarone/toxicity , Anti-Arrhythmia Agents/toxicity , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Pulmonary Fibrosis/prevention & control , Pyridones/pharmacology , Acute Disease , Animals , Bleomycin/toxicity , Cricetinae , Disease Models, Animal , Hydroxyproline/metabolism , Male , Membrane Potentials/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Oxygen Consumption/drug effects , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , RNA, Messenger/metabolism , Time Factors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1
6.
J Pharmacol Exp Ther ; 304(1): 121-9, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12490582

ABSTRACT

Hepatotoxicity induced by 1,1-dichloroethylene (DCE) is mediated by cytochrome P450-dependent metabolism to reactive intermediates, including the epoxide. We have tested the hypothesis that mitochondria are a primary target of toxicity by investigating dose- and time-dependent effects of DCE on mitochondrial respiration. Hepatotoxicity, as assessed by serum alanine aminotransferase (ALT) activity, was evaluated. We have also determined the effectiveness of N-acetyl-L-cysteine (NAC) in protecting against respiratory perturbations and hepatotoxicity. Liver mitochondria were isolated 2 h after DCE (50, 75, 100, 125, and 150 mg/kg) treatment. Glutamate (complex I)- and succinate (complex II)-supported mitochondrial respiration was assessed by measurement of state 3 (ADP-stimulated) and state 4 (resting) rates of oxygen consumption. The corresponding respiratory control ratios (RCRs, state 3/state 4) and ADP:O ratios were then calculated. A DCE dose of 125 mg/kg significantly inhibited glutamate- and succinate-supported state 3 respiration, leading to a significant reduction in corresponding RCRs and ADP:O ratios. In time-dependent studies, state 3 respiration rates and RCRs for glutamate-supported respiration were significantly decreased as early as 20 min after DCE (125 mg/kg) treatment, whereas those for succinate-supported respiration were significantly decreased at 90 min. Additionally, ADP:O ratios for glutamate-supported respiration were significantly decreased starting at 60 min, and those for succinate-supported respiration at 90 min. Alterations in mitochondrial function preceded significant increases in ALT activity, which was first manifested at 2 h. Pretreatment with NAC (1200 mg/kg) abrogated DCE-induced GSH depletion and inhibited disturbances in mitochondrial respiration. Moreover, NAC protected against increased ALT activity, suggesting that the protective effect of NAC is due to increased GSH for conjugation reactions and/or its antioxidant property. These results showed that DCE-mediated mitochondrial dysfunction is an early event that preceded the onset of hepatotoxicity.


Subject(s)
Chemical and Drug Induced Liver Injury/pathology , Dichloroethylenes/toxicity , Mitochondria, Liver/pathology , Acetylcysteine/pharmacology , Adenosine Diphosphate/metabolism , Alanine Transaminase/metabolism , Animals , Dose-Response Relationship, Drug , Female , Glutathione/metabolism , In Vitro Techniques , Indicators and Reagents , Mice , Mitochondria, Liver/drug effects , Oxygen Consumption/drug effects , Polarography , Time Factors
7.
J Pharmacol Exp Ther ; 304(1): 277-83, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12490602

ABSTRACT

Amiodarone (AM) is an efficacious antidysrhythmic agent that can cause numerous adverse effects, including potentially life-threatening pulmonary fibrosis. The current study was undertaken to investigate potential protective mechanisms of vitamin E against AM-induced pulmonary toxicity (AIPT) in the hamster. Three weeks after intratracheal administration of AM (1.83 micromol), increased pulmonary hydroxyproline content and histological damage were observed, indicative of fibrosis. These effects were preceded by increased pulmonary levels of transforming growth factor (TGF)-beta1 mRNA at 1 week post-AM, which remained elevated 3 weeks post-AM. Dietary supplementation with vitamin E resulted in rapid pulmonary accumulation of the vitamin, and prevention of AM-induced increases in TGF-beta1, hydroxyproline, and histological damage. Although dietary supplementation also markedly elevated lung mitochondrial vitamin E content, it did not attenuate AM-induced inhibition of mitochondrial respiration or disruption of mitochondrial membrane potential in vitro, or lung mitochondrial respiratory inhibition resulting from in vivo AM administration. These results suggest that vitamin E reduces the extent of pulmonary damage after AM administration via down-regulating TGF-beta1 overexpression but that it does not modify AM-induced mitochondrial dysfunction, a potential initiating event in AIPT.


Subject(s)
Amiodarone/analogs & derivatives , Amiodarone/antagonists & inhibitors , Amiodarone/toxicity , Anti-Arrhythmia Agents/antagonists & inhibitors , Anti-Arrhythmia Agents/toxicity , Antioxidants/pharmacology , Gene Expression/drug effects , Mitochondria/drug effects , Pulmonary Fibrosis/prevention & control , Transforming Growth Factor beta/biosynthesis , Vitamin E/pharmacology , Animals , Cricetinae , DNA Probes , Diet , Hydroxyproline/metabolism , In Situ Hybridization , Lung/chemistry , Lung/pathology , Male , Membrane Potentials/drug effects , Mesocricetus , Oxygen Consumption , Polarography , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , RNA, Messenger/biosynthesis , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1
SELECTION OF CITATIONS
SEARCH DETAIL
...