Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Toxicol Lett ; 234(2): 139-50, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25704631

ABSTRACT

Acetaminophen (APAP) overdosage results in hepatotoxicity, but the underlying molecular mechanisms are still not completely understood. In the current study, we focused on mitochondrial-specific oxidative liver injury induced by APAP exposure. Owning to genetic polymorphisms in the CYP2E1 gene or varying inducibility by xenobiotics, the CYP2E1 mRNA level and protein activity vary extensively among individuals. As CYP2E1 is a known ROS generating enzyme, we chose HepG2 to minimize CYP2E1-induced ROS formation, which will help us better understand the APAP induced mitochondrial-specific hepatotoxicity in a subpopulation with low CYP2E1 activity. HepG2 cells were exposed to a low and toxic dose (0.5 and 10mM) of APAP and analyzed at four time points for genome-wide gene expression. Mitochondria were isolated and electron spin resonance spectroscopy was performed to measure the formation of mitochondrial ROS. The yield of ATP was measured to confirm the impact of the toxic dose of APAP on cellular energy production. Our results indicate that 10mM APAP significantly influences the expression of mitochondrial protein-encoding genes in association with an increase in mitochondrial ROS formation. Additionally, 10mM APAP affects the expression of genes encoding the subunits of electron transport chain (ETC) complexes, which may alter normal mitochondrial functions by disrupting the assembly, stability, and structural integrity of ETC complexes, leading to a measurable depletion of ATP, and cell death. The expression of mitochondrium-specific antioxidant enzyme, SOD2, is reduced which may limit the ROS scavenging ability and cause imbalance of the mitochondrial ROS homeostasis. Overall, transcriptome analysis reveals the molecular processes involved in the observed APAP-induced increase of mitochondrial ROS formation and the associated APAP-induced oxidative stress.


Subject(s)
Acetaminophen/toxicity , Chemical and Drug Induced Liver Injury/etiology , Electron Transport Chain Complex Proteins/metabolism , Hepatocytes/drug effects , Mitochondria, Liver/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Adenosine Triphosphate/metabolism , Cell Death/drug effects , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Dose-Response Relationship, Drug , Electron Transport Chain Complex Proteins/genetics , Energy Metabolism/drug effects , Gene Expression Profiling/methods , Gene Expression Regulation , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Time Factors , Toxicogenetics/methods
2.
Toxicol In Vitro ; 29(3): 489-501, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25562108

ABSTRACT

In order to improve attrition rates of candidate-drugs there is a need for a better understanding of the mechanisms underlying drug-induced hepatotoxicity. We aim to further unravel the toxicological response of hepatocytes to a prototypical cholestatic compound by integrating transcriptomic and metabonomic profiling of HepG2 cells exposed to Cyclosporin A. Cyclosporin A exposure induced intracellular cholesterol accumulation and diminished intracellular bile acid levels. Performing pathway analyses of significant mRNAs and metabolites separately and integrated, resulted in more relevant pathways for the latter. Integrated analyses showed pathways involved in cell cycle and cellular metabolism to be significantly changed. Moreover, pathways involved in protein processing of the endoplasmic reticulum, bile acid biosynthesis and cholesterol metabolism were significantly affected. Our findings indicate that an integrated approach combining metabonomics and transcriptomics data derived from representative in vitro models, with bioinformatics can improve our understanding of the mechanisms of action underlying drug-induced hepatotoxicity. Furthermore, we showed that integrating multiple omics and thereby analyzing genes, microRNAs and metabolites of the opposed model for drug-induced cholestasis can give valuable information about mechanisms of drug-induced cholestasis in vitro and therefore could be used in toxicity screening of new drug candidates at an early stage of drug discovery.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Cyclosporine/toxicity , Immunosuppressive Agents/toxicity , Chemical and Drug Induced Liver Injury/pathology , Dose-Response Relationship, Drug , Gene Expression Profiling , Hep G2 Cells , Humans , In Vitro Techniques , Metabolomics , MicroRNAs/biosynthesis , RNA, Messenger/biosynthesis , Transcriptome
3.
Toxicology ; 324: 18-26, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25047351

ABSTRACT

The liver is responsible for drug metabolism and drug-induced hepatotoxicity is the most frequent reason for drug withdrawal, indicating that better pre-clinical toxicity tests are needed. In order to bypass animal models for toxicity screening, we exposed primary mouse hepatocytes for exploring the prototypical hepatotoxicant cyclosporin A. To elucidate the mechanisms underlying cyclosporin A-induced hepatotoxicity, we analyzed expression levels of proteins, mRNAs, microRNAs and metabolites. Integrative analysis of transcriptomics and proteomics showed that protein disulfide isomerase family A, member 4 was up-regulated on both the protein level and mRNA level. This protein is involved in protein folding and secretion in the endoplasmic reticulum. Furthermore, the microRNA mmu-miR-182-5p which is predicted to interact with the mRNA of this protein, was also differentially expressed, further emphasizing endoplasmic reticulum stress as important event in drug-induced toxicity. To further investigate the interaction between the significantly expressed proteins, a network was created including genes and microRNAs known to interact with these proteins and this network was used to visualize the experimental data. In total 6 clusters could be distinguished which appeared to be involved in several toxicity related processes, including alteration of protein folding and secretion in the endoplasmic reticulum. Metabonomic analyses resulted in 5 differentially expressed metabolites, indicative of an altered glucose, lipid and cholesterol homeostasis which can be related to cholestasis. Single and integrative analyses of transcriptomics, proteomics and metabonomics reveal mechanisms underlying cyclosporin A-induced cholestasis demonstrating that endoplasmic reticulum stress and the unfolded protein response are important processes in drug-induced liver toxicity.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Cyclosporine/toxicity , Genomics , Hepatocytes/drug effects , Immunosuppressive Agents/toxicity , Liver/drug effects , Metabolomics , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Genetic Markers , Genomics/methods , Hepatocytes/metabolism , Liver/metabolism , Male , Metabolomics/methods , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Primary Cell Culture , Protein Interaction Mapping , Protein Interaction Maps , Proteomics , RNA, Messenger/metabolism , Risk Assessment , Systems Biology
4.
OMICS ; 17(2): 71-83, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23308384

ABSTRACT

Drug-induced hepatotoxicity is a leading cause of attrition for candidate pharmaceuticals in development. New preclinical screening methods are crucial to predict drug toxicity prior to human studies. Of all in vitro hepatotoxicity models, primary human hepatocytes are considered as 'the gold standard.' However, their use is hindered by limited availability and inter-individual variation. These barriers may be overcome by using primary mouse hepatocytes. We used differential in gel electrophoresis (DIGE) to study large-scale protein expression of primary mouse hepatocytes. These hepatocytes were exposed to three well-defined hepatotoxicants: acetaminophen, amiodarone, and cyclosporin A. Each hepatotoxicant induces a different hepatotoxic phenotype. Based on the DIGE results, the mRNA expression levels of deregulated proteins from cyclosporin A-treated cells were also analyzed. We were able to distinguish cyclosporin A from controls, as well as acetaminophen and amiodarone-treated samples. Cyclosporin A induced endoplasmic reticulum (ER) stress and altered the ER-Golgi transport. Moreover, liver carboxylesterase and bile salt sulfotransferase were differentially expressed. These proteins were associated with a protective adaptive response against cyclosporin A-induced cholestasis. The results of this study are comparable with effects in HepG2 cells. Therefore, we suggest both models can be used to analyze the cholestatic properties of cyclosporin A. Furthermore, this study showed a conserved response between primary mouse hepatocytes and HepG2 cells. These findings collectively lend support for use of omics strategies in preclinical toxicology, and might inform future efforts to better link preclinical and clinical research in rational drug development.


Subject(s)
Acetaminophen/pharmacology , Amiodarone/pharmacology , Cyclosporine/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Proteome , Proteomics , Acetaminophen/toxicity , Amiodarone/toxicity , Animals , Cell Line , Cluster Analysis , Cyclosporine/toxicity , Gene Expression Profiling , Gene Expression Regulation/drug effects , Genomics , Hep G2 Cells , Humans , Male , Mice , Primary Cell Culture , Proteomics/methods
5.
Toxicol In Vitro ; 26(3): 373-85, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22274661

ABSTRACT

The safety assessment for pharmaceuticals includes in vivo repeated dose toxicity tests in laboratory animals. These in vivo studies often generate false negative results and unexpected toxicity. The appearance of this unexpected toxicity is one of the major reasons for the drawback of a drug from the market. The liver is often a target organ in toxicology since it is responsible for the metabolism and elimination of chemical compounds. Therefore, there is need for new screening methods which classify hepatotoxic compounds earlier in development. This will lead to safer drugs and a more efficient drug discovery process. Furthermore, these new screening methods are preferably in vitro test systems, aiming at reducing the use of laboratory animals. In this review the possibilities of proteomics and its promising results for improving current predictive and mechanistic toxicological studies are described. Biomarkers or protein panels for hepatotoxic mechanisms, which reflect the in vivo situation, need to be identified to allow a better toxicity screening. Therefore, in vivo studies and in vitro cell models are discussed and evaluated with regard to the protein expression of their metabolic enzymes, their similarities with liver, their use for analyzing toxicological mechanisms and hepatotoxicity screening. Studies in which proteomics are combined with other omics-technologies are also presented. The results from these integrated data analyses can be used for the development of improved panels of biomarkers for toxicity screening.


Subject(s)
Biomarkers, Pharmacological/metabolism , Chemical and Drug Induced Liver Injury/etiology , Proteomics/methods , Animal Testing Alternatives/methods , Animals , Chemical and Drug Induced Liver Injury/physiopathology , Drug Design , Drug-Related Side Effects and Adverse Reactions , Humans , Pharmaceutical Preparations/administration & dosage , Toxicity Tests/methods
6.
Toxicol Sci ; 120(1): 109-22, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21163907

ABSTRACT

Unexpected hepatotoxicity is one of the major reasons of drugs failing in clinical trials. This emphasizes the need for new screening methods that address toxicological hazards early in the drug discovery process. Here, proteomics techniques were used to gain further insight into the mechanistic processes of the hepatotoxic compounds. Drug-induced hepatotoxicity is mainly divided in hepatic steatosis, cholestasis, or necrosis. For each class, a compound was selected, respectively amiodarone, cyclosporin A, and acetaminophen. The changes in protein expressions in HepG2, after exposure to these test compounds, were studied using quantitative two-dimensional differential gel electrophoresis. Identification of differentially expressed proteins was performed by Maldi-TOF/TOF MS and liquid chromatography-tandem mass spectrometry. In this study, 254 differentially expressed protein spots were detected in a two-dimensional proteome map from which 86 were identified, showing that the proteome of HepG2 cells is responsive to hepatotoxic compounds. cyclosporin A treatment was responsible for most differentially expressed proteins and could be discriminated in the hierarchical clustering analysis. The identified differential proteins show that cyclosporin A may induce endoplasmic reticulum (ER) stress and disturbs the ER-Golgi transport, with an altered vesicle-mediated transport and protein secretion as result. Moreover, the differential protein pattern seen after cyclosporin A treatment can be related to cholestatic mechanisms. Therefore, our findings indicate that the HepG2 in vitro cell system has distinctive characteristics enabling the assessment of cholestatic properties of novel compounds at an early stage of drug discovery.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Drug-Related Side Effects and Adverse Reactions , Proteins/analysis , Proteomics/methods , Acetaminophen/administration & dosage , Acetaminophen/adverse effects , Amiodarone/administration & dosage , Amiodarone/adverse effects , Blotting, Western , Cell Survival/drug effects , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Cluster Analysis , Cyclosporine/administration & dosage , Cyclosporine/adverse effects , Electrophoresis, Gel, Two-Dimensional , Hep G2 Cells , Humans , Pharmaceutical Preparations/administration & dosage , Protein Biosynthesis/drug effects , Protein Processing, Post-Translational , Proteins/genetics , Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...