Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Arch Toxicol ; 97(9): 2303-2328, 2023 09.
Article in English | MEDLINE | ID: mdl-37402810

ABSTRACT

Genotoxicity data are mainly interpreted in a qualitative way, which typically results in a binary classification of chemical entities. For more than a decade, there has been a discussion about the need for a paradigm shift in this regard. Here, we review current opportunities, challenges and perspectives for a more quantitative approach to genotoxicity assessment. Currently discussed opportunities mainly include the determination of a reference point (e.g., a benchmark dose) from genetic toxicity dose-response data, followed by calculation of a margin of exposure (MOE) or derivation of a health-based guidance value (HBGV). In addition to new opportunities, major challenges emerge with the quantitative interpretation of genotoxicity data. These are mainly rooted in the limited capability of standard in vivo genotoxicity testing methods to detect different types of genetic damage in multiple target tissues and the unknown quantitative relationships between measurable genotoxic effects and the probability of experiencing an adverse health outcome. In addition, with respect to DNA-reactive mutagens, the question arises whether the widely accepted assumption of a non-threshold dose-response relationship is at all compatible with the derivation of a HBGV. Therefore, at present, any quantitative genotoxicity assessment approach remains to be evaluated case-by-case. The quantitative interpretation of in vivo genotoxicity data for prioritization purposes, e.g., in connection with the MOE approach, could be seen as a promising opportunity for routine application. However, additional research is needed to assess whether it is possible to define a genotoxicity-derived MOE that can be considered indicative of a low level of concern. To further advance quantitative genotoxicity assessment, priority should be given to the development of new experimental methods to provide a deeper mechanistic understanding and a more comprehensive basis for the analysis of dose-response relationships.


Subject(s)
DNA Damage , Mutagens , Mutagens/toxicity , Mutagens/analysis , DNA , Risk Assessment , Mutagenicity Tests/methods
2.
EFSA J ; 20(Suppl 2): e200920, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36531281

ABSTRACT

In the EU, any material or article intended to come into contact with food, which is placed on the market, has to comply with the requirements of the Regulation (EC) No 1935/2004 - the so called 'framework regulation' for food contact materials (FCM). FCM covers a wide range of materials, including plastics, paper, metal and glass, which contain chemicals that might migrate into food. These chemicals must not migrate into the foodstuff in quantities that could endanger human health, bring about an unacceptable change in the composition of the food, or bring about a deterioration in the organoleptic characteristics thereof. Despite of this general regulation, the safety of new and specific materials that are not covered must be assessed case-by-case. In addition, national authorities can set their own regulations, and in this context, the BfR sets recommendations, which are not legal norms, but represent a standard for the production of materials not subjected to any specific legislation and are well accepted by other European Commission member states according to the mutual recognition principle. The BfR Unit 74 is responsible not only to deal with chemical risk assessment of FCM but also to evaluate application dossiers to include new substances in the positive list of FCM chemicals. In the proposed EU-FORA programme, the fellow had the opportunity to gain experience in the evaluation of toxicological data from applicant dossiers and in the methodologies of migration tests performed in the laboratories. Moreover, the fellow also made a bibliographic review on scientific literature on the migration studies from starch-based materials.

3.
Arch Toxicol ; 96(11): 3013-3032, 2022 11.
Article in English | MEDLINE | ID: mdl-35963937

ABSTRACT

Styrene oligomers (SO) are well-known side products formed during styrene polymerization. They consist mainly of dimers (SD) and trimers (ST) that have been shown to be still residual in polystyrene (PS) materials. In this study migration of SO from PS into sunflower oil at temperatures between 5 and 70 °C and contact times between 0.5 h and 10 days was investigated. In addition, the contents of SD and ST in the fatty foodstuffs créme fraiche and coffee cream, which are typically enwrapped in PS, were measured and the amounts detected (of up to 0.123 mg/kg food) were compared to literature data. From this comparison, it became evident, that the levels of SO migrating from PS packaging into real food call for a comprehensive risk assessment. As a first step towards this direction, possible genotoxicity has to be addressed. Due to technical and experimental limitations, however, the few existing in vitro tests available are unsuited to provide a clear picture. In order to reduce uncertainty of these in vitro tests, four different knowledge and statistics-based in silico tools were applied to such SO that are known to migrate into food. Except for SD4 all evaluated SD and ST showed no alert for genotoxicity. For SD4, either the predictions were inconclusive or the substance was assigned as being out of the chemical space (out of domain) of the respective in silico tool. Therefore, the absence of genotoxicity of SD4 requires additional experimental proof. Apart from SD4, in silico studies supported the limited in vitro data that indicated the absence of genotoxicity of SO. In conclusion, the overall migration of all SO together into food of up to 50 µg/kg does not raise any health concerns, given the currently available in silico and in vitro data.


Subject(s)
Food Contamination , Polystyrenes , Coffee , Food Contamination/analysis , Food Packaging , Polystyrenes/chemistry , Polystyrenes/toxicity , Sunflower Oil
4.
EFSA J ; 20(Suppl 1): e200408, 2022 May.
Article in English | MEDLINE | ID: mdl-35634565

ABSTRACT

Food contact materials (FCMs) are materials and articles intended to be placed in direct or indirect contact with foodstuffs, or which can reasonably be expected to come into contact with food under normal or foreseeable conditions of use. Substances intentionally used to manufacture FCMs, as well as non-intentionally added substances resulting from impurities, by-products and/or degradation products, can migrate from FMCs into food and, consequently, are taken up by humans. To protect consumers' health, EU legislation requires that FCMs must be sufficiently inert to prevent substances from being transferred into the food in quantities that could endanger human health. At the German Federal Institute for Risk Assessment (BfR), Unit 74 'Safety of Food Contact Materials' deals with the risk assessment of FCMs and provides recommendations on the use of substances for the production of FCMs for which no specific European measures exist yet (e.g. silicone, rubber, paper and board). The BfR 'Recommendations on Food Contact Materials' are not legally binding; however, they represent the current state of the scientific and technical knowledge for the conditions under which these materials meet the requirements for consumer safety. As part of the EU-FORA programme, the fellow was involved in the risk assessment tasks and projects undertaken by Unit 74, which include: (i) the scientific evaluation of analytical and toxicological data from dossiers for adding new substances to the database 'BfR Recommendations on Food Contact Materials'; (ii) the hazard assessment of cyclic volatile methylsiloxanes (cVMS) migrating from silicone FCMs into foodstuff; and (iii) in vitro metabolic stability study of cyclic methylsiloxanes in the presence of S9 fraction, performed in the BfR laboratories. Moreover, the EU-FORA fellowship was a great opportunity for the fellow to build a strong network of food safety experts and to be part of an international community of risk assessment professionals.

5.
Toxicol In Vitro ; 73: 105134, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33662514

ABSTRACT

The liver is composed of different cell populations. Interactions of different cell populations can be investigated by a newly established indirect co-culture system consisting of immortalised primary human hepatocytes and human monocyte derived macrophages (MDMs). Using the time-dependent cytokine secretion of the co-cultures and single cultures, correlation networks (including the cytokines G-CSF, CCL3, MCP-1, CCL20, FGF, TGF-ß1, GM-CSF, IL-8 IL-6, IL-1ß, and IL-18) were generated and the correlations were validated by application of IL-8 and TNF-α-neutralising antibodies. The data reveal that IL-8 is crucial for the interaction between hepatocytes and macrophages in vitro. In addition, transcriptome analyses showed that a change in the ratio between macrophages and hepatocytes may trigger pro-inflammatory signalling pathways of the acute phase response and the complement system (release of, e.g., certain cyto- and chemokines). Using diclofenac and LPS showed that the release of cytokines is increasing with higher ratios of MDMs. Altogether, we could demonstrate that the current co-culture system is better suited to mirror the in vivo situation when compared to previously established co-culture systems composed of HepG2 and differentiated THP-1 cells. Further, our data reveal that the cytokine IL-8 is crucial for the interaction between hepatocytes and macrophages in vitro.


Subject(s)
Coculture Techniques , Cytokines/metabolism , Hepatocytes/metabolism , Macrophages/metabolism , Cell Differentiation , Cells, Cultured , Gene Expression , Gene Expression Profiling , Humans
6.
Toxicol In Vitro ; 68: 104957, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32739440

ABSTRACT

HepG2 and THP-1 cells, the latter differentiated by phorbol 12-myristate 13-acetate (PMA), were co-cultured and characterized for typical liver-specific functions, such as xenobiotic detoxification, lipid and cholesterol metabolism. Furthermore, liver injury-associated pathways, such as inflammation, were studied. In general, the co-cultivation of these cells produced a pro-inflammatory system, as indicated by increased levels of cytokines (IL-8, TGF-α, IL-6, GM-CSF, G-CSF, TGF-ß, and hFGF) in the respective supernatant. Increased expression levels of target genes of the aryl hydrocarbon receptor (AHR), e.g., CYP1A1, CYP1A2 and CYP1B1, were detected, accompanied by the increased enzyme activity of CYP1A1. Moreover, transcriptome analyses indicated a significant upregulation of cholesterol biosynthesis, which could be reduced to baseline levels by lovastatin. In contrast, total de novo lipid synthesis was reduced in co-cultured HepG2 cells. Key events of the adverse outcome pathway (AOP) for fibrosis were activated by the co-cultivation, however, no increase in the concentration of extracellular collagen was detected. This indicates, that AOP should be used with care. In summary, the indirect co-culture of HepG2/THP-1 cells results in an increased release of pro-inflammatory cytokines, an activation of the AHR pathway and an increased enzymatic CYP1A activity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cytokines/metabolism , Receptors, Aryl Hydrocarbon/genetics , Adenosine Triphosphate/metabolism , Cell Differentiation , Cholesterol/metabolism , Coculture Techniques , Collagen/metabolism , Cytochrome P-450 Enzyme System/genetics , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Signal Transduction , THP-1 Cells , Transcriptome
8.
Arch Toxicol ; 93(12): 3503-3521, 2019 12.
Article in English | MEDLINE | ID: mdl-31659427

ABSTRACT

Aluminium is one of the most abundant elements in earth's crust and its manifold uses result in an exposure of the population from many sources. Developmental toxicity, effects on the urinary tract and neurotoxicity are known effects of aluminium and its compounds. Here, we assessed the health risks resulting from total consumer exposure towards aluminium and various aluminium compounds, including contributions from foodstuffs, food additives, food contact materials (FCM), and cosmetic products. For the estimation of aluminium contents in foodstuff, data from the German "Pilot-Total-Diet-Study" were used, which was conducted as part of the European TDS-Exposure project. These were combined with consumption data from the German National Consumption Survey II to yield aluminium exposure via food for adults. It was found that the average weekly aluminium exposure resulting from food intake amounts to approx. 50% of the tolerable weekly intake (TWI) of 1 mg/kg body weight (bw)/week, derived by the European Food Safety Authority (EFSA). For children, data from the French "Infant Total Diet Study" and the "Second French Total Diet Study" were used to estimate aluminium exposure via food. As a result, the TWI can be exhausted or slightly exceeded-particularly for infants who are not exclusively breastfed and young children relying on specially adapted diets (e.g. soy-based, lactose free, hypoallergenic). When taking into account the overall aluminium exposure from foods, cosmetic products (cosmetics), pharmaceuticals and FCM from uncoated aluminium, a significant exceedance of the EFSA-derived TWI and even the PTWI of 2 mg/kg bw/week, derived by the Joint FAO/WHO Expert Committee on Food Additives, may occur. Specifically, high exposure levels were found for adolescents aged 11-14 years. Although exposure data were collected with special regard to the German population, it is also representative for European and comparable to international consumers. From a toxicological point of view, regular exceedance of the lifetime tolerable aluminium intake (TWI/PTWI) is undesirable, since this results in an increased risk for health impairments. Consequently, recommendations on how to reduce overall aluminium exposure are given.


Subject(s)
Aluminum/toxicity , Environmental Exposure/adverse effects , Risk Assessment/methods , Adolescent , Aluminum/pharmacokinetics , Animals , Carcinogens/toxicity , Child , Child, Preschool , Dietary Exposure/adverse effects , Dietary Exposure/analysis , Environmental Exposure/analysis , Food Additives/adverse effects , Food Contamination/analysis , Humans , Infant , Mutagens/toxicity , Toxicity Tests, Acute
9.
Toxicol In Vitro ; 40: 153-160, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28089782

ABSTRACT

The azo dye o-anisidine is known as an industrial and environmental pollutant. Metabolites of o-anisidine remain in the liver for >24h. However, the toxicological impact of o-anisidine on the liver and its individual cell types, e.g., hepatocytes and immune cells, is currently poorly understood. A novel co-culture system, composed of HepG2 or Huh-7 cells, and differentiated THP-1 cells was used to study the metabolic capacity towards o-anisidine, and compared to primary murine hepatocytes which express high enzyme activities. As model compounds the carcinogenic arylamine o-anisidine and its non-carcinogenic isomer, p-anisidine, as well as caffeine were used. Global proteome analysis revealed an activation of eIF2 and Nrf2-mediated oxidative stress response pathways only in co-cultures after treatment with o-anisidine. This was confirmed via detection of reactive oxygen species. In addition, the mitochondrial membrane potential decreased already after 3h treatment of cells, which correlated with a decrease of ATP levels (R2>0.92). In the supernatant of co-cultured, but not single-cultured HepG2 and Huh-7 cells, o-anisidine caused increases of damage-associated proteins, such as HMGB1 (high mobility group box-1) protein. In summary, only co-cultures of HepG2 and THP-1 cells predict o-anisidine induced stress responsive pathways, since the system has a higher sensitivity compared to single cultured cells.


Subject(s)
Aniline Compounds/toxicity , Carcinogens/toxicity , NF-E2-Related Factor 2/metabolism , Adenosine Triphosphate/metabolism , Caffeine/toxicity , Cell Line , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Urinary Bladder Neoplasms/metabolism
10.
Arch Toxicol ; 91(2): 799-810, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26965496

ABSTRACT

Liver injury as a result of a sterile inflammation is closely linked to the activation of immune cells, including macrophages, by damaged hepatocytes. This interaction between immune cells and hepatocytes is as yet not considered in any of the in vitro test systems applied during the generation of new drugs. Here, we established and characterized a novel in vitro co-culture model with two human cell lines, HepG2 and differentiated THP-1. Ketoconazole, an antifungal drug known for its hepatotoxicity, was used as a model compound in the testing of the co-culture. Single cultures of HepG2 and THP-1 cells were studied as controls. Different metabolism patterns of ketoconazole were observed for the single and co-culture incubations as well as for the different cell types. The main metabolite N-deacetyl ketoconazole was found in cell pellets, but not in supernatants of cell cultures. Global proteome analysis showed that the NRF2-mediated stress response and the CXCL8 (IL-8) pathway were induced by ketoconazole treatment under co-culture conditions. The upregulation and ketoconazole-induced secretion of several pro-inflammatory cytokines, including CXCL8, TNF-α and CCL3, was observed in the co-culture system only, but not in single cell cultures. Taking together, we provide evidence that the co-culture model applied might be suitable to serve as tool for the prediction of chemical-induced sterile inflammation in liver tissue in vivo.


Subject(s)
Chemical and Drug Induced Liver Injury/pathology , Ketoconazole/adverse effects , Toxicity Tests/methods , Chemical and Drug Induced Liver Injury/metabolism , Coculture Techniques , Hep G2 Cells/drug effects , Humans , Interleukin-8/metabolism , Ketoconazole/analogs & derivatives , Ketoconazole/metabolism , Ketoconazole/pharmacokinetics , Membrane Potential, Mitochondrial/drug effects , NF-kappa B/metabolism , Proteins/analysis , Proteins/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
Elife ; 52016 05 17.
Article in English | MEDLINE | ID: mdl-27185526

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease in industrialized countries and is increasing in prevalence. The pathomechanisms, however, are poorly understood. This study assessed the unexpected role of the Hedgehog pathway in adult liver lipid metabolism. Using transgenic mice with conditional hepatocyte-specific deletion of Smoothened in adult mice, we showed that hepatocellular inhibition of Hedgehog signaling leads to steatosis by altering the abundance of the transcription factors GLI1 and GLI3. This steatotic 'Gli-code' caused the modulation of a complex network of lipogenic transcription factors and enzymes, including SREBP1 and PNPLA3, as demonstrated by microarray analysis and siRNA experiments and could be confirmed in other steatotic mouse models as well as in steatotic human livers. Conversely, activation of the Hedgehog pathway reversed the "Gli-code" and mitigated hepatic steatosis. Collectively, our results reveal that dysfunctions in the Hedgehog pathway play an important role in hepatic steatosis and beyond.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Lipid Metabolism , Liver/metabolism , Nerve Tissue Proteins/metabolism , Non-alcoholic Fatty Liver Disease/physiopathology , Signal Transduction , Smoothened Receptor/metabolism , Zinc Finger Protein GLI1/metabolism , Animals , Gene Expression Profiling , Humans , Mice , Mice, Transgenic , Microarray Analysis , Smoothened Receptor/deficiency , Zinc Finger Protein Gli3
12.
J Hepatol ; 64(4): 860-71, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26639393

ABSTRACT

BACKGROUND & AIMS: Recently, spatial-temporal/metabolic mathematical models have been established that allow the simulation of metabolic processes in tissues. We applied these models to decipher ammonia detoxification mechanisms in the liver. METHODS: An integrated metabolic-spatial-temporal model was used to generate hypotheses of ammonia metabolism. Predicted mechanisms were validated using time-resolved analyses of nitrogen metabolism, activity analyses, immunostaining and gene expression after induction of liver damage in mice. Moreover, blood from the portal vein, liver vein and mixed venous blood was analyzed in a time dependent manner. RESULTS: Modeling revealed an underestimation of ammonia consumption after liver damage when only the currently established mechanisms of ammonia detoxification were simulated. By iterative cycles of modeling and experiments, the reductive amidation of alpha-ketoglutarate (α-KG) via glutamate dehydrogenase (GDH) was identified as the lacking component. GDH is released from damaged hepatocytes into the blood where it consumes ammonia to generate glutamate, thereby providing systemic protection against hyperammonemia. This mechanism was exploited therapeutically in a mouse model of hyperammonemia by injecting GDH together with optimized doses of cofactors. Intravenous injection of GDH (720 U/kg), α-KG (280 mg/kg) and NADPH (180 mg/kg) reduced the elevated blood ammonia concentrations (>200 µM) to levels close to normal within only 15 min. CONCLUSION: If successfully translated to patients the GDH-based therapy might provide a less aggressive therapeutic alternative for patients with severe hyperammonemia.


Subject(s)
Hyperammonemia/drug therapy , Liver Diseases/drug therapy , Animals , Glutamate Dehydrogenase/physiology , Ketoglutaric Acids/therapeutic use , Male , Mice , Mice, Inbred C57BL
13.
Arch Toxicol ; 89(11): 2069-78, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26438405

ABSTRACT

The rodent liver eliminates toxic ammonia. In mammals, three enzymes (or enzyme systems) are involved in this process: glutaminase, glutamine synthetase and the urea cycle enzymes, represented by carbamoyl phosphate synthetase. The distribution of these enzymes for optimal ammonia detoxification was determined by numerical optimization. This in silico approach predicted that the enzymes have to be zonated in order to achieve maximal removal of toxic ammonia and minimal changes in glutamine concentration. Using 13 compartments, representing hepatocytes, the following predictions were generated: glutamine synthetase is active only within a narrow pericentral zone. Glutaminase and carbamoyl phosphate synthetase are located in the periportal zone in a non-homogeneous distribution. This correlates well with the paradoxical observation that in a first step glutamine-bound ammonia is released (by glutaminase) although one of the functions of the liver is detoxification by ammonia fixation. The in silico approach correctly predicted the in vivo enzyme distributions also for non-physiological conditions (e.g. starvation) and during regeneration after tetrachloromethane (CCl4) intoxication. Metabolite concentrations of glutamine, ammonia and urea in each compartment, representing individual hepatocytes, were predicted. Finally, a sensitivity analysis showed a striking robustness of the results. These bioinformatics predictions were validated experimentally by immunohistochemistry and are supported by the literature. In summary, optimization approaches like the one applied can provide valuable explanations and high-quality predictions for in vivo enzyme and metabolite distributions in tissues and can reveal unknown metabolic functions.


Subject(s)
Ammonia/metabolism , Computer Simulation , Hepatocytes/metabolism , Liver/metabolism , Animals , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Glutamate-Ammonia Ligase , Glutaminase , Glutamine/metabolism , Immunohistochemistry , Inactivation, Metabolic/physiology , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Urea/metabolism
14.
Arch Toxicol ; 89(6): 823-50, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25820917

ABSTRACT

The advent of new testing systems and "omics"-technologies has left regulatory toxicology facing one of the biggest challenges for decades. That is the question whether and how these methods can be used for regulatory purposes. The new methods undoubtedly enable regulators to address important open questions of toxicology such as species-specific toxicity, mixture toxicity, low-dose effects, endocrine effects or nanotoxicology, while promising faster and more efficient toxicity testing with the use of less animals. Consequently, the respective assays, methods and testing strategies are subject of several research programs worldwide. On the other hand, the practical application of such tests for regulatory purposes is a matter of ongoing debate. This document summarizes key aspects of this debate in the light of the European "regulatory status quo", while elucidating new perspectives for regulatory toxicity testing.


Subject(s)
Animal Testing Alternatives/methods , Toxicity Tests/methods , Toxicology/methods , Animal Testing Alternatives/legislation & jurisprudence , Animals , Europe , Government Regulation , Humans , Species Specificity , Toxicity Tests/standards , Toxicity Tests/trends , Toxicology/legislation & jurisprudence , Toxicology/standards , Toxicology/trends , United States
15.
J Pharm Sci ; 104(1): 191-206, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25393841

ABSTRACT

Transfer of knowledge along the different phases of drug development is a fundamental process in pharmaceutical research. In particular, cross-species extrapolation between different laboratory animals and further on to first-in-human trials is challenging because of the uncertain comparability of physiological processes. Physiologically based pharmacokinetic (PBPK) modeling allows translation of mechanistic knowledge from one species to another by specifically considering physiological and biochemical differences in between. We here evaluated different knowledge-driven approaches for cross-species extrapolation by systematically incorporating specific model parameter domains of a target species into the PBPK model of a reference species. Altogether, 15 knowledge-driven approaches were applied to murine and human PBPK models of 10 exemplary drugs resulting in 300 different extrapolations. Statistical analysis of the quality of the different extrapolations revealed not only species-specific physiology as the key determinant in cross-species extrapolation but also identified a synergistic effect when considering both kinetic rate constants and gene expression profiles of relevant enzymes and transporters. Moreover, we show that considering species-specific physiology, plasma protein binding, enzyme and transport kinetics, as well as tissue-specific gene expression profiles in PBPK modeling increases accuracy of cross-species extrapolations and thus supports first-in-human trials based on prior preclinical knowledge.


Subject(s)
Drug Evaluation, Preclinical/methods , Drugs, Investigational/pharmacokinetics , Gene Expression Regulation/drug effects , Liver/drug effects , Models, Biological , Pharmacology, Clinical/methods , Physiology, Comparative/methods , Animals , Cells, Cultured , Computational Biology , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Drugs, Investigational/metabolism , Drugs, Investigational/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Enzymologic/drug effects , Germany , Humans , Liver/cytology , Liver/enzymology , Liver/metabolism , Mice, Inbred C57BL , Organ Specificity , Species Specificity , Specific Pathogen-Free Organisms
16.
Hepatology ; 60(6): 2040-51, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24677161

ABSTRACT

UNLABELLED: The impairment of hepatic metabolism due to liver injury has high systemic relevance. However, it is difficult to calculate the impairment of metabolic capacity from a specific pattern of liver damage with conventional techniques. We established an integrated metabolic spatial-temporal model (IM) using hepatic ammonia detoxification as a paradigm. First, a metabolic model (MM) based on mass balancing and mouse liver perfusion data was established to describe ammonia detoxification and its zonation. Next, the MM was combined with a spatial-temporal model simulating liver tissue damage and regeneration after CCl4 intoxication. The resulting IM simulated and visualized whether, where, and to what extent liver damage compromised ammonia detoxification. It allowed us to enter the extent and spatial patterns of liver damage and then calculate the outflow concentrations of ammonia, glutamine, and urea in the hepatic vein. The model was validated through comparisons with (1) published data for isolated, perfused livers with and without CCl4 intoxication and (2) a set of in vivo experiments. Using the experimentally determined portal concentrations of ammonia, the model adequately predicted metabolite concentrations over time in the hepatic vein during toxin-induced liver damage and regeneration in rodents. Further simulations, especially in combination with a simplified model of blood circulation with three ammonia-detoxifying compartments, indicated a yet unidentified process of ammonia consumption during liver regeneration and revealed unexpected concomitant changes in amino acid metabolism in the liver and at extrahepatic sites. CONCLUSION: The IM of hepatic ammonia detoxification considerably improves our understanding of the metabolic impact of liver disease and highlights the importance of integrated modeling approaches on the way toward virtual organisms.


Subject(s)
Ammonia/metabolism , Liver Diseases/metabolism , Liver Regeneration , Models, Biological , Animals , In Vitro Techniques , Inactivation, Metabolic , Male , Mice, Inbred C57BL , Perfusion
17.
BMC Syst Biol ; 6: 147, 2012 Nov 29.
Article in English | MEDLINE | ID: mdl-23190768

ABSTRACT

BACKGROUND: Network inference is an important tool to reveal the underlying interactions of biological systems. In the liver, a complex system of transcription factors is active to distribute signals and induce the cellular response following extracellular stimuli. Plenty of information is available about single transcription factors important for the different functions of the liver, but little is known about their causal relations to each other. RESULTS: Given a DNA microarray time series dataset of collagen monolayers cultured murine hepatocytes, we identified 22 differentially expressed genes for which the corresponding protein is known to exhibit transcription factor activity. We developed the Extended TILAR (ExTILAR) network inference algorithm based on the modeling concept of the previously published TILAR algorithm. Using ExTILAR, we inferred a transcription factor network based on gene expression data which puts these important genes into a functional context. This way, we identified a previously unknown relationship between Tgif1 and Atf3 which we validated experimentally. Beside its known role in metabolic processes, this extends the knowledge about Tgif1 in hepatocytes towards a possible influence of processes such as proliferation and cell cycle. Moreover, two positive (i.e. double negative) regulatory loops were predicted that could give rise to bistable behavior. We further evaluated the performance of ExTILAR by systematic inference of an in silico network. CONCLUSIONS: We present the ExTILAR algorithm, which combines the advantages of the regression based inference algorithm TILAR, like large network sizes processable and low computational costs, with the advantages of dynamic network models based on ordinary differential equation (i.e. in silico knock-down simulations). Like TILAR, ExTILAR makes use of various prior-knowledge types such as transcription factor binding site information and gene interaction knowledge to infer biologically meaningful gene regulatory networks. Therefore, ExTILAR is especially useful when a large number of genes is modeled using a small number of experimental data points.


Subject(s)
Gene Expression Regulation , Gene Regulatory Networks , Hepatocytes/cytology , Hepatocytes/metabolism , Models, Biological , Systems Biology/methods , Transcription Factors/metabolism , Animals , Cell Culture Techniques , Cell Cycle/genetics , Cell Proliferation , Mice , Reproducibility of Results
18.
Nat Cell Biol ; 14(8): 829-37, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22820375

ABSTRACT

The PI(3)K-PKB-FOXO signalling network provides a major intracellular hub for the regulation of cell proliferation, survival and stress resistance. Here we report an unexpected role for FOXO transcription factors in regulating autophagy by modulating intracellular glutamine levels. To identify transcriptional targets of this network, we performed global transcriptional analyses after conditional activation of the key components PI(3)K, PKB/Akt, FOXO3 and FOXO4. Using this pathway approach, we identified glutamine synthetase as being transcriptionally regulated by PI(3)K-PKB-FOXO signalling. Conditional activation of FOXO also led to an increased level of glutamine production. FOXO activation resulted in mTOR inhibition by preventing the translocation of mTOR to lysosomal membranes in a glutamine-synthetase-dependent manner. This resulted in an increased level of autophagy as measured by LC3 lipidation, p62 degradation and fluorescent imaging of multiple autophagosomal markers. Inhibition of FOXO3-mediated autophagy increased the level of apoptosis, suggesting that the induction of autophagy by FOXO3-mediated glutamine synthetase expression is important for cellular survival. These findings reveal a growth-factor-responsive network that can directly modulate autophagy through the regulation of glutamine metabolism.


Subject(s)
Autophagy , Forkhead Transcription Factors/metabolism , Glutamine/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Base Sequence , Blotting, Western , Cell Proliferation , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Glutamate Synthase/genetics , Glutamate Synthase/metabolism , Humans , Mice , Microarray Analysis , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/genetics , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/genetics , Rats , Sequence Alignment , Signal Transduction
19.
Rapid Commun Mass Spectrom ; 25(18): 2619-26, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-23657956

ABSTRACT

The liver is an important organ that is particularly involved in the lipid metabolism of the organism. Thus, high interest is nowadays focused on the lipid composition of the liver and particularly the liver parenchymal cells, the hepatocytes. Hepatocytes contain common phospholipids (PL) such as phosphatidylcholines, -ethanolamines and -inositols, for instance, that can be easily analyzed by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) even without previous separation of the PL mixture. However, in addition to common PL, hepatocytes possess also significant amounts of cardiolipin (CLP). The MS analysis of this PL is quite challenging because it (a) has a higher mass than common lipids and (b) possesses a higher negative charge. We will show here that caution is required if CLP is analyzed directly from the total lipid extract because PC dimers may be interpreted as cardiolipins if the positive ion MALDI mass spectra are analyzed.


Subject(s)
Cardiolipins/chemistry , Complex Mixtures/chemistry , Hepatocytes/chemistry , Phosphatidylcholines/chemistry , Tandem Mass Spectrometry/methods , Animals , Cardiolipins/metabolism , Cells, Cultured , Complex Mixtures/metabolism , Dimerization , Hepatocytes/metabolism , Lipids/chemistry , Male , Mice , Mice, Inbred C57BL , Phosphatidylcholines/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
20.
Hepatology ; 52(6): 2127-36, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20979052

ABSTRACT

UNLABELLED: The cellular basis of liver regeneration has been intensely investigated for many years. However, the mechanisms initiating hepatocyte "plasticity" and priming for proliferation are not yet fully clear. We investigated alterations in gene expression patterns during the first 72 hours of C57BL/6N mouse hepatocyte culture on collagen monolayers (CM), which display a high basal frequency of proliferation in the absence of cytokines. Although many metabolic genes were down-regulated, genes related to mitogen-activated protein kinase (MAPK) signaling and cell cycle were up-regulated. The latter genes showed an overrepresentation of transcription factor binding sites (TFBS) for ETF (TEA domain family member 2), E2F1 (E2F transcription factor 1), and SP-1 (Sp1 transcription factor) (P < 0.001), all depending on MAPK signaling. Time-dependent increase of ERK1/2 phosphorylation occurred during the first 48 hours (and beyond) in the absence of cytokines, accompanied by an enhanced bromodeoxyuridine labeling index of 20%. The MEK inhibitor PD98059 blunted these effects indicating MAPK signaling as major trigger for this cytokine-independent proliferative response. In line with these in vitro findings, liver tissue of mice challenged with CCl(4) displayed hepatocytes with intense p-ERK1/2 staining and nuclear SP-1 and E2F1 expression. Furthermore, differentially expressed genes in mice after partial hepatectomy contained overrepresented TFBS for ETF, E2F1, and SP-1 and displayed increased expression of E2F1. CONCLUSION: Cultivation of murine hepatocytes on CM primes cells for proliferation through cytokine-independent activation of MAPK signaling. The transcription factors ETF, E2F1, and SP-1 seem to play a pronounced role in mediating proliferation-dependent differential gene expression. Similar events, but on a shorter time-scale, occur very early after liver damage in vivo.


Subject(s)
Cell Proliferation , DNA-Binding Proteins/physiology , E2F1 Transcription Factor/physiology , Hepatocytes/cytology , Sp1 Transcription Factor/physiology , Transcription Factors/physiology , Animals , Binding Sites/physiology , Carbon Tetrachloride Poisoning/physiopathology , Gene Expression , Hepatectomy , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Signal Transduction/drug effects , TEA Domain Transcription Factors , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...