Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Nanotoxicology ; 9 Suppl 1: 118-32, 2015 May.
Article in English | MEDLINE | ID: mdl-25923349

ABSTRACT

In spite of recent advances in describing the health outcomes of exposure to nanoparticles (NPs), it still remains unclear how exactly NPs interact with their cellular targets. Size, surface, mass, geometry, and composition may all play a beneficial role as well as causing toxicity. Concerns of scientists, politicians and the public about potential health hazards associated with NPs need to be answered. With the variety of exposure routes available, there is potential for NPs to reach every organ in the body but we know little about the impact this might have. The main objective of the FP7 NanoTEST project ( www.nanotest-fp7.eu ) was a better understanding of mechanisms of interactions of NPs employed in nanomedicine with cells, tissues and organs and to address critical issues relating to toxicity testing especially with respect to alternatives to tests on animals. Here we describe an approach towards alternative testing strategies for hazard and risk assessment of nanomaterials, highlighting the adaptation of standard methods demanded by the special physicochemical features of nanomaterials and bioavailability studies. The work has assessed a broad range of toxicity tests, cell models and NP types and concentrations taking into account the inherent impact of NP properties and the effects of changes in experimental conditions using well-characterized NPs. The results of the studies have been used to generate recommendations for a suitable and robust testing strategy which can be applied to new medical NPs as they are developed.


Subject(s)
Nanomedicine/methods , Nanoparticles/toxicity , Toxicity Tests/methods , Humans , In Vitro Techniques/standards , Toxicity Tests/standards
2.
Talanta ; 130: 442-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25159433

ABSTRACT

Metabolomics based on liquid chromatography-mass spectrometry (LC-MS) is a powerful tool for studying dynamic responses of biological systems to different physiological or pathological conditions. Differences in the instrumental response within and between batches introduce unwanted and uncontrolled data variation that should be removed to extract useful information. This work exploits a recently developed method for the identification of batch effects in high throughput genomic data based on the calculation of a δ statistic through principal component analysis (PCA) and guided PCA. Its applicability to LC-MS metabolomic data was tested on two real examples. The first example involved the repeated analysis of 42 plasma samples and 6 blanks in three independent batches, and the second data set involved the analysis of 101 plasma and 18 blank samples in a single batch with a total runtime of 50h. The first and second data set were used to evaluate between and within-batch effects using the δ statistic, respectively. Results obtained showed the usefulness of using the δ statistic together with other approaches such as summary statistics of peak intensity distributions, PCA scores plots or the monitoring of IS peak intensities, to detect and identify instrumental instabilities in LC-MS.


Subject(s)
Chromatography, Liquid/methods , Mass Spectrometry/methods , Metabolomics , Plasma/chemistry , Principal Component Analysis/methods , Calibration , Humans , Quality Control
3.
Toxicol In Vitro ; 27(2): 954-63, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23357416

ABSTRACT

No consensus exists on how to address possible toxicity of nanomaterials as they interfere with most in vitro screening tests based on colorimetric and fluorimetric probes such as the dichloro-dihydro-fluorescein diacetate (DCFH-DA) assay for detection of oxidative species. In the present research, nanomaterial interaction with DCFH-DA was studied in relation to its nature and/or assay conditions (cell-based and time exposure) by incubating Rhodamine (Rhd)-labeled 25nm and 50nm silica (SiO2), naked and oleic acid coated magnetite, (Fe3O4) and maghemite (Fe2O3) iron oxide, titanium dioxide (TiO2) and poly(ethylene oxide)-poly(lactide/glycolide) acid (PLGA-PEO) nanoparticles (NPs) with metabolically active rat hepatocytes for 4 and 24-h periods. Data indicated that nanoparticle uptake correlated with quenching of dye fluorescence emission. In spite of their masking effect, the oxidative potential of NPs could be detected at a limited threshold concentration when exposed for periods of time longer than those frequently used for this test. However, changes in the experimental conditions did not systematically result in free radical formation for all nanomaterials tested. Overall data indicate that despite the quenching effect of nanoparticles on DCFH-DA assay, it can be considered as a useful tool for quantitative measurement of NPs-induced oxidative stress by minor modifications of standardized protocols.


Subject(s)
Biological Assay/methods , Fluoresceins , Fluorescent Dyes , Nanoparticles/toxicity , Oxidative Stress/drug effects , Animals , Cell Survival/drug effects , Cells, Cultured , Ferric Compounds/toxicity , Ferrosoferric Oxide/toxicity , Hepatocytes , Lactic Acid/toxicity , Male , Polyethylene Glycols/toxicity , Polyglycolic Acid/toxicity , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Silicon Dioxide/toxicity , Titanium/toxicity
4.
Toxicol In Vitro ; 27(4): 1357-76, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22922246

ABSTRACT

ACuteTox is a project within the 6th European Framework Programme which had as one of its goals to develop, optimise and prevalidate a non-animal testing strategy for predicting human acute oral toxicity. In its last 6 months, a challenging exercise was conducted to assess the predictive capacity of the developed testing strategies and final identification of the most promising ones. Thirty-two chemicals were tested blind in the battery of in vitro and in silico methods selected during the first phase of the project. This paper describes the classification approaches studied: single step procedures and two step tiered testing strategies. In summary, four in vitro testing strategies were proposed as best performing in terms of predictive capacity with respect to the European acute oral toxicity classification. In addition, a heuristic testing strategy is suggested that combines the prediction results gained from the neutral red uptake assay performed in 3T3 cells, with information on neurotoxicity alerts identified by the primary rat brain aggregates test method. Octanol-water partition coefficients and in silico prediction of intestinal absorption and blood-brain barrier passage are also considered. This approach allows to reduce the number of chemicals wrongly predicted as not classified (LD50>2000 mg/kg b.w.).


Subject(s)
Neural Networks, Computer , Toxicity Tests, Acute , Administration, Oral , Animal Testing Alternatives , Animals , Blood-Brain Barrier/metabolism , Cell Line , Cell Survival , Colony-Forming Units Assay , Computer Simulation , Cytokines/metabolism , Humans , Intestinal Absorption , Lethal Dose 50 , Mice , Oxidative Stress , Rats , Risk Assessment
5.
Toxicol In Vitro ; 25(3): 589-604, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21167275

ABSTRACT

There are now numerous in vitro and in silico ADME alternatives to in vivo assays but how do different industries incorporate them into their decision tree approaches for risk assessment, bearing in mind that the chemicals tested are intended for widely varying purposes? The extent of the use of animal tests is mainly driven by regulations or by the lack of a suitable in vitro model. Therefore, what considerations are needed for alternative models and how can they be improved so that they can be used as part of the risk assessment process? To address these issues, the European Partnership for Alternative Approaches to Animal Testing (EPAA) working group on prioritization, promotion and implementation of the 3Rs research held a workshop in November, 2008 in Duesseldorf, Germany. Participants included different industry sectors such as pharmaceuticals, cosmetics, industrial- and agro-chemicals. This report describes the outcome of the discussions and recommendations (a) to reduce the number of animals used for determining the ADME properties of chemicals and (b) for considerations and actions regarding in vitro and in silico assays. These included: standardisation and promotion of in vitro assays so that they may become accepted by regulators; increased availability of industry in vivo kinetic data for a central database to increase the power of in silico predictions; expansion of the applicability domains of in vitro and in silico tools (which are not necessarily more applicable or even exclusive to one particular sector) and continued collaborations between regulators, academia and industry. A recommended immediate course of action was to establish an expert panel of users, developers and regulators to define the testing scope of models for different chemical classes. It was agreed by all participants that improvement and harmonization of alternative approaches is needed for all sectors and this will most effectively be achieved by stakeholders from different sectors sharing data.


Subject(s)
Animal Testing Alternatives , Congresses as Topic , Xenobiotics , Animals , Cells, Cultured , Computer Simulation , Europe , Industry , International Cooperation , Models, Chemical , Quantitative Structure-Activity Relationship , Xenobiotics/chemistry , Xenobiotics/pharmacokinetics , Xenobiotics/toxicity
6.
Curr Pharm Des ; 16(17): 1963-77, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20236064

ABSTRACT

The liver is the most important target for toxicity caused by drugs. This vulnerability is a consequence of the functional features of the liver and their role in the metabolic elimination of most drugs. Therefore, evaluation of potential hepatotoxicity represents a critical step in the development of new drugs. The liver is very active in metabolising foreign compounds and, although biotransformation reactions generally parallel detoxification processes, the formation of reactive metabolites is relatively frequent. Thus, drug-induced hepatotoxicity can be due to the administered compound itself or to metabolites formed by hepatic metabolism. The most important systems to study hepatotoxicity and metabolic activity in vitro are liver slices, isolated liver cells in suspensions or in primary cultures including co-culture methods and special 3D techniques, various subcellular fractions and hepatic cell lines. These models can be used for cytotoxicity and genotoxicity screening, and also to identify the mechanisms involved in drug-induced hepatotoxicity. Assessment of current cytotoxicity and hepatic-specific biochemical effects are limited by the inability to measure a wide spectrum of potential mechanistic changes involved in the drug-induced toxic injury. A convenient selection of end-points allows a multiparametric evaluation of drug toxicity. In this regard, omic (cytomic, metabonomic, proteomic and toxicogemic) approaches help defining patterns of hepatotoxicity for early identification of potential adverse effects of the drug to the liver. The development of robust in vitro-based multiparametric screening assays covering a wider spectrum of key effects will heighten the predictive capacity for human hepatotoxicity, and accelerate the drug development process.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Drug Design , Drug-Related Side Effects and Adverse Reactions , Animals , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Mutagenicity Tests , Pharmaceutical Preparations/metabolism , Tissue Culture Techniques , Toxicity Tests/methods
8.
Mini Rev Med Chem ; 6(9): 1053-62, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17018004

ABSTRACT

The study of the ADME features of the huge number of new chemical entities (NCEs) produced mainly by combinatorial chemistry has become a bottleneck in the drug development process. In response the pharmaceutical industry is involved in the development of new medium/high-throughput screening capabilities. The aim of this paper is to review some of the available in vitro ADME systems adapted to screening requirements together with the technological approaches which can be linked to medium/high-throughput molecular screening.


Subject(s)
Combinatorial Chemistry Techniques , Drug Evaluation, Preclinical/methods , Pharmacokinetics , Animals , Cells, Cultured , Humans
9.
Toxicol In Vitro ; 20(4): 464-71, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16263237

ABSTRACT

The extract of the fern Polypodium leucotomos (PL, Fernblock) is an oral photoprotectant with strong antioxidative properties. Recent studies to determine its chemical composition have shown 4-hydroxycinnamic acid (p-coumaric), 3 methoxy-4-hydroxycinnamic acid (ferulic), 3,4-dihydroxycinnamic acid (caffeic), 3-methoxy-4-hydroxybenzoic acid (vanillic) and 3-caffeoilquinic acid (chlorogenic) to be among its major phenolic components. No conclusive data are available, however, on the H2O2-scavenging capacity of these compounds, or on their absorption and metabolism following their oral intake. In the present work, their antioxidative capacity was assessed by the luminol/H2O2 assay, their absorption studied using Caco-2 cells to resemble the intestinal barrier, and their metabolism investigated using cultured primary rat hepatocytes. The antioxidant capacity of PL components increased in a concentration-dependent manner, with ferulic and caffeic acids the most powerful antioxidants. The apparent permeability results correspond to a human post-oral administration absorption of 70-100% for all tested substances. Coumaric, ferulic and vanillic acids were metabolized by CYP450-dependent mono-oxygenases and partially conjugated to glucuronic acid and sulfate. These phenolic compounds may contribute to the health benefits afforded by this oral photoprotectant.


Subject(s)
Antioxidants/pharmacokinetics , Hepatocytes/metabolism , Hydroxybenzoates/pharmacokinetics , Polypodium/chemistry , Animals , Antioxidants/chemistry , Caco-2 Cells , Cell Survival/drug effects , Dose-Response Relationship, Drug , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Hydroxybenzoates/analysis , Plant Extracts/chemistry , Plant Extracts/pharmacokinetics , Rats , Rats, Sprague-Dawley
10.
J Biol Chem ; 276(30): 28233-41, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11359777

ABSTRACT

Serine proteases are implicated in a variety of processes during neurogenesis, including cell migration, axon outgrowth, and synapse elimination. Tissue-type plasminogen activator and urokinase-type activator are expressed in the floor plate during embryonic development. F-spondin, a gene also expressed in the floor plate, encodes a secreted, extracellular matrix-attached protein that promotes outgrowth of commissural axons and inhibits outgrowth of motor axons. F-spondin is processed in vivo to yield an amino half protein that contains regions of homology to reelin and mindin, and a carboxyl half protein that contains either six or four thrombospondin type I repeats (TSRs). We have tested F-spondin to see whether it is subjected to processing by plasmin and to determine whether the processing modulates its biological activity. Plasmin cleaves F-spondin at its carboxyl terminus. By using nested deletion proteins and mutating potential plasmin cleavage sites, we have identified two cleavage sites, the first between the fifth and sixth TSRs, and the second at the fifth TSR. Analysis of the extracellular matrix (ECM) attachment properties of the TSRs revealed that the fifth and sixth TSRs bind to the ECM, but repeats 1-4 do not. Structural functional experiments revealed that two basic motives are required to elicit binding of TSR module to the ECM. We demonstrate further that plasmin releases the ECM-bound F-spondin protein.


Subject(s)
Extracellular Matrix/metabolism , Fibrinolysin/chemistry , Fibrinolysin/physiology , Growth Substances , Neural Cell Adhesion Molecules/chemistry , Neural Cell Adhesion Molecules/metabolism , Peptides , Amino Acid Sequence , Binding Sites , Cell Division , Cell Line , Cell Movement , DNA/metabolism , Dose-Response Relationship, Drug , Extracellular Matrix Proteins , Fibrinolysin/metabolism , Gene Deletion , Humans , Molecular Sequence Data , Mutation , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Reelin Protein , Sequence Homology, Amino Acid , Structure-Activity Relationship , Tissue Plasminogen Activator/metabolism , Transfection
11.
Neurosci Lett ; 299(1-2): 77-80, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11166942

ABSTRACT

Methylazoximethanol (MAM) acetate-induced cell death in the external granule cell layer of the developing cerebellum affects clusters of cells with morphological features of apoptosis. This is accompanied by selective induction of active caspase-3 expression and increased c-Jun/AP-1 (N) immunoreactivity in dying cells, as revealed with immunohistochemistry. Since the antibody to cJun/AP-1 (N) cross-reacts with epitopes emerging after caspase-mediated proteolysis during apoptosis, these results indicate that MAM-induced cell death is associated with active caspase-3 expression and function in dying cells. In order to investigate the involvement of tissue-type plasminogen activator (tPA), which has been implicated in certain forms of neuronal cell death, MAM-induced cell death has been examined in tPA-/- and tPA+/+ mice. No differences in the number of dying cells, as seen with haematoxylin and eosin staining and in situ end-labelling of fragmented nuclear DNA-processed sections, were seen between tPA-/- and tPA+/+ mice. These results indicate that tPA is not involved in MAM-induced cell death in the developing brain.


Subject(s)
Apoptosis/drug effects , Caspases/drug effects , Cerebellar Cortex/drug effects , Methylazoxymethanol Acetate/pharmacology , Neurons/drug effects , Nucleic Acid Synthesis Inhibitors/pharmacology , Tissue Plasminogen Activator/drug effects , Animals , Animals, Newborn , Apoptosis/physiology , Caspase 3 , Caspases/metabolism , Cerebellar Cortex/growth & development , Cerebellar Cortex/metabolism , Fas Ligand Protein , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism , fas Receptor/drug effects , fas Receptor/metabolism
12.
J Biol Chem ; 272(41): 26005-8, 1997 Oct 10.
Article in English | MEDLINE | ID: mdl-9325336

ABSTRACT

Botulinum neurotoxin type A (BoNT/A) inhibits neurotransmitter release by specific cleavage of SNAP-25, a synaptosome-associated protein also expressed in the ACTH secretory cell line AtT-20. Expression of light chain BoNT/A (L-BoNT/A) gene transfected into AtT-20 cells resulted in a cleaved form of SNAP-25 indistinguishable from that generated by bona fide BoNT/A. L-BoNT/A-transfected cells showed no difference in replication rate, viability, or phenotype, compared with control AtT-20 cells. In contrast, L-BoNT/A-transfected cells could not be induced to secrete ACTH upon stimulation by 8-bromo-cAMP or KCl. In addition, alpha-latrotoxin induced ACTH release from control cells, but not from L-BoNT/A-transfected cells. These experiments suggest an important role for SNAP-25 in regulated secretion from AtT-20 cells and underline the usefulness of this cell system as a tool for the study of the molecular mechanism of peptide hormone secretion.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Botulinum Toxins, Type A/genetics , Membrane Proteins , Nerve Tissue Proteins/physiology , Pituitary Gland, Anterior/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Botulinum Toxins, Type A/metabolism , Cell Survival , Male , Nerve Tissue Proteins/metabolism , Pituitary Gland, Anterior/cytology , Rats , Rats, Sprague-Dawley , Synaptosomal-Associated Protein 25 , Transfection , Tumor Cells, Cultured
13.
FEBS Lett ; 393(2-3): 216-20, 1996 Sep 16.
Article in English | MEDLINE | ID: mdl-8814293

ABSTRACT

Because recent information suggests that the localized deposition of protease inhibitors is one mechanism by which cells regulate pericellular proteolysis during tissue invasion, the distribution of type 1 plasminogen activator inhibitor (PA1-1) associated with the invasive human glioma cell line U-251 was investigated. Direct and reverse fibrin zymography indicated the presence of urokinase-like plasminogen activator (u-PA) and PAI-1 in U-251 conditioned media and cell lysates. PA1-1 antigen was detected immunologically in cytoplasmic granules present within cellular processes of U-251 cells and these organelles could be isolated on Percoll density gradients in a high density band. In contrast, u-PA activity and another secreted protein, amyloid beta-protein precursor, were only present in the low density region of the gradients. Functional analysis of PAI-1 in the granules contained within the high density fractions revealed the presence of active PAI-1. Incubation of U-251 cells with the secretagogue, 8-bromoadenosine 3':5'-cyclic monophosphate, resulted in a 3-fold increase in the release of PAI-1 in the media conditioned by these cells. These data suggest that the human glioma cell line U-251 contains PAI-1 in a rapidly releasable form, which may provide another mechanism by which these tumors could regulate proteolytic activity in a localized manner.


Subject(s)
Fibrinolysis , Plasminogen Activator Inhibitor 1/metabolism , Urokinase-Type Plasminogen Activator/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Cell Fractionation , Cell Line , Centrifugation, Density Gradient , Fibrin , Glioma , Humans , Immunohistochemistry , Neoplasm Invasiveness , Organelles/drug effects , Organelles/metabolism , Organelles/ultrastructure , Plasminogen Activator Inhibitor 1/isolation & purification , Povidone , Silicon Dioxide , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/isolation & purification
14.
J Biol Chem ; 269(5): 3875-80, 1994 Feb 04.
Article in English | MEDLINE | ID: mdl-8106433

ABSTRACT

To understand the processing of type 1 plasminogen activator inhibitor (PAI-1) into the storage granules of platelets, we utilized a eukaryotic expression vector (pRC/CMV) to transfer the human cDNA for PAI-1 into AtT-20 cells, a mouse pituitary cell line known to sort proteins in a regulated fashion. Immunofluorescence staining of PAI-1-transfected AtT-20 clones revealed co-localization of PAI-1 with an endogenously produced and stored hormone (i.e. adrenocorticotropic hormone, ACTH). Stimulation of PAI-1-transfected AtT-20 cells with a secretagogue resulted in the release of both active PAI-1 and the latent form. In comparison, PAI-1-transfected Chinese hamster ovary cells (i.e. a nonpackaging cell line) did not release PAI-1 in response to a secretagogue and exhibited immunoreactivity for PAI-1 primarily confined to the Golgi region. Percoll density gradient fractionation of AtT-20 cells revealed a codistribution of PAI-1 and ACTH in cellular compartments of the same density. The half-life of PAI-1 activity at 37 degrees C was prolonged in intact granules (t1/2, 5 h) in comparison with its half-life in lysed granules (t1/2, 2 h). These studies demonstrate the presence of a new functional property associated with the PAI-1 molecule that directs this inhibitor into the storage secretory pathway.


Subject(s)
Blood Platelets/metabolism , Cytoplasmic Granules/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Adrenocorticotropic Hormone/analysis , Adrenocorticotropic Hormone/biosynthesis , Animals , Cell Line , DNA, Complementary/metabolism , Fluorescent Antibody Technique , Humans , Kinetics , Laminin/analysis , Mice , Pituitary Neoplasms , Plasminogen Activator Inhibitor 1/analysis , Plasminogen Activator Inhibitor 1/metabolism , Protein Processing, Post-Translational , Time Factors , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...