Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Toxicol Sci ; 178(1): 71-87, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32866265

ABSTRACT

Seizures are life-threatening adverse drug reactions which are investigated late in drug development using rodent models. Consequently, if seizures are detected, a lot of time, money and animals have been used. Thus, there is a need for in vitro screening models using human cells to circumvent interspecies translation. We assessed the suitability of cocultures of human-induced pluripotent stem cell (hiPSC)-derived neurons and astrocytes compared with rodent primary cortical cultures for in vitro seizure liability assessment using microelectrode arrays. hiPSC-derived and rodent primary cortical neuronal cocultures were exposed to 9 known (non)seizurogenic compounds (pentylenetetrazole, amoxapine, enoxacin, amoxicillin, linopirdine, pilocarpine, chlorpromazine, phenytoin, and acetaminophen) to assess effects on neuronal network activity using microelectrode array recordings. All compounds affect activity in hiPSC-derived cocultures. In rodent primary cultures all compounds, except amoxicillin changed activity. Changes in activity patterns for both cell models differ for different classes of compounds. Both models had a comparable sensitivity for exposure to amoxapine (lowest observed effect concentration [LOEC] 0.03 µM), linopirdine (LOEC 1 µM), and pilocarpine (LOEC 0.3 µM). However, hiPSC-derived cultures were about 3 times more sensitive for exposure to pentylenetetrazole (LOEC 30 µM) than rodent primary cortical cultures (LOEC 100 µM). Sensitivity of hiPSC-derived cultures for chlorpromazine, phenytoin, and enoxacin was 10-30 times higher (LOECs 0.1, 0.3, and 0.1 µM, respectively) than in rodent cultures (LOECs 10, 3, and 3 µM, respectively). Our data indicate that hiPSC-derived neuronal cocultures may outperform rodent primary cortical cultures with respect to detecting seizures, thereby paving the way towards animal-free seizure assessment.


Subject(s)
Induced Pluripotent Stem Cells/drug effects , Neurons/drug effects , Seizures/diagnosis , Animals , Cells, Cultured , Coculture Techniques , Humans , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Rodentia , Seizures/chemically induced
3.
ALTEX ; 37(1): 121-135, 2020.
Article in English | MEDLINE | ID: mdl-31686111

ABSTRACT

A sizeable proportion of drug attrition is due to drug-induced seizures. Current available animal models frequently fail to predict human seizure liability. Therefore, there is a need for in vitro alternatives, preferably based on human-derived neurons to circumvent interspecies translation. The increasing number of commercially available human induced pluripotent stem cell (hiPSC)-derived neuronal models holds great promise for replacing rodent primary cultures. We therefore tested three different hiPSC-derived neuronal models for their applicability for in vitro seizure liability assessment. Using immunofluorescent staining and multi-well micro-electrode arrays we show that all models develop functional neuronal networks that exhibit spontaneous activity and (network) bursting behavior. Developmental patterns differ between the models, probably due to differences in model composition and seeding density. Nevertheless, neuronal activity and (network) bursting can be reproducibly modulated with the seizurogenic compounds strychnine, picrotoxin (PTX) and 4-aminopyridine (4-AP). However, the sensitivity and degree of chemical-induced effects differs between the models, which can likely be explained by differences in seeding density, maturation and different ratios of inhibitory and excitatory cell types. Importantly, compared to rat primary cortical neurons, the hiPSC-derived neuronal models were equally, or even better in the case of 4-AP, suited to detect seizurogenicity. Overall, our data indicate that hiPSC-derived neuronal models may in the future be used as a first screening tool for in vitro seizure liability assessment. However, before hiPSC-derived neuronal models can fully replace animal experiments, more compounds should be tested and the available models must be further characterized to fully understand their applicability.


Subject(s)
Animal Use Alternatives , Induced Pluripotent Stem Cells/drug effects , Neurons/drug effects , Seizures/chemically induced , Animals , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/physiology , Neurons/physiology , Rats
4.
Neurotoxicology ; 67: 215-225, 2018 07.
Article in English | MEDLINE | ID: mdl-29909083

ABSTRACT

Neurotoxicity testing still relies on ethically debated, expensive and time consuming in vivo experiments, which are unsuitable for high-throughput toxicity screening. There is thus a clear need for a rapid in vitro screening strategy that is preferably based on human-derived neurons to circumvent interspecies translation. Recent availability of commercially obtainable human induced pluripotent stem cell (hiPSC)-derived neurons and astrocytes holds great promise in assisting the transition from the current standard of rat primary cortical cultures to an animal-free alternative. We therefore composed several hiPSC-derived neuronal models with different ratios of excitatory and inhibitory neurons in the presence or absence of astrocytes. Using immunofluorescent stainings and multi-well micro-electrode array (mwMEA) recordings we demonstrate that these models form functional neuronal networks that become spontaneously active. The differences in development of spontaneous neuronal activity and bursting behavior as well as spiking patterns between our models confirm the importance of the presence of astrocytes. Preliminary neurotoxicity assessment demonstrates that these cultures can be modulated with known seizurogenic compounds, such as picrotoxin (PTX) and endosulfan, and the neurotoxicant methylmercury (MeHg). However, the chemical-induced effects on different parameters for neuronal activity, such as mean spike rate (MSR) and mean burst rate (MBR), may depend on the ratio of inhibitory and excitatory neurons. Our results thus indicate that hiPSC-derived neuronal models must be carefully designed and characterized prior to large-scale use in neurotoxicity screening.


Subject(s)
Action Potentials/drug effects , Astrocytes/drug effects , Induced Pluripotent Stem Cells/physiology , Neurons/drug effects , Action Potentials/physiology , Astrocytes/physiology , Cells, Cultured , Coculture Techniques/methods , Endosulfan/toxicity , Humans , Induced Pluripotent Stem Cells/drug effects , Methylmercury Compounds/toxicity , Neurons/physiology , Picrotoxin/toxicity
5.
Toxicol In Vitro ; 45(Pt 1): 60-71, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28506818

ABSTRACT

The prevalence and use of new psychoactive substances (NPS) is increasing and currently over 600 NPS exist. Many illicit drugs and NPS increase brain monoamine levels by inhibition and/or reversal of monoamine reuptake transporters (DAT, NET and SERT). This is often investigated using labor-intensive, radiometric endpoint measurements. We investigated the applicability of a novel and innovative assay that is based on a fluorescent monoamine mimicking substrate. DAT, NET or SERT-expressing human embryonic kidney (HEK293) cells were exposed to common drugs (cocaine, dl-amphetamine or MDMA), NPS (4-fluoroamphetamine, PMMA, α-PVP, 5-APB, 2C-B, 25B-NBOMe, 25I-NBOMe or methoxetamine) or the antidepressant fluoxetine. We demonstrate that this fluorescent microplate reader-based assay detects inhibition of different transporters by various drugs and discriminates between drugs. Most IC50 values were in line with previous results from radiometric assays and within estimated human brain concentrations. However, phenethylamines showed higher IC50 values on hSERT, possibly due to experimental differences. Compared to radiometric assays, this high-throughput fluorescent assay is uncomplicated, can measure at physiological conditions, requires no specific facilities and allows for kinetic measurements, enabling detection of transient effects. This assay is therefore a good alternative for radiometric assays to investigate effects of illicit drugs and NPS on monoamine reuptake transporters.


Subject(s)
Amphetamines/pharmacology , Neurotransmitter Transport Proteins/antagonists & inhibitors , Psychotropic Drugs/pharmacology , Cocaine/pharmacology , HEK293 Cells , Humans , Molecular Structure , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Psychotropic Drugs/chemistry
6.
Neuropharmacology ; 123: 1-9, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28454981

ABSTRACT

The use of new psychoactive substances (NPS) is steadily increasing. One commonly used NPS is methoxetamine (MXE), a ketamine analogue. Several adverse effects have been reported following MXE exposure, while only limited data are available on its neuropharmacological modes of action. We investigated the effects of MXE and ketamine on several endpoints using multiple in vitro models. These included rat primary cortical cells, human SH-SY5Y cells, human induced pluripotent stem cell (hiPSC)-derived iCell® Neurons, DopaNeurons and astrocyte co-cultures, and human embryonic kidney (HEK293) cells. We investigated effects on several neurotransmitter receptors using single cell intracellular calcium [Ca2+]i imaging, effects on neuronal activity using micro-electrode array (MEA) recordings and effects on human monoamine transporters using a fluorescence-based plate reader assay. In rat primary cortical cells, 10 µM MXE increased the glutamate-evoked increase in [Ca2+]i, whereas 10 µM ketamine was without effect. MXE and ketamine did not affect voltage-gated calcium channels (VGCCs), but inhibited spontaneous neuronal activity (IC50 0.5 µM and 1.2 µM respectively). In human SH-SY5Y cells, 10 µM MXE slightly inhibited the K+- and acetylcholine-evoked increase in [Ca2+]i. In hiPSC-derived iCell®(Dopa)Neurons, only the ATP-evoked increase in [Ca2+]i was slightly reduced. Additionally, MXE inhibited spontaneous neuronal activity (IC50 between 10 and 100 µM). Finally, MXE potently inhibits uptake via monoamine transporters (DAT, NET and SERT), with IC50 values in the low micromolar range (33, 20, 2 µM respectively). Our combined in vitro data provide an urgently needed first insight into the multiple modes of action of MXE. The use of different models and different (neuronal) endpoints can be complementary in pharmacological profiling. Rapid in vitro screening methods as those presented here, could be of utmost importance for gaining a first mechanistic insight to aid the risk assessment of emerging NPS.


Subject(s)
Cyclohexanones/pharmacology , Cyclohexylamines/pharmacology , Neurons/drug effects , Psychotropic Drugs/pharmacology , Animals , Calcium/metabolism , Calcium Channels/metabolism , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Cations, Divalent/metabolism , Cell Line, Tumor , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Coculture Techniques , Glutamic Acid/metabolism , Glycerol Kinase , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/physiology , Ketamine/analogs & derivatives , Ketamine/pharmacology , Neurons/physiology , Rats, Wistar , Vesicular Monoamine Transport Proteins/metabolism
7.
Neurotoxicology ; 57: 194-202, 2016 12.
Article in English | MEDLINE | ID: mdl-27720795

ABSTRACT

There is an increasing demand for in vitro test systems to detect neurotoxicity for use in chemical risk assessment. In this study, we evaluated the applicability of rat primary cortical cultures grown on multi-well micro-electrode arrays (mwMEAs) to detect effects of chronic 14-day exposure to structurally different insecticides or methylmercury on neuronal activity (mean spike rate; MSR). Effects of chronic exposure to α-cypermethrin, endosulfan, carbaryl, chlorpyrifos(-oxon), methylmercury or solvent control [14days exposure, initiated after baseline recording at day in vitro (DIV)7] were studied in five successive recordings between DIV10 and DIV21. The results were compared to effects of acute exposure to these same compounds (activity recorded immediately after the start of exposure after baseline recording at DIV10-11). Chronic 14-day exposure to methylmercury, chlorpyrifos and α-cypermethrin inhibited MSR, all with a lowest-observed effect concentration (LOEC) of 0.1µM, while exposure to endosulfan increased MSR [LOEC: 1µM]. No significant effects were observed for chlorpyrifos-oxon and carbaryl. Similar to the observations in the chronic 14-day exposure studies, MSR was inhibited by acute 30-min exposure to methylmercury, chlorpyrifos, and α-cypermethrin [LOECs: 1µM, 10µM, and 1µM, respectively], whereas endosulfan increased MSR [LOEC: 0.3µM]. While not observed in the chronic 14-day exposure study, acute exposure to chlorpyrifos-oxon and carbaryl resulted in inhibition of MSR [LOECs: 10µM, and100 µM, respectively]. Effects on median interspike intervals (mISI; a measure for neuronal firing pattern) were not detected following chronic 14-day or acute 30-min exposure, except for increased mISI at acute chlorpyrifos and α-cypermethrin exposures at concentrations that also inhibited MSR. These data indicate that the effects of chronic 14-day exposures to methylmercury and insecticides at low concentrations on spontaneous neuronal activity in vitro can be predicted in rapid acute screening studies using mwMEAs.


Subject(s)
Cerebral Cortex/cytology , Neurons/drug effects , Action Potentials/drug effects , Animals , Animals, Newborn , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Insecticides/pharmacology , Methylmercury Compounds/pharmacology , Rats , Rats, Wistar , Time Factors
8.
ALTEX ; 33(3): 261-71, 2016.
Article in English | MEDLINE | ID: mdl-27010910

ABSTRACT

Current neurotoxicity testing heavily relies on expensive, time consuming and ethically debated in vivo animal experiments that are unsuitable for screening large number of chemicals. Consequently, there is a clear need for (high-throughput) in vitro test strategies, preferably using human cells as this increases relevance and eliminates the need for interspecies translation. However, human stem cell-derived neurons used to date are not well characterised, require prolonged differentiation and are potentially subject to batch-to-batch variation, ethical concerns and country-specific legislations. Recently, a number of human induced pluripotent stem cell (iPSC)-derived neurons became commercially available that may circumvent these concerns. We therefore used immunofluorescent stainings to demonstrate that human iPSC-derived neurons from various suppliers form mixed neuronal cultures, consisting of different types of (excitatory and inhibitory) neurons. Using multi-well microelectrode array (mwMEA) recordings, we demonstrate that these human iPSC-derived cultures develop spontaneous neuronal activity over time, which can be modulated by different physiological, toxicological and pharmacological compounds. Additional single cell calcium imaging illustrates the presence of functional GABA, glutamate, and acetylcholine receptors as well as voltage-gated calcium channels. While human iPSC-derived neuronal cultures appear not yet suitable to fully replace the rat primary cortical model, our data indicate that these rapidly differentiating, commercially available human iPSC-derived neuronal cultures are already suitable for in vitro prioritisation and effect screening studies. Further characterisation and toxicological validation is now required to facilitate acceptance and large-scale implementation of these animal-free, physiologically-relevant human iPSC-based modelsfor future neurotoxicity testing.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Neurons/physiology , Toxicity Tests/methods , Animals , Cell Differentiation , Cells, Cultured , Coculture Techniques , Electrophysiological Phenomena , Fluoroimmunoassay , Gene Expression Regulation/physiology , Humans , Induced Pluripotent Stem Cells/cytology , Ion Channels/physiology , Neurons/cytology , Rats , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , Staining and Labeling
9.
Toxicol Sci ; 147(2): 607-17, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26187449

ABSTRACT

We previously demonstrated that acute inhibition of voltage-gated calcium channels (VGCCs) is a common mode of action for (sub)micromolar concentrations of chemicals, including insecticides. However, because human exposure to chemicals is usually chronic and repeated, we investigated if selected insecticides from different chemical classes (organochlorines, organophosphates, pyrethroids, carbamates, and neonicotinoids) also disturb calcium homeostasis after subchronic (24 h) exposure and after a subsequent (repeated) acute exposure. Effects on calcium homeostasis were investigated with single-cell fluorescence (Fura-2) imaging of PC12 cells. Cells were depolarized with high-K(+) saline to study effects of subchronic or repeated exposure on VGCC-mediated Ca(2+) influx. The results demonstrate that except for carbaryl and imidacloprid, all selected insecticides inhibited depolarization (K(+))-evoked Ca(2+) influx after subchronic exposure (IC50's: approximately 1-10 µM) in PC12 cells. These inhibitory effects were not or only slowly reversible. Moreover, repeated exposure augmented the inhibition of the K(+)-evoked increase in intracellular calcium concentration induced by subchronic exposure to cypermethrin, chlorpyrifos, chlorpyrifos-oxon, and endosulfan (IC50's: approximately 0.1-4 µM). In rat primary cortical cultures, acute and repeated chlorpyrifos exposure also augmented inhibition of VGCCs compared with subchronic exposure. In conclusion, compared with subchronic exposure, repeated exposure increases the potency of insecticides to inhibit VGCCs. However, the potency of insecticides to inhibit VGCCs upon repeated exposure was comparable with the inhibition previously observed following acute exposure, with the exception of chlorpyrifos. The data suggest that an acute exposure paradigm is sufficient for screening chemicals for effects on VGCCs and that PC12 cells are a sensitive model for detection of effects on VGCCs.


Subject(s)
Calcium Channel Blockers/toxicity , Insecticides/toxicity , Action Potentials/drug effects , Animals , Brain/cytology , Brain/drug effects , Calcium/metabolism , Calcium Channel Blockers/administration & dosage , Calcium Channels/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Insecticides/administration & dosage , PC12 Cells/drug effects , Rats , Rats, Wistar , Toxicity Tests, Subchronic/methods
10.
Toxicol Rep ; 1: 360-372, 2014.
Article in English | MEDLINE | ID: mdl-28962252

ABSTRACT

Phytoestrogens are plant-derived estrogen-like compounds that are increasingly used for their suggested health promoting properties, even by healthy, young women. However, scientific concerns exist regarding potential adverse effects on female reproduction. In this study, naringenin (NAR), 8-prenylnaringenin (8-PN), genistein (GEN), coumestrol (COU), quercetin (QUE) and resveratrol (RSV) up-regulated steroidogenic acute regulatory protein (StaR) mRNA levels in KGN human granulosa-like tumor cells. Most of the phytoestrogens tested also increased CYP19A1 (aromatase) mRNA levels via activation of ovary-specific I.3 and II promoters. Yet, only NAR (3 and 10 µM), COU (10 and 30 µM) and QUE (10 µM) also statistically significantly induced aromatase activity in KGN cells after 24 h. 8-PN, aromatase inhibitor letrozole and estrogen receptor antagonist ICI 182,780 concentration-dependently inhibited aromatase activity with IC50 values of 8 nM, 10 nM and 72 nM, respectively. Co-exposure with ICI 182,780 (0.1 µM) statistically significantly attenuated the induction of aromatase activity by QUE and COU, but not NAR. Cell cycle status and proliferation of KGN cells were not affected by any of the phytoestrogens tested. Nonetheless, the migration of KGN cells was significantly reduced with approximately 30% by COU, RSV and QUE and 46% by GEN at 10 µM, but not NAR and 8-PN. Our results indicate that phytoestrogens can affect various pathways in granulosa-like cells in vitro at concentrations that can be found in plasma upon supplement intake. This implies that phytoestrogens may interfere with ovarian function and caution is in place regarding the use of supplements with high contents of phytoestrogens.

11.
PLoS One ; 8(4): e61555, 2013.
Article in English | MEDLINE | ID: mdl-23613873

ABSTRACT

PURPOSE: The combination of systemic cisplatin with local and regional radiotherapy as primary treatment of head and neck squamous cell carcinoma (HNSCC) leads to cure in approximately half of the patients. The addition of cisplatin has significant effects on outcome, but despite extensive research the mechanism underlying cisplatin response is still not well understood. METHODS: We examined 19 HNSCC cell lines with variable cisplatin sensitivity. We determined the TP53 mutational status of each cell line and investigated the expression levels of 11 potentially relevant genes by quantitative real-time PCR. In addition, we measured cisplatin accumulation and retention, as well as the level of platinum-DNA adducts. RESULTS: We found that the IC50 value was significantly correlated with the platinum-DNA adduct levels that accumulated during four hours of cisplatin incubation (p = 0.002). We could not find a significant correlation between cisplatin sensitivity and any of the other parameters tested, including the expression levels of established cisplatin influx and efflux transporters. Furthermore, adduct accumulation did not correlate with mRNA expression of the investigated influx pumps (CTR1 and OCT3) nor with that of the examined DNA repair genes (ATR, ATM, BRCA1, BRCA2 and ERCC1). CONCLUSION: Our findings suggest that the cisplatin-DNA adduct level is the most important determinant of cisplatin sensitivity in HNSCC cells. Imaging with radio-labeled cisplatin might have major associations with outcome.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Cisplatin/therapeutic use , DNA, Neoplasm/metabolism , Head and Neck Neoplasms/drug therapy , Platinum/therapeutic use , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/metabolism , Cisplatin/pharmacology , DNA Adducts/metabolism , DNA Adducts/pharmacology , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Genes, Neoplasm/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Inhibitory Concentration 50 , Mutation/genetics , Platinum/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Squamous Cell Carcinoma of Head and Neck , Statistics, Nonparametric , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...