1
|
Campàs M, Leonardo S, Sanchez-Henao A, Real F, Diogène J. Electrochemical Biosensor for the Assessment of Cell Viability Using Methylene Blue. Application to the Detection of Ciguatoxins in Fish from the Canary Islands. Anal Chem 2024; 96:19207-19212. [PMID: 39602562 DOI: 10.1021/acs.analchem.4c05174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Cell-based biosensors (CBBs) for the detection of marine neurotoxins such as ciguatoxins (CTXs) are of high interest due to the composite toxicological response they can provide and the low limits of quantification (LOQs) they can achieve with the use of sensitive neural cells. However, the development and validation of CBBs are challenging due to the use of living material and the need for appropriate signal transduction strategies. In this work, Neuro-2a cells have been immobilized on thin-film gold electrodes, and their viability after exposure to CTX1B has been evaluated with light optical microscopy as well as cyclic voltammetry (CV) and differential pulse voltammetry (DPV) using methylene blue (MB) as a redox indicator. An LOQ of 0.93 pg CTX1B/mL has been obtained. The CBB has been applied to the analysis of fish samples from the Canary Islands, one of them implicated in a ciguatera poisoning (CP) outbreak, and results have been compared with those obtained with a conventional cell-based assay (CBA), showing a very good agreement. The combination of the benefits of cells with those provided by biosensor platforms in terms of ease of use, miniaturization, automatization, and portability could result in the ideal analytical tool for CP management. Additionally, this is the first time MB is used as a cell viability indicator in a CBB, providing a new versatile approach for multiple applications.
Collapse
Affiliation(s)
- Mònica Campàs
- IRTA, Marine and Continental Waters, Ctra. Poble Nou km 5.5, 43540 La Ràpita, Spain
| | - Sandra Leonardo
- IRTA, Marine and Continental Waters, Ctra. Poble Nou km 5.5, 43540 La Ràpita, Spain
| | - Andrés Sanchez-Henao
- IRTA, Marine and Continental Waters, Ctra. Poble Nou km 5.5, 43540 La Ràpita, Spain
- University Institute of Animal Health and Food Safety (IUSA), University of Las Palmas de Gran Canaria, C/Trasmontan ̃a s/n, 35416 Arucas, Spain
| | - Fernando Real
- University Institute of Animal Health and Food Safety (IUSA), University of Las Palmas de Gran Canaria, C/Trasmontan ̃a s/n, 35416 Arucas, Spain
| | - Jorge Diogène
- IRTA, Marine and Continental Waters, Ctra. Poble Nou km 5.5, 43540 La Ràpita, Spain
| |
Collapse
|
2
|
Tarasiuk O, Invernizzi C, Alberti P. In vitro neurotoxicity testing: lessons from chemotherapy-induced peripheral neurotoxicity. Expert Opin Drug Metab Toxicol 2024; 20:1037-1052. [PMID: 39246127 DOI: 10.1080/17425255.2024.2401584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
INTRODUCTION Chemotherapy induced peripheral neurotoxicity (CIPN) is a long-lasting, or even permanent, late toxicity caused by largely used anticancer drugs. CIPN affects a growing population of cancer survivors and diminishes their quality of life since there is no curative/preventive treatment. Among several reasons for this unmet clinical need, there is an incomplete knowledge on mechanisms leading to CIPN. Therefore, bench side research is still greatly needed: in vitro studies are pivotal to both evaluate neurotoxicity mechanisms and potential neuroprotection strategies. AREAS COVERED Advantages and disadvantages of in vitro approaches are addressed with respect to their applicability to the CIPN field. Different cell cultures and techniques to assess neurotoxicity/neuroprotection are described. PubMed search-string: (chemotherapy-induced) AND (((neuropathy) OR neurotoxicity) OR neuropathic pain) AND (in vitro) AND (((((model) OR SH-SY5Y) OR PC12) OR iPSC) OR DRG neurons); (chemotherapy-induced) AND (((neuropathy) OR neurotoxicity) OR neuropathic pain) AND (model) AND (((neurite elongation) OR cell viability) OR morphology). No articles published before 1990 were selected. EXPERT OPINION CIPN is an ideal experimental setting to test axonal damage and, in general, peripheral nervous system mechanisms of disease and neuroprotection. Therefore, starting from robust preclinical data in this field, potentially, relevant biological rationale can be transferred to other human spontaneous diseases of the peripheral nervous system.
Collapse
Affiliation(s)
- Olga Tarasiuk
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- NeuroMI (Milan Center for Neuroscience), Milan, Italy
| | - Chiara Invernizzi
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- Neuroscience, School of Medicine and Surgery, Monza, Italy
| | - Paola Alberti
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- NeuroMI (Milan Center for Neuroscience), Milan, Italy
- Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| |
Collapse
|
3
|
Mi W, Liu S. Tetrodotoxin and the state-of-the-art progress of its associated analytical methods. Front Microbiol 2024; 15:1413741. [PMID: 39290516 PMCID: PMC11407752 DOI: 10.3389/fmicb.2024.1413741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Tetrodotoxin (TTX), which is found in various marine organisms, including pufferfish, shellfish, shrimp, crab, marine gastropods, and gobies, is an effective marine toxin and the cause of many seafood poisoning incidents. Owing to its toxicity and threat to public health, the development of simple, rapid, and efficient analytical methods to detect TTX in various food matrices has garnered increasing interest worldwide. Herein, we reviewed the structure and properties, origin and sources, toxicity and poisoning, and relevant legislative measures of TTX. Additionally, we have mainly reviewed the state-of-the-art progress of analytical methods for TTX detection in the past five years, such as bioassays, immunoassays, instrumental analysis, and biosensors, and summarized their advantages and limitations. Furthermore, this review provides an in-depth discussion of the most advanced biosensors, including cell-based biosensors, immunosensors, and aptasensors. Overall, this study provides useful insights into the future development and wide application of biosensors for TTX detection.
Collapse
Affiliation(s)
- Wei Mi
- School of Public Health, Binzhou Medical University, Yantai, China
| | - Sha Liu
- School of Public Health, Binzhou Medical University, Yantai, China
| |
Collapse
|
4
|
Ishibashi Y, Nagafuku N, Kinoshita K, Okamura A, Shirakawa T, Suzuki I. Verification of the seizure liability of compounds based on their in vitro functional activity in cultured rat cortical neurons and co-cultured human iPSC-derived neurons with astrocytes and in vivo extrapolation to cerebrospinal fluid concentration. Toxicol Appl Pharmacol 2023; 476:116675. [PMID: 37661062 DOI: 10.1016/j.taap.2023.116675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/26/2023] [Accepted: 08/30/2023] [Indexed: 09/05/2023]
Abstract
Methodical screening of safe and efficient drug candidate compounds is crucial for drug development. A high-throughput and accurate compound evaluation method targeting the central nervous system can be developed using in vitro neural networks. In particular, an evaluation system based on a human-derived neural network that can act as an alternative to animal experiments is desirable to avoid interspecific differences. A microelectrode array (MEA) is one such evaluation system, and can measure in vitro neural activity; however, studies on compound evaluation criteria and in vitro to in vivo extrapolation are scarce. In this study, we identified the parameters that can eliminate the effects of solvents from neural activity data obtained using MEA allow for accurate compound evaluation. Additionally, we resolved the issue associated with compound evaluation criteria during MEA using principal component analysis by considering the neuronal activity exceeding standard deviation (SD) of the solvent as indicator of seizurogenic potential. Overall, 10 seizurogenic compounds and three negative controls were assessed using MEA-based co-cultured human-induced pluripotent stem cell-derived neurons and astrocytes, and primary rat cortical neurons. In addition, we determined rat cerebrospinal fluid (CSF) concentrations during tremor and convulsion in response to exposure to test compounds. To characterize the in vitro to in vivo extrapolation and species differences, we compared the concentrations at which neuronal activity exceeding the SD range of the solvent was detectable using the MEA system and rat CSF concentration.
Collapse
Affiliation(s)
- Y Ishibashi
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi 982-8577, Japan
| | - N Nagafuku
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi 982-8577, Japan
| | - K Kinoshita
- Drug Safety Research Labs, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - A Okamura
- Drug Safety Research Labs, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - T Shirakawa
- Drug Safety Research Labs, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - I Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi 982-8577, Japan.
| |
Collapse
|
5
|
Chen J, Noorlander A, Wesseling S, Bouwmeester H, Kramer NI, Rietjens IMCM. Integrating In Vitro Data and Physiologically Based Kinetic Modeling to Predict and Compare Acute Neurotoxic Doses of Saxitoxin in Rats, Mice, and Humans. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023. [PMID: 37478462 PMCID: PMC10399293 DOI: 10.1021/acs.est.3c01987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
Current climate trends are likely to expand the geographic distribution of the toxigenic microalgae and concomitant phycotoxins, making intoxications by such toxins a global phenomenon. Among various phycotoxins, saxitoxin (STX) acts as a neurotoxin that might cause severe neurological symptoms in mammals following consumptions of contaminated seafood. To derive a point of departure (POD) for human health risk assessment upon acute neurotoxicity induced by oral STX exposure, a physiologically based kinetic (PBK) modeling-facilitated quantitative in vitro to in vivo extrapolation (QIVIVE) approach was employed. The PBK models for rats, mice, and humans were built using parameters from the literature, in vitro experiments, and in silico predictions. Available in vitro toxicity data for STX were converted to in vivo dose-response curves via the PBK models established for these three species, and POD values were derived from the predicted curves and compared to reported in vivo toxicity data. Interspecies differences in acute STX toxicity between rodents and humans were found, and they appeared to be mainly due to differences in toxicokinetics. The described approach resulted in adequate predictions for acute oral STX exposure, indicating that new approach methodologies, when appropriately integrated, can be used in a 3R-based chemical risk assessment paradigm.
Collapse
Affiliation(s)
- Jiaqi Chen
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, Wageningen, Gelderland 6708 WE, The Netherlands
| | - Annelies Noorlander
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, Wageningen, Gelderland 6708 WE, The Netherlands
| | - Sebastiaan Wesseling
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, Wageningen, Gelderland 6708 WE, The Netherlands
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, Wageningen, Gelderland 6708 WE, The Netherlands
| | - Nynke I Kramer
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, Wageningen, Gelderland 6708 WE, The Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, Wageningen, Gelderland 6708 WE, The Netherlands
| |
Collapse
|
6
|
Reverté J, Alkassar M, Diogène J, Campàs M. Detection of Ciguatoxins and Tetrodotoxins in Seafood with Biosensors and Other Smart Bioanalytical Systems. Foods 2023; 12:foods12102043. [PMID: 37238861 DOI: 10.3390/foods12102043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
The emergence of marine toxins such as ciguatoxins (CTXs) and tetrodotoxins (TTXs) in non-endemic regions may pose a serious food safety threat and public health concern if proper control measures are not applied. This article provides an overview of the main biorecognition molecules used for the detection of CTXs and TTXs and the different assay configurations and transduction strategies explored in the development of biosensors and other biotechnological tools for these marine toxins. The advantages and limitations of the systems based on cells, receptors, antibodies, and aptamers are described, and new challenges in marine toxin detection are identified. The validation of these smart bioanalytical systems through analysis of samples and comparison with other techniques is also rationally discussed. These tools have already been demonstrated to be useful in the detection and quantification of CTXs and TTXs, and are, therefore, highly promising for their implementation in research activities and monitoring programs.
Collapse
Affiliation(s)
- Jaume Reverté
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Ctra. Poble Nou km 5.5, 43540 La Ràpita, Spain
| | - Mounira Alkassar
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Ctra. Poble Nou km 5.5, 43540 La Ràpita, Spain
| | - Jorge Diogène
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Ctra. Poble Nou km 5.5, 43540 La Ràpita, Spain
| | - Mònica Campàs
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Ctra. Poble Nou km 5.5, 43540 La Ràpita, Spain
| |
Collapse
|
7
|
Gerber LS, van Kleef RGDM, Fokkens P, Cassee FR, Westerink RH. In vitro neurotoxicity screening of engine oil- and hydraulic fluid-derived aircraft cabin bleed-air contamination. Neurotoxicology 2023; 96:184-196. [PMID: 37120036 DOI: 10.1016/j.neuro.2023.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/01/2023]
Abstract
In most airplanes, cabin air is extracted from the turbine compressors, so-called bleed air. Bleed air can become contaminated by leakage of engine oil or hydraulic fluid and possible neurotoxic constituents, like triphenyl phosphate (TPhP) and tributyl phosphate (TBP). The aim of this study was to characterize the neurotoxic hazard of TBP and TPhP, and to compare this with the possible hazard of fumes originating from engine oils and hydraulic fluids in vitro. Effects on spontaneous neuronal activity were recorded in rat primary cortical cultures grown on microelectrode arrays following exposure for 0.5h (acute), and 24h and 48h (prolonged) to TBP and TPhP (0.01 - 100µM) or fume extracts (1 - 100µg/mL) prepared from four selected engine oils and two hydraulic fluids by a laboratory bleed air simulator. TPhP and TBP concentration-dependently reduced neuronal activity with equal potency, particularly during acute exposure (TPhP IC50: 10 - 12µM; TBP IC50: 15 - 18µM). Engine oil-derived fume extracts persistently reduced neuronal activity. Hydraulic fluid-derived fume extracts showed a stronger inhibition during 0.5h exposure, but the degree of inhibition attenuates during 48h. Overall, fume extracts from hydraulic fluids were more potent than those from engine oils, in particular during 0.5h exposure, although the higher toxicity is unlikely to be due only to higher levels of TBP and TPhP in hydraulic fluids. Our combined data show that bleed air contaminants originating from selected engine oils or hydraulic fluids exhibit neurotoxic hazard in vitro, with fumes derived from the selected hydraulic fluids being most potent.
Collapse
Affiliation(s)
- Lora-Sophie Gerber
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Regina G D M van Kleef
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Paul Fokkens
- National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Flemming R Cassee
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Remco Hs Westerink
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
8
|
Enterovirus D-68 Infection of Primary Rat Cortical Neurons: Entry, Replication, and Functional Consequences. mBio 2023; 14:e0024523. [PMID: 36877033 PMCID: PMC10127580 DOI: 10.1128/mbio.00245-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Enterovirus D68 (EV-D68) is an emerging pathogen associated with mild to severe respiratory disease. Since 2014, EV-D68 is also linked to acute flaccid myelitis (AFM), causing paralysis and muscle weakness in children. However, it remains unclear whether this is due to an increased pathogenicity of contemporary EV-D68 clades or increased awareness and detection of this virus. Here, we describe an infection model of primary rat cortical neurons to study the entry, replication, and functional consequences of different EV-D68 strains, including historical and contemporary strains. We demonstrate that sialic acids are important (co)receptors for infection of both neurons and respiratory epithelial cells. Using a collection of glycoengineered isogenic HEK293 cell lines, we show that sialic acids on either N-glycans or glycosphingolipids can be used for infection. Additionally, we show that both excitatory glutamatergic and inhibitory GABA-ergic neurons are susceptible and permissive to historical and contemporary EV-D68 strains. EV-D68 infection of neurons leads to the reorganization of the Golgi-endomembranes forming replication organelles, first in the soma and later in the processes. Finally, we demonstrate that the spontaneous neuronal activity of EV-D68-infected neuronal network cultured on microelectrode arrays (MEA) is decreased, independent of the virus strain. Collectively, our findings provide novel insights into neurotropism and -pathology of different EV-D68 strains, and argue that it is unlikely that increased neurotropism is a recently acquired phenotype of a specific genetic lineage. IMPORTANCE Acute flaccid myelitis (AFM) is a serious neurological illness characterized by muscle weakness and paralysis in children. Since 2014, outbreaks of AFM have emerged worldwide, and they appear to be caused by nonpolio enteroviruses, particularly enterovirus-D68 (EV-D68), an unusual enterovirus that is known to mainly cause respiratory disease. It is unknown whether these outbreaks reflect a change of EV-D68 pathogenicity or are due to increased detection and awareness of this virus in recent years. To gain more insight herein, it is crucial to define how historical and circulating EV-D68 strains infect and replicate in neurons and how they affect their physiology. This study compares the entry and replication in neurons and the functional consequences on the neural network upon infection with an old "historical" strain and contemporary "circulating" strains of EV-D68.
Collapse
|
9
|
Wei LN, Luo L, Wang BZ, Lei HT, Guan T, Shen YD, Wang H, Xu ZL. Biosensors for detection of paralytic shellfish toxins: Recognition elements and transduction technologies. Trends Food Sci Technol 2023. [DOI: 10.1016/j.tifs.2023.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
10
|
Tukker AM, Vrolijk MF, van Kleef RGDM, Sijm DTHM, Westerink RHS. Mixture effects of tetrodotoxin (TTX) and drugs targeting voltage-gated sodium channels on spontaneous neuronal activity in vitro. Toxicol Lett 2023; 373:53-61. [PMID: 36375636 DOI: 10.1016/j.toxlet.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/19/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
Tetrodotoxin (TTX) potently inhibits TTX-sensitive voltage-gated sodium (NaV) channels in nerve and muscle cells, potentially resulting in depressed neurotransmission, paralysis and death from respiratory failure. Since a wide range of pharmaceutical drugs is known to also act on NaV channels, the use of medicines could predispose individuals to a higher susceptibility towards TTX toxicity. We therefore first assessed the inhibitory effect of selected medicines that act on TTX-sensitive (Riluzole, Chloroquine, Fluoxetine, Valproic acid, Lamotrigine, Lidocaine) and TTX-resistant (Carbamazepine, Mexiletine, Flecainide) NaV channels on spontaneous neuronal activity of rat primary cortical cultures grown on microelectrode arrays (MEA). After establishing concentration-effect curves, binary mixtures of the medicines with TTX at calculated NOEC, IC20 and IC50 values were used to determine if pharmacodynamic interactions occur between TTX and these drugs on spontaneous neuronal activity. At IC20 and IC50 values, all medicines significantly increased the inhibitory effect of TTX on spontaneous neuronal activity of rat cortical cells in vitro. Subsequent experiments using human iPSC-derived neuronal co-cultures grown on MEAs confirmed the ability of selected medicines (Carbamazepine, Flecainide, Riluzole, Lidocaine) to inhibit spontaneous neuronal activity. Despite the need for additional experiments using human iPSC-derived neuronal co-cultures, our combined data already highlight the importance of identifying and including vulnerable risk groups in the risk assessment of TTX.
Collapse
Affiliation(s)
- Anke M Tukker
- Neurotoxicology Research Group, Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 TD Utrecht, the Netherlands
| | - Misha F Vrolijk
- Faculty of Health, Medicine and Life Sciences, Department of Pharmacology and Toxicology, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Regina G D M van Kleef
- Neurotoxicology Research Group, Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 TD Utrecht, the Netherlands
| | - Dick T H M Sijm
- Faculty of Health, Medicine and Life Sciences, Department of Pharmacology and Toxicology, Maastricht University, 6229 ER Maastricht, the Netherlands; Office for Risk Assessment and Research, Netherlands Food and Consumer Product Safety Authority (NVWA), 3540 AA Utrecht, the Netherlands
| | - Remco H S Westerink
- Neurotoxicology Research Group, Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 TD Utrecht, the Netherlands.
| |
Collapse
|
11
|
Choo SS, Keever JY, Brown J, Strickland JD, Shafer TJ. Assaying Spontaneous Network Activity and Cellular Viability Using Multi-Well Microelectrode Arrays. Methods Mol Biol 2023; 2644:133-154. [PMID: 37142920 DOI: 10.1007/978-1-0716-3052-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Microelectrode array (MEA) technology is a neurophysiological method that allows for the measurement of spontaneous or evoked neural activity to determine chemical effects thereon. Following assessment of compound effects on multiple endpoints that evaluate network function, a cell viability endpoint in the same well is determined using a multiplexed approach. Recently, it has become possible to measure electrical impedance of cells attached to the electrodes, where greater impedance indicates greater number of cells attached. This would allow rapid and repeated assessments of cell health as the neural network develops in longer exposure assays without impacting cell health. Typically, the lactate dehydrogenase (LDH) assay for cytotoxity and CellTiter-Blue® (CTB) assay for cell viability are only performed at the end of the chemical exposure period because these assays involve lysing of the cells. Procedures describing the multiplexed methods in acute and network formation screening are included in this chapter.
Collapse
Affiliation(s)
- Seline S Choo
- Oak Ridge Institute for Science and Engineering, Oak Ridge, TN, USA
- Rapid Assay Development Branch, Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Jackson Y Keever
- Rapid Assay Development Branch, Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC, USA
- Oak Ridge Associated Universities Student Contractor, Oak Ridge, TN, USA
| | - Jasmine Brown
- Rapid Assay Development Branch, Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| | - Jenna D Strickland
- Axion Biosystems, Atlanta, GA, USA
- LabCorp Drug Development, Madison, WI, USA
| | - Timothy J Shafer
- Rapid Assay Development Branch, Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC, USA.
| |
Collapse
|
12
|
Gerber LS, Heusinkveld HJ, Langendoen C, Stahlmecke B, Schins RPF, Westerink RHS. Acute, sub-chronic and chronic exposures to TiO2 and Ag nanoparticles differentially affects neuronal function in vitro. Neurotoxicology 2022; 93:311-323. [DOI: 10.1016/j.neuro.2022.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/07/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
|
13
|
Alkassar M, Leonardo S, Diogène J, Campàs M. Immobilisation of Neuro-2a cells on electrodes and electrochemical detection of MTT formazan crystals to assess their viability. Bioelectrochemistry 2022; 148:108274. [DOI: 10.1016/j.bioelechem.2022.108274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/02/2022]
|
14
|
Sun X, Xiang Y, Liu M, Xu X, Zhang L, Zhuang L, Wang P, Wang Q. High-performance and -efficiency cardiomyocyte-based potential biosensor for temporal-specific detection of ion channel marine toxins. Biosens Bioelectron 2022; 220:114837. [DOI: 10.1016/j.bios.2022.114837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/15/2022] [Accepted: 10/18/2022] [Indexed: 11/30/2022]
|
15
|
Recent advances of three-dimensional micro-environmental constructions on cell-based biosensors and perspectives in food safety. Biosens Bioelectron 2022; 216:114601. [DOI: 10.1016/j.bios.2022.114601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 06/29/2022] [Accepted: 07/25/2022] [Indexed: 11/21/2022]
|
16
|
Functional and Structural Biological Methods for Palytoxin Detection. JOURNAL OF MARINE SCIENCE AND ENGINEERING 2022. [DOI: 10.3390/jmse10070916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Palytoxin (PLTX) and its analogues are marine polyethers identified in Palythoa and Zoanthus corals, Ostreopsis dinoflagellates, and Trichodesmium cyanobacteria. Humans can be exposed to these toxins by different routes with a series of adverse effects but the most severe risk is associated with poisonings by the consumption of edible marine organisms accumulating these toxins, as occurs in (sub)-tropical areas. In temperate areas, adverse effects ascribed to PLTXs have been recorded after inhalation of marine aerosols and/or cutaneous contact with seawater during Ostreopsis blooms, as well as during cleaning procedures of Palythoa-containing home aquaria. Besides instrumental analytical methods, in the last years a series of alternative or complementary methods based on biological/biochemical tools have been developed for the rapid and specific PLTX detection required for risk assessment. These methods are usually sensitive, cost- and time-effective, and do not require highly specialized operators. Among them, structural immunoassays and functional cell-based assays are reviewed. The availability of specific anti-PLTX antibodies allowed the development of different sensitive structural assays, suitable for its detection also in complex matrices, such as mussels. In addition, knowing the mechanism of PLTX action, a series of functional identification methods has been developed. Despite some of them being limited by matrix effects and specificity issues, biological methods for PLTX detection represent a feasible tool, suitable for rapid screening.
Collapse
|
17
|
Dobreniecki S, Mendez E, Lowit A, Freudenrich TM, Wallace K, Carpenter A, Wetmore BA, Kreutz A, Korol-Bexell E, Friedman KP, Shafer TJ. Integration of toxicodynamic and toxicokinetic new approach methods into a weight-of-evidence analysis for pesticide developmental neurotoxicity assessment: A case-study with DL- and L-glufosinate. Regul Toxicol Pharmacol 2022; 131:105167. [PMID: 35413399 DOI: 10.1016/j.yrtph.2022.105167] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/14/2022] [Accepted: 04/06/2022] [Indexed: 01/13/2023]
Abstract
DL-glufosinate ammonium (DL-GLF) is a registered herbicide for which a guideline Developmental Neurotoxicity (DNT) study has been conducted. Offspring effects included altered brain morphometrics, decreased body weight, and increased motor activity. Guideline DNT studies are not available for its enriched isomers L-GLF acid and L-GLF ammonium; conducting one would be time consuming, resource-intensive, and possibly redundant given the existing DL-GLF DNT. To support deciding whether to request a guideline DNT study for the L-GLF isomers, DL-GLF and the L-GLF isomers were screened using in vitro assays for network formation and neurite outgrowth. DL-GLF and L-GLF isomers were without effects in both assays. DL-GLF and L-GLF (1-100 μM) isomers increased mean firing rate of mature networks to 120-140% of baseline. In vitro toxicokinetic assessments were used to derive administered equivalent doses (AEDs) for the in vitro testing concentrations. The AED for L-GLF was ∼3X higher than the NOAEL from the DL-GLF DNT indicating that the available guideline study would be protective of potential DNT due to L-GLF exposure. Based in part on the results of these in vitro studies, EPA is not requiring L-GLF isomer guideline DNT studies, thereby providing a case study for a useful application of DNT screening assays.
Collapse
Affiliation(s)
| | | | - Anna Lowit
- Office of Pesticide Programs USEPA, Washington, DC, USA
| | - Theresa M Freudenrich
- Center for Computational Toxicology and Exposure, Office of Research and Development. US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Kathleen Wallace
- Center for Computational Toxicology and Exposure, Office of Research and Development. US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Amy Carpenter
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Barbara A Wetmore
- Center for Computational Toxicology and Exposure, Office of Research and Development. US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Anna Kreutz
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | | | - Katie Paul Friedman
- Center for Computational Toxicology and Exposure, Office of Research and Development. US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Timothy J Shafer
- Center for Computational Toxicology and Exposure, Office of Research and Development. US Environmental Protection Agency, Research Triangle Park, NC, USA.
| |
Collapse
|
18
|
Noorlander A, Zhang M, van Ravenzwaay B, Rietjens IMCM. Use of physiologically based kinetic modeling-facilitated reverse dosimetry to predict in vivo acute toxicity of tetrodotoxin in rodents. Toxicol Sci 2022; 187:127-138. [PMID: 35218365 PMCID: PMC9041554 DOI: 10.1093/toxsci/kfac022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
In this study, the ability of a new in vitro/in silico quantitative in vitro–in vivo extrapolation (QIVIVE) methodology was assessed to predict the in vivo neurotoxicity of tetrodotoxin (TTX) in rodents. In vitro concentration–response data of TTX obtained in a multielectrode array assay with primary rat neonatal cortical cells and in an effect study with mouse neuro-2a cells were quantitatively extrapolated into in vivo dose–response data, using newly developed physiologically based kinetic (PBK) models for TTX in rats and mice. Incorporating a kidney compartment accounting for active renal excretion in the PBK models proved to be essential for its performance. To evaluate the predictions, QIVIVE-derived dose–response data were compared with in vivo data on neurotoxicity in rats and mice upon oral and parenteral dosing. The results revealed that for both rats and mice the predicted dose–response data matched the data from available in vivo studies well. It is concluded that PBK modeling-based reserve dosimetry of in vitro TTX effect data can adequately predict the in vivo neurotoxicity of TTX in rodents, providing a novel proof-of-principle for this methodology.
Collapse
Affiliation(s)
- Annelies Noorlander
- Division of Toxicology, Wageningen University, Stippeneng 4, Wageningen, 6708 WE, the Netherlands
| | - Mengying Zhang
- Division of Toxicology, Wageningen University, Stippeneng 4, Wageningen, 6708 WE, the Netherlands
| | - Bennard van Ravenzwaay
- Division of Toxicology, Wageningen University, Stippeneng 4, Wageningen, 6708 WE, the Netherlands.,Experimental Toxicology and Ecology, BASF SE, Z 470, Ludwigshafen, 67056, Germany
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University, Stippeneng 4, Wageningen, 6708 WE, the Netherlands
| |
Collapse
|
19
|
Katikou P, Gokbulut C, Kosker AR, Campàs M, Ozogul F. An Updated Review of Tetrodotoxin and Its Peculiarities. Mar Drugs 2022; 20:md20010047. [PMID: 35049902 PMCID: PMC8780202 DOI: 10.3390/md20010047] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 12/19/2022] Open
Abstract
Tetrodotoxin (TTX) is a crystalline, weakly basic, colorless organic substance and is one of the most potent marine toxins known. Although TTX was first isolated from pufferfish, it has been found in numerous other marine organisms and a few terrestrial species. Moreover, tetrodotoxication is still an important health problem today, as TTX has no known antidote. TTX poisonings were most commonly reported from Japan, Thailand, and China, but today the risk of TTX poisoning is spreading around the world. Recent studies have shown that TTX-containing fish are being found in other regions of the Pacific and in the Indian Ocean, as well as the Mediterranean Sea. This review aims to summarize pertinent information available to date on the structure, origin, distribution, mechanism of action of TTX and analytical methods used for the detection of TTX, as well as on TTX-containing organisms, symptoms of TTX poisoning, and incidence worldwide.
Collapse
Affiliation(s)
- Panagiota Katikou
- Ministry of Rural Development and Food, Directorate of Research, Innovation and Education, Hapsa & Karatasou 1, 54626 Thessaloniki, Greece
- Correspondence: (P.K.); (F.O.)
| | - Cengiz Gokbulut
- Department of Pharmacology, Faculty of Medicine, Balikesir University, Balikesir 10145, Turkey;
| | - Ali Rıza Kosker
- Department of Seafood Processing Technology, Faculty of Fisheries, Cukurova University, Adana 01330, Turkey;
| | - Mònica Campàs
- IRTA, Ctra Poble Nou km 5.5, 43540 Sant Carles de la Ràpita, Spain;
| | - Fatih Ozogul
- Department of Seafood Processing Technology, Faculty of Fisheries, Cukurova University, Adana 01330, Turkey;
- Correspondence: (P.K.); (F.O.)
| |
Collapse
|
20
|
Tukker AM, Westerink RHS. Novel test strategies for in vitro seizure liability assessment. Expert Opin Drug Metab Toxicol 2021; 17:923-936. [PMID: 33595380 PMCID: PMC8367052 DOI: 10.1080/17425255.2021.1876026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/11/2021] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The increasing incidence of mental illnesses and neurodegenerative diseases results in a high demand for drugs targeting the central nervous system (CNS). These drugs easily reach the CNS, have a high affinity for CNS targets, and are prone to cause seizures as an adverse drug reaction. Current seizure liability assessment heavily depends on in vivo or ex vivo animal models and is therefore ethically debated, labor intensive, expensive, and not always predictive for human risk. AREAS COVERED The demand for CNS drugs urges the development of alternative safety assessment strategies. Yet, the complexity of the CNS hampers reliable detection of compound-induced seizures. This review provides an overview of the requirements of in vitro seizure liability assays and highlights recent advances, including micro-electrode array (MEA) recordings using rodent and human cell models. EXPERT OPINION Successful and cost-effective replacement of in vivo and ex vivo models for seizure liability screening can reduce animal use for drug development, while increasing the predictive value of the assays, particularly if human cell models are used. However, these novel test strategies require further validation and standardization as well as additional refinements to better mimic the human in vivo situation and increase their predictive value.
Collapse
Affiliation(s)
- Anke M. Tukker
- School of Health Sciences, Purdue University, Hall for Discovery and Learning Research (DLR 339), INUSA
| | - Remco H. S. Westerink
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, TD Utrecht, The Netherlands
| |
Collapse
|
21
|
Gerber LS, van Melis LVJ, van Kleef RGDM, de Groot A, Westerink RHS. Culture of Rat Primary Cortical Cells for Microelectrode Array (MEA) Recordings to Screen for Acute and Developmental Neurotoxicity. Curr Protoc 2021; 1:e158. [PMID: 34152700 DOI: 10.1002/cpz1.158] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Neurotoxicity testing of chemicals, drug candidates, and environmental pollutants still relies on extensive in vivo studies that are very costly, time-consuming, and ethically debated due to the large number of animals typically used. Currently, rat primary cortical cultures are widely used for in vitro neurotoxicity studies, as they closely resemble the in vitro brain with respect to the diversity of cell types, their physiological functions, and the pathological processes that they undergo. Common in vitro assays for neurotoxicity screening often focus on very target-specific endpoints such as morphological, biochemical, or electrophysiological changes, and such narrow focus can hamper translation and interpretation. Microelectrode array (MEA) recordings provide a non-invasive platform for extracellular recording of electrical activity of cultured neuronal cells, thereby enabling the evaluation of changes in neuronal (network) function as a sensitive and integrated endpoint for neurotoxicity screening. Here, we describe an in vitro approach for assessing changes in neuronal network function as a measure for neurotoxicity, using rat primary cortical cultures grown on MEAs. We provide a detailed protocol for the culture of rat primary cortical cells, and describe several experimental procedures to address acute, subchronic, and chronic exposure scenarios. We additionally describe the steps for processing and analyzing MEA and cell viability data. © 2021 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Isolation and culture of rat primary cortical cells on 48-well MEA plates Support Protocol 1: Pretreatment and washing of 48-well MEA plates before first use or for re-use Support Protocol 2: Coating of 48-well MEA plates with 0.1% PEI solution Basic Protocol 2: MEA measurements during acute exposure Alternate Protocol 1: MEA measurements during subchronic exposure Alternate Protocol 2: MEA measurements during chronic exposure Support Protocol 3: Determination of cell viability after MEA experiments Basic Protocol 3: MEA data processing Basic Protocol 4: Analyzing MEA experiments after acute and subchronic exposure Alternate Protocol 3: Analyzing MEA experiments after chronic exposure.
Collapse
Affiliation(s)
- Lora-Sophie Gerber
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Lennart V J van Melis
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Regina G D M van Kleef
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Aart de Groot
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Remco H S Westerink
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
22
|
Puschhof J, Post Y, Beumer J, Kerkkamp HM, Bittenbinder M, Vonk FJ, Casewell NR, Richardson MK, Clevers H. Derivation of snake venom gland organoids for in vitro venom production. Nat Protoc 2021; 16:1494-1510. [PMID: 33504990 DOI: 10.1038/s41596-020-00463-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 11/12/2020] [Indexed: 11/09/2022]
Abstract
More than 400,000 people each year suffer adverse effects following bites from venomous snakes. However, snake venom is also a rich source of bioactive molecules with known or potential therapeutic applications. Manually 'milking' snakes is the most common method to obtain venom. Safer alternative methods to produce venom would facilitate the production of both antivenom and novel therapeutics. This protocol describes the generation, maintenance and selected applications of snake venom gland organoids. Snake venom gland organoids are 3D culture models that can be derived within days from embryonic or adult venom gland tissues from several snake species and can be maintained long-term (we have cultured some organoids for more than 2 years). We have successfully used the protocol with glands from late-stage embryos and recently deceased adult snakes. The cellular heterogeneity of the venom gland is maintained in the organoids, and cell type composition can be controlled through changes in media composition. We describe in detail how to derive and grow the organoids, how to dissociate them into single cells, and how to cryopreserve and differentiate them into toxin-producing organoids. We also provide guidance on useful downstream assays, specifically quantitative real-time PCR, bulk and single-cell RNA sequencing, immunofluorescence, immunohistochemistry, fluorescence in situ hybridization, scanning and transmission electron microscopy and genetic engineering. This stepwise protocol can be performed in any laboratory with tissue culture equipment and enables studies of venom production, differentiation and cellular heterogeneity.
Collapse
Affiliation(s)
- Jens Puschhof
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, the Netherlands.,Oncode Institute, Hubrecht Institute, Utrecht, the Netherlands
| | - Yorick Post
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, the Netherlands.,Oncode Institute, Hubrecht Institute, Utrecht, the Netherlands
| | - Joep Beumer
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, the Netherlands.,Oncode Institute, Hubrecht Institute, Utrecht, the Netherlands
| | - Harald M Kerkkamp
- Naturalis Biodiversity Center, Leiden, the Netherlands.,Institute of Biology Leiden, Department of Animal Science and Health, Leiden, the Netherlands
| | - Matyas Bittenbinder
- Naturalis Biodiversity Center, Leiden, the Netherlands.,Amsterdam Institute for Molecules Medicines and Systems, Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Freek J Vonk
- Naturalis Biodiversity Center, Leiden, the Netherlands
| | - Nicholas R Casewell
- Centre for Snakebite Research & Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Michael K Richardson
- Institute of Biology Leiden, Department of Animal Science and Health, Leiden, the Netherlands
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, the Netherlands. .,Oncode Institute, Hubrecht Institute, Utrecht, the Netherlands.
| |
Collapse
|
23
|
Loser D, Schaefer J, Danker T, Möller C, Brüll M, Suciu I, Ückert AK, Klima S, Leist M, Kraushaar U. Human neuronal signaling and communication assays to assess functional neurotoxicity. Arch Toxicol 2021; 95:229-252. [PMID: 33269408 PMCID: PMC7811517 DOI: 10.1007/s00204-020-02956-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/16/2020] [Indexed: 01/08/2023]
Abstract
Prediction of drug toxicity on the human nervous system still relies mainly on animal experiments. Here, we developed an alternative system allowing assessment of complex signaling in both individual human neurons and on the network level. The LUHMES cultures used for our approach can be cultured in 384-well plates with high reproducibility. We established here high-throughput quantification of free intracellular Ca2+ concentrations [Ca2+]i as broadly applicable surrogate of neuronal activity and verified the main processes by patch clamp recordings. Initially, we characterized the expression pattern of many neuronal signaling components and selected the purinergic receptors to demonstrate the applicability of the [Ca2+]i signals for quantitative characterization of agonist and antagonist responses on classical ionotropic neurotransmitter receptors. This included receptor sub-typing and the characterization of the anti-parasitic drug suramin as modulator of the cellular response to ATP. To exemplify potential studies on ion channels, we characterized voltage-gated sodium channels and their inhibition by tetrodotoxin, saxitoxin and lidocaine, as well as their opening by the plant alkaloid veratridine and the food-relevant marine biotoxin ciguatoxin. Even broader applicability of [Ca2+]i quantification as an end point was demonstrated by measurements of dopamine transporter activity based on the membrane potential-changing activity of this neurotransmitter carrier. The substrates dopamine or amphetamine triggered [Ca2+]i oscillations that were synchronized over the entire culture dish. We identified compounds that modified these oscillations by interfering with various ion channels. Thus, this new test system allows multiple types of neuronal signaling, within and between cells, to be assessed, quantified and characterized for their potential disturbance.
Collapse
Affiliation(s)
- Dominik Loser
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
- NMI TT GmbH, 72770, Reutlingen, Germany
- Life Sciences Faculty, Albstadt-Sigmaringen University, 72488, Sigmaringen, Germany
| | - Jasmin Schaefer
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
- NMI TT GmbH, 72770, Reutlingen, Germany
| | | | - Clemens Möller
- Life Sciences Faculty, Albstadt-Sigmaringen University, 72488, Sigmaringen, Germany
| | - Markus Brüll
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Ilinca Suciu
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Anna-Katharina Ückert
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Stefanie Klima
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany.
| | - Udo Kraushaar
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, 72770, Reutlingen, Germany
| |
Collapse
|
24
|
Tukker AM, Wijnolts FMJ, de Groot A, Westerink RHS. Applicability of hiPSC-Derived Neuronal Cocultures and Rodent Primary Cortical Cultures for In Vitro Seizure Liability Assessment. Toxicol Sci 2020; 178:71-87. [PMID: 32866265 PMCID: PMC7657345 DOI: 10.1093/toxsci/kfaa136] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
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.
Collapse
Affiliation(s)
- Anke M Tukker
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, NL-3508 TD Utrecht, The Netherlands
| | - Fiona M J Wijnolts
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, NL-3508 TD Utrecht, The Netherlands
| | - Aart de Groot
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, NL-3508 TD Utrecht, The Netherlands
| | - Remco H S Westerink
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, NL-3508 TD Utrecht, The Netherlands
| |
Collapse
|
25
|
Funada M, Takebayashi-Ohsawa M, Tomiyama KI. Synthetic cannabinoids enhanced ethanol-induced motor impairments through reduction of central glutamate neurotransmission. Toxicol Appl Pharmacol 2020; 408:115283. [PMID: 33068620 DOI: 10.1016/j.taap.2020.115283] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/29/2020] [Accepted: 10/11/2020] [Indexed: 01/05/2023]
Abstract
Marijuana or synthetic cannabinoids and alcohol are often used together, with these combinations causing motor impairments that can subsequently lead to motor vehicle accidents. This study investigated the combined use of both synthetic cannabinoids and ethanol and their effect on motor coordination in mice in addition to examining the neurochemical changes in the cerebellum. Ethanol (2 g/kg, i.p.) significantly induced motor impairment in the accelerating rotarod test in mice. Furthermore, ethanol-induced motor impairments were further accentuated when combined with the synthetic cannabinoid, JWH-018 or AB-CHMINACA. The enhancement effects of the synthetic cannabinoids were completely antagonized by pretreatment with the selective CB1 receptor antagonist AM251, but not by the selective CB2 receptor antagonist AM630. Neurochemical study results showed that ethanol caused a reduction in the extracellular glutamate levels in the cerebellum during periods of ethanol-induced motor impairment. In addition to the enhanced motor impairment seen when ethanol was combined with JWH-018, these combinations also enhanced the reduction of the extracellular glutamate levels in the cerebellum. We additionally used microelectrode array recordings to examine the effects of ethanol and/or JWH-018 on the spontaneous network activity in primary cultures from mouse cerebellum. Results showed that ethanol combined with JWH-018 significantly reduced spontaneous neuronal network activity in the primary cerebellar culture. Our findings demonstrate that ethanol-induced motor impairments are enhanced by synthetic cannabinoids, with these effects potentially mediated by CB1 receptors. An accentuated reduction of neurotransmissions in the cerebellum may play an important role in motor impairments caused by ethanol combined with synthetic cannabinoids.
Collapse
Affiliation(s)
- Masahiko Funada
- Department of Drug Dependence Research, National Institute of Mental Health, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8553, Japan.
| | - Mika Takebayashi-Ohsawa
- Department of Drug Dependence Research, National Institute of Mental Health, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8553, Japan
| | - Ken-Ich Tomiyama
- Department of Drug Dependence Research, National Institute of Mental Health, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8553, Japan
| |
Collapse
|
26
|
Alloisio S, Clericuzio M, Nobile M, Salis A, Damonte G, Canali C, Fortuna-Perez AP, Cornara L, Burlando B. Cannabis-like activity of Zornia latifolia Sm. detected in vitro on rat cortical neurons: major role of the flavone syzalterin. Drug Chem Toxicol 2020; 45:919-931. [PMID: 32628037 DOI: 10.1080/01480545.2020.1788057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Zornia latifolia is a plant suspected to possess psychoactive properties and marketed as a marijuana substitute under the name 'maconha brava'. In this study, the effects of fractions obtained from a 2-propanol extract of aerial portions of the plant were determined by multielectrode array (MEA) analyses on cultured networks of rat cortical neurons. Lipophilic (ZL_lipo, mainly containing flavonoid aglycones), and hydrophilic (ZL_hydro, mainly containing flavonoid glycosides) fractions were initially obtained from the raw extract. ZL_lipo significantly inhibited mean firing rate (MFR) and mean bursting rate (MBR) of MEA recordings, while ZL_hydro induced no inhibition. Column chromatography separation of ZL_lipo yielded five fractions (ZL1-ZL5), among which ZL1 induced the strongest MFR and MBR inhibition. NMR and HPLC-MS analyses of ZL1 revealed the prevalence of the common flavonoids genistein (1) and apigenin (2) (in about a 1:1 ratio), and the presence of the rare flavone syzalterin (6,8-dimethylapigenin) (3) as a minor compound. Exposures of MEA to apigenin and genistein standards did not induce the MFR and MBR inhibition observed with ZL1, whereas exposure to syzalterin standard or to a 1:9 mixture syzalterin-genistein induced effects similar to ZL1. These inhibitory effects were comparable to that observed with high-THC hashish, possibly accounting for the plant psychoactive properties. Data indicate that Z. latifolia, currently marketed as a free herbal product, should be subjected to measures of control. In addition, syzalterin showed distinctive pharmacological properties, opening the way to its possible exploitation as a neuroactive drug.
Collapse
Affiliation(s)
- Susanna Alloisio
- ETT Spa, Genova, Italy.,National Research Council (CNR), Institute of Biophysics, Genova, Italy
| | - Marco Clericuzio
- Department of Sciences and Technological Innovation (DISIT), University of Eastern Piedmont, Alessandria, Italy
| | - Mario Nobile
- National Research Council (CNR), Institute of Biophysics, Genova, Italy
| | - Annalisa Salis
- Center of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy
| | - Gianluca Damonte
- Center of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy.,Department of Experimental Medicine (DIMES), Section of Biochemistry, University of Genova, Genova, Italy
| | - Claudia Canali
- Forensic Science Police Service, Italian National Police, Genova, Italy
| | | | - Laura Cornara
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, Genova, Italy
| | - Bruno Burlando
- Department of Pharmacy (DIFAR), University of Genova, Genova, Italy
| |
Collapse
|
27
|
Post Y, Puschhof J, Beumer J, Kerkkamp HM, de Bakker MAG, Slagboom J, de Barbanson B, Wevers NR, Spijkers XM, Olivier T, Kazandjian TD, Ainsworth S, Iglesias CL, van de Wetering WJ, Heinz MC, van Ineveld RL, van Kleef RGDM, Begthel H, Korving J, Bar-Ephraim YE, Getreuer W, Rios AC, Westerink RHS, Snippert HJG, van Oudenaarden A, Peters PJ, Vonk FJ, Kool J, Richardson MK, Casewell NR, Clevers H. Snake Venom Gland Organoids. Cell 2020; 180:233-247.e21. [PMID: 31978343 DOI: 10.1016/j.cell.2019.11.038] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 10/29/2019] [Accepted: 11/27/2019] [Indexed: 12/12/2022]
Abstract
Wnt dependency and Lgr5 expression define multiple mammalian epithelial stem cell types. Under defined growth factor conditions, such adult stem cells (ASCs) grow as 3D organoids that recapitulate essential features of the pertinent epithelium. Here, we establish long-term expanding venom gland organoids from several snake species. The newly assembled transcriptome of the Cape coral snake reveals that organoids express high levels of toxin transcripts. Single-cell RNA sequencing of both organoids and primary tissue identifies distinct venom-expressing cell types as well as proliferative cells expressing homologs of known mammalian stem cell markers. A hard-wired regional heterogeneity in the expression of individual venom components is maintained in organoid cultures. Harvested venom peptides reflect crude venom composition and display biological activity. This study extends organoid technology to reptilian tissues and describes an experimentally tractable model system representing the snake venom gland.
Collapse
Affiliation(s)
- Yorick Post
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, 3584 CT Utrecht, the Netherlands; Oncode Institute, Hubrecht Institute, 3584 CT Utrecht, the Netherlands
| | - Jens Puschhof
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, 3584 CT Utrecht, the Netherlands; Oncode Institute, Hubrecht Institute, 3584 CT Utrecht, the Netherlands
| | - Joep Beumer
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, 3584 CT Utrecht, the Netherlands; Oncode Institute, Hubrecht Institute, 3584 CT Utrecht, the Netherlands
| | - Harald M Kerkkamp
- Naturalis Biodiversity Center, 2333 CR Leiden, the Netherlands; Institute of Biology Leiden, Department of Animal Science and Health, 2333 BE Leiden, the Netherlands
| | - Merijn A G de Bakker
- Institute of Biology Leiden, Department of Animal Science and Health, 2333 BE Leiden, the Netherlands
| | - Julien Slagboom
- Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, 1081 LA Amsterdam, the Netherlands
| | - Buys de Barbanson
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, 3584 CT Utrecht, the Netherlands; Oncode Institute, Hubrecht Institute, 3584 CT Utrecht, the Netherlands
| | - Nienke R Wevers
- Mimetas BV, Organ-on-a-Chip Company, 2333 CH Leiden, the Netherlands; Department of Cell and Chemical Biology, Leiden University Medical Centre, Einthovenweg 20, 2333 ZC Leiden, the Netherlands
| | - Xandor M Spijkers
- Mimetas BV, Organ-on-a-Chip Company, 2333 CH Leiden, the Netherlands; Department of Translational Neuroscience, Utrecht University Medical Center, 3584 CG Utrecht, the Netherlands
| | - Thomas Olivier
- Mimetas BV, Organ-on-a-Chip Company, 2333 CH Leiden, the Netherlands
| | - Taline D Kazandjian
- Centre for Snakebite Research & Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK
| | - Stuart Ainsworth
- Centre for Snakebite Research & Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK
| | - Carmen Lopez Iglesias
- The Maastricht Multimodal Molecular Imaging Institute, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Willine J van de Wetering
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, 3584 CT Utrecht, the Netherlands; The Maastricht Multimodal Molecular Imaging Institute, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Maria C Heinz
- Oncode Institute, Hubrecht Institute, 3584 CT Utrecht, the Netherlands; Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, the Netherlands
| | - Ravian L van Ineveld
- Oncode Institute, Hubrecht Institute, 3584 CT Utrecht, the Netherlands; The Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, the Netherlands
| | - Regina G D M van Kleef
- Neurotoxicology Research Group, Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Harry Begthel
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, 3584 CT Utrecht, the Netherlands; Oncode Institute, Hubrecht Institute, 3584 CT Utrecht, the Netherlands
| | - Jeroen Korving
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, 3584 CT Utrecht, the Netherlands; Oncode Institute, Hubrecht Institute, 3584 CT Utrecht, the Netherlands
| | - Yotam E Bar-Ephraim
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, 3584 CT Utrecht, the Netherlands; Oncode Institute, Hubrecht Institute, 3584 CT Utrecht, the Netherlands
| | | | - Anne C Rios
- Oncode Institute, Hubrecht Institute, 3584 CT Utrecht, the Netherlands; The Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, the Netherlands
| | - Remco H S Westerink
- Neurotoxicology Research Group, Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Hugo J G Snippert
- Oncode Institute, Hubrecht Institute, 3584 CT Utrecht, the Netherlands; Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, the Netherlands
| | - Alexander van Oudenaarden
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, 3584 CT Utrecht, the Netherlands; Oncode Institute, Hubrecht Institute, 3584 CT Utrecht, the Netherlands
| | - Peter J Peters
- The Maastricht Multimodal Molecular Imaging Institute, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Freek J Vonk
- Naturalis Biodiversity Center, 2333 CR Leiden, the Netherlands
| | - Jeroen Kool
- Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, 1081 LA Amsterdam, the Netherlands; Division of BioAnalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - Michael K Richardson
- Institute of Biology Leiden, Department of Animal Science and Health, 2333 BE Leiden, the Netherlands
| | - Nicholas R Casewell
- Centre for Snakebite Research & Interventions, Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, 3584 CT Utrecht, the Netherlands; Oncode Institute, Hubrecht Institute, 3584 CT Utrecht, the Netherlands; The Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, the Netherlands.
| |
Collapse
|
28
|
Shirakawa T, Suzuki I. Approach to Neurotoxicity using Human iPSC Neurons: Consortium for Safety Assessment using Human iPS Cells. Curr Pharm Biotechnol 2020; 21:780-786. [DOI: 10.2174/1389201020666191129103730] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/27/2019] [Accepted: 11/03/2019] [Indexed: 01/05/2023]
Abstract
Neurotoxicity, as well as cardiotoxicity and hepatotoxicity, resulting from administration of
a test article is considered a major adverse effect both pre-clinically and clinically. Among the different
types of neurotoxicity occurring during the drug development process, seizure is one of the most serious
one. Seizure occurrence is usually assessed using in vivo animal models, the Functional Observational
Battery, the Irwin test or electroencephalograms. In in vitro studies, a number of assessments can
be performed using animal organs/cells. Interestingly, recent developments in stem cell biology, especially
the development of Human-Induced Pluripotent Stem (iPS) cells, are enabling the assessment of
neurotoxicity in human iPS cell-derived neurons. Further, a Multi-Electrode Array (MEA) using rodent
neurons is a useful tool for identifying seizure-inducing compounds. The Consortium for Safety Assessment
using Human iPS Cells (CSAHi; http://csahi.org/en/) was established in 2013 by the Japan
Pharmaceutical Manufacturers Association (JPMA) to verify the application of human iPS cell-derived
neuronal cells to drug safety evaluation. The Neuro Team of CSAHi has been attempting to evaluate the
seizure risk of compounds using the MEA platform. Here, we review the current status of neurotoxicity
and recent work, including problems related to the use of the MEA assay with human iPS neuronal
cell-derived neurons, and future developments.
Collapse
Affiliation(s)
- Takafumi Shirakawa
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Neuro Team, Japan
| | - Ikuro Suzuki
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Neuro Team, Japan
| |
Collapse
|
29
|
Viallon J, Chinain M, Darius HT. Revisiting the Neuroblastoma Cell-Based Assay (CBA-N2a) for the Improved Detection of Marine Toxins Active on Voltage Gated Sodium Channels (VGSCs). Toxins (Basel) 2020; 12:E281. [PMID: 32349302 PMCID: PMC7290318 DOI: 10.3390/toxins12050281] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
The neuroblastoma cell-based assay (CBA-N2a) is widely used for the detection of marine biotoxins in seafood products, yet a consensus protocol is still lacking. In this study, six key parameters of CBA-N2a were revisited: cell seeding densities, cell layer viability after 26 h growth, MTT incubation time, Ouabain and Veratridine treatment and solvent and matrix effects. A step-by-step protocol was defined identifying five viability controls for the validation of CBA-N2a results. Specific detection of two voltage gated sodium channel activators, pacific ciguatoxin (P-CTX3C) and brevetoxin (PbTx3) and two inhibitors, saxitoxin (STX) and decarbamoylsaxitoxin (dc-STX) was achieved, with EC50 values of 1.7 ± 0.35 pg/mL, 5.8 ± 0.9 ng/mL, 3 ± 0.5 ng/mL and 15.8 ± 3 ng/mL, respectively. When applied to the detection of ciguatoxin (CTX)-like toxicity in fish samples, limit of detection (LOD) and limit of quantification (LOQ) values were 0.031 ± 0.008 and 0.064 ± 0.016 ng P-CTX3C eq/g of flesh, respectively. Intra and inter-assays comparisons of viability controls, LOD, LOQ and toxicity in fish samples gave coefficients of variation (CVs) ranging from 3% to 29%. This improved test adaptable to either high throughput screening or composite toxicity estimation is a useful starting point for a standardization of the CBA-N2a in the field of marine toxin detection.
Collapse
Affiliation(s)
| | | | - Hélène Taiana Darius
- Institut Louis Malardé (ILM), Laboratory of Marine Biotoxins-UMR 241-EIO, 98713 Papeete-Tahiti, French Polynesia; (J.V.); (M.C.)
| |
Collapse
|
30
|
Zwartsen A, Hondebrink L, de Lange DW, Westerink RHS. Hyperthermia exacerbates the acute effects of psychoactive substances on neuronal activity measured using microelectrode arrays (MEAs) in rat primary cortical cultures in vitro. Toxicol Appl Pharmacol 2020; 397:115015. [PMID: 32320794 DOI: 10.1016/j.taap.2020.115015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 11/21/2022]
Abstract
Hyperthermia is a well-known, potentially life-threatening, side effect of stimulant psychoactive substances that worsens the neurological outcome of hospitalized patients. However, current in vitro methods to assess the hazard of psychoactive substances do not account for hyperthermia. Therefore, this study determined the potency of five psychoactive substances (cocaine, MDMA (3,4-methylenedioxymethamphetamine), methamphetamine, 3-MMC (3-methylmethcathinone) and TFMPP (3-trifluoromethylphenylpiperazine)) to affect neuronal activity at physiological and hyperthermic conditions. Neuronal activity of rat cortical cultures grown on microelectrode arrays (MEAs) was recorded at 37 °C before exposure. Following 30 min and 4.5 h drug exposure (1-1000 μM) at 37 °C or 41 °C, neuronal activity was measured at either 37 °C or 41 °C. Without drug exposure, hyperthermia induced a modest decrease in neuronal activity. Following acute (30 min) exposure at 37 °C, all drugs concentration-dependently inhibited neuronal activity. Increasing the temperature to 41 °C significantly exacerbated the reduction of neuronal activity ~ 2-fold for all drugs compared to 37 °C. Prolonged (4.5 h) exposure at 41 °C decreased neuronal activity comparable to 37 °C. Neuronal activity (partly) recovered following drug exposure at both temperatures, although recovery from exposure at 41 °C was less pronounced for most drugs. None of the exposure conditions affected viability. Since acute exposure at hyperthermic conditions exacerbates the decrease in neuronal activity induced by psychoactive substances, effects of hyperthermia should be included in future hazard assessment of illicit drugs and new psychoactive substances (NPS).
Collapse
Affiliation(s)
- Anne Zwartsen
- Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Dutch Poisons Information Center (DPIC), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Laura Hondebrink
- Dutch Poisons Information Center (DPIC), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Dylan W de Lange
- Dutch Poisons Information Center (DPIC), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Remco H S Westerink
- Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
31
|
Tukker AM, Bouwman LMS, van Kleef RGDM, Hendriks HS, Legler J, Westerink RHS. Perfluorooctane sulfonate (PFOS) and perfluorooctanoate (PFOA) acutely affect human α 1β 2γ 2L GABA A receptor and spontaneous neuronal network function in vitro. Sci Rep 2020; 10:5311. [PMID: 32210279 PMCID: PMC7093421 DOI: 10.1038/s41598-020-62152-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/04/2020] [Indexed: 11/28/2022] Open
Abstract
Concerns about the neurotoxic potential of polyfluoroalkyl substances (PFAS) such as perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) increase, although their neurotoxic mechanisms of action remain debated. Considering the importance of the GABAA receptor in neuronal function, we investigated acute effects of PFAS on this receptor and on spontaneous neuronal network activity. PFOS (Lowest Observed Effect Concentration (LOEC) 0.1 µM) and PFOA (LOEC 1 µM) inhibited the GABA-evoked current and acted as non-competitive human GABAA receptor antagonists. Network activity of rat primary cortical cultures increased following exposure to PFOS (LOEC 100 µM). However, exposure of networks of human induced pluripotent stem cell (hiPSC)-derived neurons decreased neuronal activity. The higher sensitivity of the α1β2γ2L GABAA receptor for PFAS as compared to neuronal networks suggests that PFAS have additional mechanisms of action, or that compensatory mechanisms are at play. Differences between rodent and hiPSC-derived neuronal networks highlight the importance of proper model composition. LOECs for PFAS on GABAA receptor and neuronal activity reported here are within or below the range found in blood levels of occupationally exposed humans. For PFOS, LOECs are even within the range found in human serum and plasma of the general population, suggesting a clear neurotoxic risk.
Collapse
Affiliation(s)
- Anke M Tukker
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508, TD, Utrecht, The Netherlands
| | - Lianne M S Bouwman
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508, TD, Utrecht, The Netherlands
| | - Regina G D M van Kleef
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508, TD, Utrecht, The Netherlands
| | - Hester S Hendriks
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508, TD, Utrecht, The Netherlands
| | - Juliette Legler
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508, TD, Utrecht, The Netherlands
| | - Remco H S Westerink
- Neurotoxicology Research Group, Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508, TD, Utrecht, The Netherlands.
| |
Collapse
|
32
|
Kosnik MB, Strickland JD, Marvel SW, Wallis DJ, Wallace K, Richard AM, Reif DM, Shafer TJ. Concentration-response evaluation of ToxCast compounds for multivariate activity patterns of neural network function. Arch Toxicol 2020; 94:469-484. [PMID: 31822930 PMCID: PMC7371233 DOI: 10.1007/s00204-019-02636-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/26/2019] [Indexed: 01/01/2023]
Abstract
The US Environmental Protection Agency's ToxCast program has generated toxicity data for thousands of chemicals but does not adequately assess potential neurotoxicity. Networks of neurons grown on microelectrode arrays (MEAs) offer an efficient approach to screen compounds for neuroactivity and distinguish between compound effects on firing, bursting, and connectivity patterns. Previously, single concentrations of the ToxCast Phase II library were screened for effects on mean firing rate (MFR) in rat primary cortical networks. Here, we expand this approach by retesting 384 of those compounds (including 222 active in the previous screen) in concentration-response across 43 network activity parameters to evaluate neural network function. Using hierarchical clustering and machine learning methods on the full suite of chemical-parameter response data, we identified 15 network activity parameters crucial in characterizing activity of 237 compounds that were response actives ("hits"). Recognized neurotoxic compounds in this network function assay were often more potent compared to other ToxCast assays. Of these chemical-parameter responses, we identified three k-means clusters of chemical-parameter activity (i.e., multivariate MEA response patterns). Next, we evaluated the MEA clusters for enrichment of chemical features using a subset of ToxPrint chemotypes, revealing chemical structural features that distinguished the MEA clusters. Finally, we assessed distribution of neurotoxicants with known pharmacology within the clusters and found that compounds segregated differentially. Collectively, these results demonstrate that multivariate MEA activity patterns can efficiently screen for diverse chemical activities relevant to neurotoxicity, and that response patterns may have predictive value related to chemical structural features.
Collapse
Affiliation(s)
- Marissa B Kosnik
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
- Science for Life Laboratory, Department of Environmental Science and Analytical Chemistry, Stockholm University, Stockholm, Sweden
| | - Jenna D Strickland
- Axion Biosystems, Atlanta, GA, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Skylar W Marvel
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Dylan J Wallis
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Kathleen Wallace
- Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, MD B105-05, Research Triangle Park, NC, 27711, USA
| | - Ann M Richard
- Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, MD B105-05, Research Triangle Park, NC, 27711, USA
| | - David M Reif
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Timothy J Shafer
- Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, MD B105-05, Research Triangle Park, NC, 27711, USA.
| |
Collapse
|
33
|
Zwartsen A, Hondebrink L, Westerink RH. Changes in neuronal activity in rat primary cortical cultures induced by illicit drugs and new psychoactive substances (NPS) following prolonged exposure and washout to mimic human exposure scenarios. Neurotoxicology 2019; 74:28-39. [PMID: 31078573 DOI: 10.1016/j.neuro.2019.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 04/29/2019] [Accepted: 05/07/2019] [Indexed: 12/13/2022]
Abstract
The use of new psychoactive substances (NPS) is increasing despite associated health risks and limited pharmacological and toxicological knowledge. Information is available mainly for acute effects on specific targets like monoamine transporters and receptors. Recently, we have shown the ability of several NPS and illicit drugs to modulate neuronal activity during acute exposure. While these acute measurements provide valuable information regarding the potency and possible structure-activity relationships, an exposure scenario more representative of human exposure would increase insight and aid translation to the human situation. Therefore, we investigated the effects on neuronal activity after acute (30 min) and prolonged (5 h) exposure to amphetamine-type stimulants, cathinones, hallucinogens, piperazines and cocaine using rat primary cortical cultures grown on multi-well microelectrode arrays. To investigate the reversibility of effects, activity was also measured after a washout period of 19 h. During acute exposure, all compounds concentration-dependently decreased neuronal activity. Compared to acute exposure, prolonged exposure did not further decrease neuronal activity. Following washout, effects of 3 out of 11 drugs (methamphetamine, cocaine, and benzylpiperazine) were fully reversible, whereas effects induced by MDMA, PMMA and α-PVP were partially reversible. Neuronal activity did not recover after 19 h washout following exposure to the highest concentration of MDPV, 2C-B, 25B-NBOMe, and TFMPP. On the contrary, exposure to low concentrations of methylone, and to some extent of 2C-B, increased neuronal activity after the washout period. Hazard characterization of emerging NPS should include at least an acute exposure to determine a potency rank order. Supplementing the (acute and prolonged) exposure scenario with a washout period allows investigation of the reversibility of effects. The possibility of a neuronal network to regain activity after drug exposure appears independent of drug class or IC50 values for acute and prolonged exposure. Even though neuronal activity (partly) recovers after washout following exposure to most drugs, it is perturbing that complete recovery of neuronal activity is observed only for a minority of the tested drugs.
Collapse
Affiliation(s)
- Anne Zwartsen
- Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Dutch Poisons Information Center (DPIC), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Laura Hondebrink
- Dutch Poisons Information Center (DPIC), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Remco Hs Westerink
- Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
34
|
Shafer TJ. Application of Microelectrode Array Approaches to Neurotoxicity Testing and Screening. ADVANCES IN NEUROBIOLOGY 2019; 22:275-297. [PMID: 31073941 DOI: 10.1007/978-3-030-11135-9_12] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Neurotoxicity can be defined by the ability of a drug or chemical to alter the physiology, biochemistry, or structure of the nervous system in a manner that may negatively impact the health or function of the individual. Electrophysiological approaches have been utilized to study the mechanisms underlying neurotoxic actions of drugs and chemicals for over 50 years, and in more recent decades, high-throughput patch-clamp approaches have been utilized by the pharmaceutical industry for drug development. The use of microelectrode array recordings to study neural network electrophysiology is a relatively newer approach, with commercially available systems becoming available only in the early 2000s. However, MEAs have been rapidly adopted as a useful approach for neurotoxicity testing. In this chapter, I will review the use of MEA approaches as they have been applied to the field of neurotoxicity testing, especially as they have been applied to the need to screen large numbers of chemicals for neurotoxicity and developmental neurotoxicity. In addition, I will also identify challenges for the field that when addressed will improve the utility of MEA approaches for toxicity testing.
Collapse
Affiliation(s)
- Timothy J Shafer
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory (NHEERL), US EPA, Research Triangle Park, NC, USA.
| |
Collapse
|
35
|
Abstract
'Bursting', defined as periods of high-frequency firing of a neuron separated by periods of quiescence, has been observed in various neuronal systems, both in vitro and in vivo. It has been associated with a range of neuronal processes, including efficient information transfer and the formation of functional networks during development, and has been shown to be sensitive to genetic and pharmacological manipulations. Accurate detection of periods of bursting activity is thus an important aspect of characterising both spontaneous and evoked neuronal network activity. A wide variety of computational methods have been developed to detect periods of bursting in spike trains recorded from neuronal networks. In this chapter, we review several of the most popular and successful of these methods.
Collapse
|
36
|
Black BJ, Atmaramani R, Plagens S, Campbell ZT, Dussor G, Price TJ, Pancrazio JJ. Emerging neurotechnology for antinoceptive mechanisms and therapeutics discovery. Biosens Bioelectron 2018; 126:679-689. [PMID: 30544081 DOI: 10.1016/j.bios.2018.11.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 11/01/2018] [Accepted: 11/10/2018] [Indexed: 12/20/2022]
Abstract
The tolerance, abuse, and potential exacerbation associated with classical chronic pain medications such as opioids creates a need for alternative therapeutics. Phenotypic screening provides a complementary approach to traditional target-based drug discovery. Profiling cellular phenotypes enables quantification of physiologically relevant traits central to a disease pathology without prior identification of a specific drug target. For complex disorders such as chronic pain, which likely involves many molecular targets, this approach may identify novel treatments. Sensory neurons, termed nociceptors, are derived from dorsal root ganglia (DRG) and can undergo changes in membrane excitability during chronic pain. In this review, we describe phenotypic screening paradigms that make use of nociceptor electrophysiology. The purpose of this paper is to review the bioelectrical behavior of DRG neurons, signaling complexity in sensory neurons, various sensory neuron models, assays for bioelectrical behavior, and emerging efforts to leverage microfabrication and microfluidics for assay development. We discuss limitations and advantages of these various approaches and offer perspectives on opportunities for future development.
Collapse
Affiliation(s)
- Bryan J Black
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA.
| | - Rahul Atmaramani
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Sarah Plagens
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Zachary T Campbell
- Department of Biological Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Gregory Dussor
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Joseph J Pancrazio
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| |
Collapse
|
37
|
Bader BM, Jügelt K, Schultz L, Schroeder OHU. Ginkgo biloba L. (Ginkgoaceae) Leaf Extract Medications From Different Providers Exhibit Differential Functional Effects on Mouse Frontal Cortex Neuronal Networks. Front Pharmacol 2018; 9:848. [PMID: 30123130 PMCID: PMC6085676 DOI: 10.3389/fphar.2018.00848] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/13/2018] [Indexed: 12/27/2022] Open
Abstract
Background: Details of the extraction and purification procedure can have a profound impact on the composition of plant-derived extracts, and thus on their efficacy and safety. So far, studies with head-to-head comparison of the pharmacology of Ginkgo extracts rendered by different procedures have been rare. Objective: The objective of this study was to explore whether Ginkgo biloba L. (Ginkgoaceae) leaf extract medications of various sources protect against amyloid beta toxicity on primary mouse cortex neurons growing on microelectrode arrays, and whether the effects differ between different Ginkgo extracts. Design: Our brain-on-chip platform integrates microelectrode array data recorded on neuronal tissue cultures from embryonic mouse cortex. Amyloid beta 42 (Aβ42) and various Ginkgo extract preparations were added to the networks in vitro before evaluation of electrophysiological parameters by multi-parametric analysis. A Multi-variate data analysis, called Effect Score, was designed to compare effects between different products. Results: The results show that Ginkgo extracts protected against Aβ42-induced electrophysiological alterations. Different Ginkgo extracts exhibited different effects. Of note, the reference Ginkgo biloba L. (Ginkgoaceae) leaf medication Tebonin had the most pronounced rescuing effect. Conclusion: Here, we show for the first time a side-by-side analysis of a large number of Ginkgo medications in a relevant in vitro system modeling early functional effects induced by amyloid beta peptides on neuronal transmission and connectivity. Ginkgo biloba L. (Ginkgoaceae) leaf extract from different manufactures exhibit differential functional effects in this neural network model. This in-depth analysis of functional phenotypes of neurons cultured on MEAs chips allows identifying optimal plant extract formulations protecting against toxin-induced functional effects in vitro.
Collapse
|
38
|
Tukker AM, Wijnolts FMJ, de Groot A, Westerink RHS. Human iPSC-derived neuronal models for in vitro neurotoxicity assessment. Neurotoxicology 2018; 67:215-225. [PMID: 29909083 DOI: 10.1016/j.neuro.2018.06.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 04/24/2018] [Accepted: 06/12/2018] [Indexed: 10/28/2022]
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.
Collapse
Affiliation(s)
- Anke M Tukker
- Neurotoxicology Research Group, Toxicology and Pharmacology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 TD, Utrecht, The Netherlands
| | - Fiona M J Wijnolts
- Neurotoxicology Research Group, Toxicology and Pharmacology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 TD, Utrecht, The Netherlands
| | - Aart de Groot
- Neurotoxicology Research Group, Toxicology and Pharmacology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 TD, Utrecht, The Netherlands
| | - Remco H S Westerink
- Neurotoxicology Research Group, Toxicology and Pharmacology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 TD, Utrecht, The Netherlands.
| |
Collapse
|
39
|
Zwartsen A, Hondebrink L, Westerink RH. Neurotoxicity screening of new psychoactive substances (NPS): Effects on neuronal activity in rat cortical cultures using microelectrode arrays (MEA). Neurotoxicology 2018; 66:87-97. [PMID: 29572046 DOI: 10.1016/j.neuro.2018.03.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 03/09/2018] [Accepted: 03/19/2018] [Indexed: 12/13/2022]
Abstract
While the prevalence and the use of new psychoactive substances (NPS) is steadily increasing, data on pharmacological, toxicological and clinical effects is limited. Considering the large number of NPS available, there is a clear need for efficient in vitro screening techniques that capture multiple mechanisms of action. Neuronal cultures grown on multi-well microelectrode arrays (mwMEAs) have previously proven suitable for neurotoxicity screening of chemicals, pharmaceuticals and (illicit) drugs. We therefore used rat primary cortical cultures grown on mwMEA plates to investigate the effects of eight NPS (PMMA, α-PVP, methylone, MDPV, 2C-B, 25B-NBOMe, BZP and TFMPP) and two 'classic' illicit drugs (cocaine, methamphetamine) on spontaneous neuronal activity. All tested drugs rapidly and concentration-dependently decreased the weighted mean firing rate (wMFR) and the weighted mean burst rate (wMBR) during a 30 min acute exposure. Of the 'classic' drugs, cocaine most potently inhibited the wMFR (IC50 9.8 μM), whereas methamphetamine and the structurally-related NPS PMMA were much less potent (IC50 100 μM and IC50 112 μM, respectively). Of the cathinones, MDPV and α-PVP showed comparable IC50 values (29 μM and 21 μM, respectively), although methylone was 10-fold less potent (IC50 235 μM). Comparable 10-fold differences in potency were also observed between the hallucinogenic phenethylamines 2C-B (IC50 27 μM) and 25B-NBOMe (IC50 2.4 μM), and between the piperazine derivatives BZP (IC50 161 μM) and TFMPP (IC50 19 μM). All drugs also inhibited the wMBR and concentration-response curves for wMBR and wMFR were comparable. For most drugs, IC50 values are close to the estimated human brain concentrations following recreational doses of these drugs, highlighting the importance of this efficient in vitro screening approach for classification and prioritization of emerging NPS. Moreover, the wide range of IC50 values observed for these and previously tested drugs of abuse, both within and between different classes of NPS, indicates that additional investigation of structure-activity relationships could aid future risk assessment of emerging NPS.
Collapse
Affiliation(s)
- Anne Zwartsen
- Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands; Dutch Poisons Information Center (DPIC), University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Laura Hondebrink
- Dutch Poisons Information Center (DPIC), University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Remco Hs Westerink
- Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
40
|
Strickland JD, Martin MT, Richard AM, Houck KA, Shafer TJ. Screening the ToxCast phase II libraries for alterations in network function using cortical neurons grown on multi-well microelectrode array (mwMEA) plates. Arch Toxicol 2018; 92:487-500. [PMID: 28766123 PMCID: PMC6438628 DOI: 10.1007/s00204-017-2035-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 07/12/2017] [Indexed: 12/12/2022]
Abstract
Methods are needed for rapid screening of environmental compounds for neurotoxicity, particularly ones that assess function. To demonstrate the utility of microelectrode array (MEA)-based approaches as a rapid neurotoxicity screening tool, 1055 chemicals from EPA's phase II ToxCast library were evaluated for effects on neural function and cell health. Primary cortical networks were grown on multi-well microelectrode array (mwMEA) plates. On day in vitro 13, baseline activity (40 min) was recorded prior to exposure to each compound (40 µM). Changes in spontaneous network activity [mean firing rate (MFR)] and cell viability (lactate dehydrogenase and CellTiter Blue) were assessed within the same well following compound exposure. Following exposure, 326 compounds altered (increased or decreased) normalized MFR beyond hit thresholds based on 2× the median absolute deviation of DMSO-treated wells. Pharmaceuticals, pesticides, fungicides, chemical intermediates, and herbicides accounted for 86% of the hits. Further, changes in MFR occurred in the absence of cytotoxicity, as only eight compounds decreased cell viability. ToxPrint chemotype analysis identified several structural domains (e.g., biphenyls and alkyl phenols) significantly enriched with MEA actives relative to the total test set. The top 5 enriched ToxPrint chemotypes were represented in 26% of the MEA hits, whereas the top 11 ToxPrints were represented in 34% of MEA hits. These results demonstrate that large-scale functional screening using neural networks on MEAs can fill a critical gap in assessment of neurotoxicity potential in ToxCast assay results. Further, a data-mining approach identified ToxPrint chemotypes enriched in the MEA-hit subset, which define initial structure-activity relationship inferences, establish potential mechanistic associations to other ToxCast assay endpoints, and provide working hypotheses for future studies.
Collapse
Affiliation(s)
- Jenna D Strickland
- Axion Biosystems, Atlanta, GA, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Matthew T Martin
- National Center for Computational Toxicology, U.S. Environmental Protection Agency, MD D143-02, Research Triangle Park, NC, 27711, USA
- Pfizer Inc, Groton, CT, USA
| | - Ann M Richard
- National Center for Computational Toxicology, U.S. Environmental Protection Agency, MD D143-02, Research Triangle Park, NC, 27711, USA
| | - Keith A Houck
- National Center for Computational Toxicology, U.S. Environmental Protection Agency, MD D143-02, Research Triangle Park, NC, 27711, USA
| | - Timothy J Shafer
- Integrated Systems Toxicology Division, U.S. Environmental Protection Agency, MD105-05, Research Triangle Park, NC, 27711, USA.
| |
Collapse
|
41
|
Targeted Stimulation Using Differences in Activation Probability across the Strength–Duration Space. Processes (Basel) 2017. [DOI: 10.3390/pr5020014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
42
|
Duarte DJ, Rutten JM, van den Berg M, Westerink RH. In vitro neurotoxic hazard characterization of different tricresyl phosphate (TCP) isomers and mixtures. Neurotoxicology 2017; 59:222-230. [DOI: 10.1016/j.neuro.2016.02.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 02/01/2016] [Accepted: 02/01/2016] [Indexed: 12/01/2022]
|
43
|
Kasteel EEJ, Westerink RHS. Comparison of the acute inhibitory effects of Tetrodotoxin (TTX) in rat and human neuronal networks for risk assessment purposes. Toxicol Lett 2017; 270:12-16. [PMID: 28192153 DOI: 10.1016/j.toxlet.2017.02.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 02/07/2017] [Accepted: 02/08/2017] [Indexed: 11/29/2022]
Abstract
Tetrodotoxin (TTX) is an extremely toxic marine neurotoxin. TTX inhibits voltage-gated sodium channels, resulting in a potentially lethal inhibition of neurotransmission. Despite numerous intoxications in Asia and Europe, limited (human) toxicological data are available for TTX. Additionally, the degree of interspecies differences for TTX is not well established, hampering the use of available (animal) data for human risk assessment and establishing regulatory limits for TTX concentrations in (shell)fish. We therefore used micro-electrode array (MEA) recordings as an integrated measure of neurotransmission to demonstrate that TTX inhibits neuronal electrical activity in both primary rat cortical cultures and human-induced pluripotent stem cell (hIPSC)-derived iCell® neurons in co-culture with hIPSC-derived iCell® astrocytes, with IC50 values of 7 and 10nM, respectively. From these data combined with LD50 values and IC50 concentrations of voltage-gated sodium channels derived from literature it can be concluded that interspecies differences are limited for TTX. Consequently, we used experimental animal data to derive a human acute reference dose of 1.33μg/kg body weight, which corresponds to maximum concentration of TTX in shellfish of 200μg/kg.
Collapse
Affiliation(s)
- Emma E J Kasteel
- Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 TD, Utrecht, The Netherlands
| | - Remco H S Westerink
- Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 TD, Utrecht, The Netherlands.
| |
Collapse
|
44
|
Marine Toxins Analysis for Consumer Protection. RECENT ADVANCES IN THE ANALYSIS OF MARINE TOXINS 2017. [DOI: 10.1016/bs.coac.2017.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
45
|
Brown JP, Lynch BS, Curry-Chisolm IM, Shafer TJ, Strickland JD. Assaying Spontaneous Network Activity and Cellular Viability Using Multi-well Microelectrode Arrays. Methods Mol Biol 2017; 1601:153-170. [PMID: 28470525 DOI: 10.1007/978-1-4939-6960-9_13] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Microelectrode array (MEA) technology is a neurophysiological method that allows for the spontaneous measure of activity in neural cultures and determination of drug and chemical effects thereon. Recent introduction of multi-well MEA (mwMEA) formats have dramatically increased the throughput of this technology, allowing more efficient compound screening. Rapid characterization of compounds for neuroactivity or neurotoxicity hazard evaluation following acute, chronic, or developmental exposures ideally would also consider compound effects on cell health, and to do so in the same well requires a multiplexed approach. Procedures describing the multiplexed method to acute and developmental screening are described in this chapter.
Collapse
Affiliation(s)
- Jasmine P Brown
- Integrated Systems Toxicology Division, NHEERL, US EPA, MD105-05, Research Triangle Park, NC, 27711, USA
| | - Brittany S Lynch
- Integrated Systems Toxicology Division, NHEERL, US EPA, MD105-05, Research Triangle Park, NC, 27711, USA
| | - Itaevia M Curry-Chisolm
- Integrated Systems Toxicology Division, NHEERL, US EPA, MD105-05, Research Triangle Park, NC, 27711, USA
| | - Timothy J Shafer
- Integrated Systems Toxicology Division, NHEERL, US EPA, MD105-05, Research Triangle Park, NC, 27711, USA.
| | - Jenna D Strickland
- Axion Biosystems, Atlanta, GA, USA
- Department of Pharmacology and Toxicology, Michigan State University, E. Lansing, MI, USA
| |
Collapse
|
46
|
Dingemans MM, Schütte MG, Wiersma DM, de Groot A, van Kleef RG, Wijnolts FM, Westerink RH. Chronic 14-day exposure to insecticides or methylmercury modulates neuronal activity in primary rat cortical cultures. Neurotoxicology 2016; 57:194-202. [DOI: 10.1016/j.neuro.2016.10.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/29/2016] [Accepted: 10/04/2016] [Indexed: 10/20/2022]
|
47
|
Brown JP, Hall D, Frank CL, Wallace K, Mundy WR, Shafer TJ. Editor's Highlight: Evaluation of a Microelectrode Array-Based Assay for Neural Network Ontogeny Using Training Set Chemicals. Toxicol Sci 2016; 154:126-139. [PMID: 27492221 DOI: 10.1093/toxsci/kfw147] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Thousands of compounds in the environment have not been characterized for developmental neurotoxicity (DNT) hazard. To address this issue, methods to screen compounds rapidly for DNT hazard evaluation are necessary and are being developed for key neurodevelopmental processes. In order to develop an assay for network formation, this study evaluated effects of a training set of chemicals on network ontogeny by measuring spontaneous electrical activity in neural networks grown on microelectrode arrays (MEAs). Rat (0-24 h old) primary cortical cells were plated in 48 well-MEA plates and exposed to 6 compounds: acetaminophen, bisindolylmaleimide-1 (Bis-1), domoic acid, mevastatin, sodium orthovanadate, and loperamide for a period of 12 days. Spontaneous network activity was recorded on days 2, 5, 7, 9, and 12 and viability was assessed using the Cell Titer Blue assay on day 12. Network activity (e.g. mean firing rate [MFR], burst rate [BR], etc), increased between days 5 and 12. Random Forest analysis indicated that across all compounds and times, temporal correlation of firing patterns (r), MFR, BR, number of active electrodes and % of spikes in a burst were the most influential parameters in separating control from treated wells. All compounds except acetaminophen (≤ 30 µM) caused concentration-related effects on one or more of these parameters. Domoic acid and sodium orthovanadate altered several of these parameters in the absence of cytotoxicity. Although cytotoxicity was observed with Bis1, mevastatin, and loperamide, some parameters were affected by these compounds at concentrations below those resulting in cytotoxicity. These results demonstrate that this assay may be suitable for screening of compounds for DNT hazard identification.
Collapse
Affiliation(s)
| | - Diana Hall
- NHEERL, US EPA, Research Triangle Park, NC, USA
| | | | | | | | | |
Collapse
|
48
|
Cotterill E, Hall D, Wallace K, Mundy WR, Eglen SJ, Shafer TJ. Characterization of Early Cortical Neural Network Development in Multiwell Microelectrode Array Plates. JOURNAL OF BIOMOLECULAR SCREENING 2016; 21:510-9. [PMID: 27028607 PMCID: PMC4904353 DOI: 10.1177/1087057116640520] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 03/01/2016] [Accepted: 03/02/2016] [Indexed: 11/16/2022]
Abstract
We examined neural network ontogeny using microelectrode array (MEA) recordings made in multiwell MEA (mwMEA) plates over the first 12 days in vitro (DIV). In primary cortical cultures, action potential spiking activity developed rapidly between DIV 5 and 12. Spiking was sporadic and unorganized at early DIV, and became progressively more organized with time, with bursting parameters, synchrony, and network bursting increasing between DIV 5 and 12. We selected 12 features to describe network activity; principal components analysis using these features demonstrated segregation of data by age at both the well and plate levels. Using random forest classifiers and support vector machines, we demonstrated that four features (coefficient of variation [CV] of within-burst interspike interval, CV of interburst interval, network spike rate, and burst rate) could predict the age of each well recording with >65% accuracy. When restricting the classification to a binary decision, accuracy improved to as high as 95%. Further, we present a novel resampling approach to determine the number of wells needed for comparing different treatments. Overall, these results demonstrate that network development on mwMEA plates is similar to development in single-well MEAs. The increased throughput of mwMEAs will facilitate screening drugs, chemicals, or disease states for effects on neurodevelopment.
Collapse
Affiliation(s)
- Ellese Cotterill
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, UK
| | - Diana Hall
- Office of Research and Development, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Kathleen Wallace
- Office of Research and Development, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - William R Mundy
- Office of Research and Development, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Stephen J Eglen
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, UK
| | - Timothy J Shafer
- Office of Research and Development, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| |
Collapse
|
49
|
Hondebrink L, Verboven AHA, Drega WS, Schmeink S, de Groot MWGDM, van Kleef RGDM, Wijnolts FMJ, de Groot A, Meulenbelt J, Westerink RHS. Neurotoxicity screening of (illicit) drugs using novel methods for analysis of microelectrode array (MEA) recordings. Neurotoxicology 2016; 55:1-9. [PMID: 27149913 DOI: 10.1016/j.neuro.2016.04.020] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/18/2016] [Accepted: 04/29/2016] [Indexed: 01/11/2023]
Abstract
Annual prevalence of the use of common illicit drugs and new psychoactive substances (NPS) is high, despite the often limited knowledge on the health risks of these substances. Recently, cortical cultures grown on multi-well microelectrode arrays (mwMEAs) have been used for neurotoxicity screening of chemicals, pharmaceuticals, and toxins with a high sensitivity and specificity. However, the use of mwMEAs to investigate the effects of illicit drugs on neuronal activity is largely unexplored. We therefore first characterised the cortical cultures using immunocytochemistry and show the presence of astrocytes, glutamatergic and GABAergic neurons. Neuronal activity is concentration-dependently affected following exposure to six neurotransmitters (glutamate, GABA, serotonin, dopamine, acetylcholine and nicotine). Most neurotransmitters inhibit neuronal activity, although glutamate and acetylcholine transiently increase activity at specific concentrations. These transient effects are not detected when activity is determined during the entire 30min exposure window, potentially resulting in false-negative results. As expected, exposure to the GABAA-receptor antagonist bicuculline increases neuronal activity. Exposure to a positive allosteric modulator of the GABAA-receptor (diazepam) or to glutamate receptor antagonists (CNQX and MK-801) reduces neuronal activity. Further, we demonstrate that exposure to common drugs (3,4-methylenedioxymethamphetamine (MDMA) and amphetamine) and NPS (1-(3-chlorophenyl)piperazine (mCPP), 4-fluoroamphetamine (4-FA) and methoxetamine (MXE)) decreases neuronal activity. MXE most potently inhibits neuronal activity with an IC50 of 0.5μM, whereas 4-FA is least potent with an IC50 of 113μM. Our data demonstrate the importance of analysing neuronal activity within different time windows during exposure to prevent false-negative results. We also show that cortical cultures grown on mwMEAs can successfully be applied to investigate the effects of different (illicit) drugs on neuronal activity. Compared to investigating multiple single endpoints for neurotoxicity or neuromodulation, such as receptor activation or calcium channel function, mwMEAs can provide information on integrated aspects of drug-induced neurotoxicity more rapidly. Therefore, this approach could contribute to a faster insight in possible health risks and shorten the regulation process.
Collapse
Affiliation(s)
- L Hondebrink
- National Poisons Information Center (NVIC), University Medical Center Utrecht, P.O. Box 85500, NL-3508 GA Utrecht, The Netherlands.
| | - A H A Verboven
- Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 Utrecht, The Netherlands
| | - W S Drega
- Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 Utrecht, The Netherlands
| | - S Schmeink
- Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 Utrecht, The Netherlands
| | - M W G D M de Groot
- Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 Utrecht, The Netherlands
| | - R G D M van Kleef
- Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 Utrecht, The Netherlands
| | - F M J Wijnolts
- Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 Utrecht, The Netherlands
| | - A de Groot
- Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 Utrecht, The Netherlands
| | - J Meulenbelt
- National Poisons Information Center (NVIC), University Medical Center Utrecht, P.O. Box 85500, NL-3508 GA Utrecht, The Netherlands; Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 Utrecht, The Netherlands; Department of Intensive Care Medicine, University Medical Center Utrecht, P.O. Box 85500, NL-3508 GA, Utrecht, The Netherlands
| | - R H S Westerink
- Neurotoxicology Research Group, Division Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.177, NL-3508 Utrecht, The Netherlands
| |
Collapse
|
50
|
Alloisio S, Giussani V, Nobile M, Chiantore M, Novellino A. Microelectrode array (MEA) platform as a sensitive tool to detect and evaluate Ostreopsis cf. ovata toxicity. HARMFUL ALGAE 2016; 55:230-237. [PMID: 28073536 DOI: 10.1016/j.hal.2016.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 03/04/2016] [Accepted: 03/04/2016] [Indexed: 06/06/2023]
Abstract
In the last decade, the occurrence of harmful dinoflagellate blooms of the genus Ostreopsis has increased both in frequency and in geographic distribution with adverse impacts on public health and the economy. Ostreopsis species are producers of palytoxin-like toxins (putative palytoxin and ovatoxins) which are among the most potent natural non-protein compounds known to date, exhibiting extreme toxicity in mammals, including humans. Most existing toxicological data are derived from in vivo mouse assay and are related to acute effects of pure palytoxin, without considering that the toxicity mechanism of dinoflagellates can be dependent on the varying composition of complex biotoxins mixture and on the presence of cellular components. In this study, in vitro neuronal networks coupled to microelectrode array (MEA)-based system are proposed, for the first time, as sensitive biosensors for the evaluation of marine alga toxicity on mammalian cells. Toxic effect was investigated by testing three different treatments of laboratory cultured Ostreopsis cf. ovata cells: filtered and re-suspended algal cells; filtered, re-suspended and sonicated algal cells; conditioned growth medium devoid of algal cells. The great sensitivity of this system revealed the mixture of PTLX-complex analogues naturally released in the growth medium and the different potency of the three treatments to inhibit the neuronal network spontaneous electrical activity. Moreover, by means of the multiparametric analysis of neuronal network activity, the approach revealed a different toxicity mechanism of the cellular component compared to the algal conditioned growth medium, highlighting the potential active role of the first treatment.
Collapse
Affiliation(s)
- Susanna Alloisio
- ETT S.p.A., via Sestri 37, Genoa 16154, Italy; CNR-Institute of Biophysics (IBF), Via De Marini 6, 16149 Genoa, Italy.
| | | | - Mario Nobile
- CNR-Institute of Biophysics (IBF), Via De Marini 6, 16149 Genoa, Italy
| | | | - Antonio Novellino
- ETT S.p.A., via Sestri 37, Genoa 16154, Italy; CNR-Institute of Biophysics (IBF), Via De Marini 6, 16149 Genoa, Italy
| |
Collapse
|