1
|
Choi MS, Park SM, Kim S, Jegal H, Lee HA, Han HY, Yoon S, Kim SK, Oh JH. Enhanced electrophysiological activity and neurotoxicity screening of environmental chemicals using 3D neurons from human neural precursor cells purified with PSA-NCAM. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 280:116516. [PMID: 38820819 DOI: 10.1016/j.ecoenv.2024.116516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/02/2024]
Abstract
The assessment of neurotoxicity for environmental chemicals is of utmost importance in ensuring public health and environmental safety. Multielectrode array (MEA) technology has emerged as a powerful tool for assessing disturbances in the electrophysiological activity. Although human embryonic stem cell (hESC)-derived neurons have been used in MEA for neurotoxicity screening, obtaining a substantial and sufficiently active population of neurons from hESCs remains challenging. In this study, we successfully differentiated neurons from a large population of human neuronal precursor cells (hNPC) purified using a polysialylated neural cell adhesion molecule (PSA-NCAM), referred to as hNPCPSA-NCAM+. The functional characterization demonstrated that hNPCPSA-NCAM+-derived neurons improve functionality by enhancing electrophysiological activity compared to total hNPC-derived neurons. Furthermore, three-dimensional (3D) neurons derived from hNPCPSA-NCAM+ exhibited reduced maturation time and enhanced electrophysiological activity on MEA. We employed subdivided population analysis of active mean firing rate (MFR) based on electrophysiological intensity to characterize the electrophysiological properties of hNPCPSA-NCAM+-3D neurons. Based on electrophysiological activity including MFR and burst parameters, we evaluated the sensitivity of hNPCPSA-NCAM+-3D neurons on MEA to screen both inhibitory and excitatory neuroactive environmental chemicals. Intriguingly, electrophysiologically active hNPCPSA-NCAM+-3D neurons demonstrated good sensitivity to evaluate neuroactive chemicals, particularly in discriminating excitatory chemicals. Our findings highlight the effectiveness of MEA approaches using hNPCPSA-NCAM+-3D neurons in the assessment of neurotoxicity associated with environmental chemicals. Furthermore, we emphasize the importance of selecting appropriate signal intensity thresholds to enhance neurotoxicity prediction and screening of environmental chemicals.
Collapse
Affiliation(s)
- Mi-Sun Choi
- Department of predictive toxicology, Korea Institute of Toxicology (KIT), Daejeon, the Republic of Korea; College of Pharmacy, Chungnam National University, Daejeon, the Republic of Korea
| | - Se-Myo Park
- Department of predictive toxicology, Korea Institute of Toxicology (KIT), Daejeon, the Republic of Korea
| | - Soojin Kim
- Department of predictive toxicology, Korea Institute of Toxicology (KIT), Daejeon, the Republic of Korea
| | - Hyun Jegal
- Department of predictive toxicology, Korea Institute of Toxicology (KIT), Daejeon, the Republic of Korea; Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, the Republic of Korea
| | - Hyang-Ae Lee
- Department of predictive toxicology, Korea Institute of Toxicology (KIT), Daejeon, the Republic of Korea
| | - Hyoung-Yun Han
- Department of predictive toxicology, Korea Institute of Toxicology (KIT), Daejeon, the Republic of Korea; Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, the Republic of Korea
| | - Seokjoo Yoon
- Department of predictive toxicology, Korea Institute of Toxicology (KIT), Daejeon, the Republic of Korea; Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, the Republic of Korea
| | - Sang-Kyum Kim
- College of Pharmacy, Chungnam National University, Daejeon, the Republic of Korea.
| | - Jung-Hwa Oh
- Department of predictive toxicology, Korea Institute of Toxicology (KIT), Daejeon, the Republic of Korea; Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, the Republic of Korea.
| |
Collapse
|
2
|
Kanungo J, Sorkin BC, Krzykwa J, Mitchell CA, Embry M, Spencer P, Harry GJ, Cannon J, Liu F, McPherson CA, Gafner S, Westerink RH. Screening tools to evaluate the neurotoxic potential of botanicals: building a strategy to assess safety. Expert Opin Drug Metab Toxicol 2024; 20:629-646. [PMID: 38984683 PMCID: PMC11542175 DOI: 10.1080/17425255.2024.2378895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/08/2024] [Indexed: 07/11/2024]
Abstract
AREAS COVERED This paper outlines the selection of NAMs, including in vitro assays using primary rat cortical neurons, zebrafish embryos, and Caenorhabditis elegans. These assays aim to assess neurotoxic endpoints such as neuronal activity and behavioral responses. Microelectrode array recordings of rat cortical neurons provide insights into the impact of botanical extracts on neuronal function, while the zebrafish embryos and C. elegans assays evaluate neurobehavioral responses. The paper also provides an account of the selection of botanical case studies based on expert judgment and existing neuroactivity/toxicity information. The proposed battery of assays will be tested with these case studies to evaluate their utility for neurotoxicity screening. EXPERT OPINION The complexity of botanicals necessitates the use of multiple NAMs for effective neurotoxicity screening. This paper discusses the evaluation of methodologies to develop a robust framework for evaluating botanical safety, including complex neuronal models and key neurodevelopmental process assays. It aims to establish a comprehensive screening framework.
Collapse
Affiliation(s)
- Jyotshna Kanungo
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079
| | - Barbara C. Sorkin
- Office of Dietary Supplements, Division of Program Coordination, Planning, and Strategic Initiatives, U.S. National Institutes of Health, Bethesda, MD
| | - Julie Krzykwa
- Health and Environmental Sciences Institute, Washington, DC, USA
| | | | - Michelle Embry
- Health and Environmental Sciences Institute, Washington, DC, USA
| | - Peter Spencer
- Department of Neurology, School of Medicine, Oregon Health & Science University
| | - G. Jean Harry
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Jason Cannon
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA
| | - Fang Liu
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079
| | - Christopher A. McPherson
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Stefan Gafner
- American Botanical Council, 6200 Manor Road, Austin, Texas 78723, United States
| | - Remco H.S. Westerink
- Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
3
|
Torres-Ruiz M, de Alba Gonzalez M, Cañas Portilla AI, Coronel R, Liste I, González-Caballero MC. Effects of nanomolar methylmercury on developing human neural stem cells and zebrafish Embryo. Food Chem Toxicol 2024; 188:114684. [PMID: 38663761 DOI: 10.1016/j.fct.2024.114684] [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: 02/27/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
Exposure to mercury and its organic form methylmercury (MeHg), is of great concern for the developing nervous system. Despite available literature on MeHg neurotoxicity, there is still uncertainty about its mechanisms of action and the doses that trigger developmental effects. Our study combines two alternative methodologies, the human neural stem cells (NSC) and the zebrafish (ZF) embryo, to address the neurotoxic effects of early exposure to nanomolar concentrations of MeHg. Our results show linear or nonmonotonic (hormetic) responses depending on studied parameters. In ZF, we observed a hormetic response in locomotion and larval rotation, but a concentration-dependent response for sensory organ size and habituation. We also observed a possible delayed response as MeHg had greater effects on larval activity at 5 days than at 24 h. In NSC cells, some parameters show a clear dose dependence, such as increased apoptosis and differentiation to glial cells or decreased neuronal precursors; while others show a hormetic response: neuronal differentiation or cell proliferation. This study shows that the ZF model was more susceptible than NSC to MeHg neurotoxicity. The combination of different models has improved the understanding of the underlying mechanisms of toxicity and possible compensatory mechanisms at the cellular and organismal level.
Collapse
Affiliation(s)
- Mónica Torres-Ruiz
- Environmental Toxicology Unit, Centro Nacional de Sanidad Ambiental (CNSA), Instituto de Salud Carlos III (ISCIII), Ctra. Majadahonda-Pozuelo Km. 2,2., Majadahonda, Madrid, 28220, Spain.
| | - Mercedes de Alba Gonzalez
- Environmental Toxicology Unit, Centro Nacional de Sanidad Ambiental (CNSA), Instituto de Salud Carlos III (ISCIII), Ctra. Majadahonda-Pozuelo Km. 2,2., Majadahonda, Madrid, 28220, Spain
| | - Ana I Cañas Portilla
- Environmental Toxicology Unit, Centro Nacional de Sanidad Ambiental (CNSA), Instituto de Salud Carlos III (ISCIII), Ctra. Majadahonda-Pozuelo Km. 2,2., Majadahonda, Madrid, 28220, Spain
| | - Raquel Coronel
- Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III, Madrid, Spain; Departamento de Biología de Sistemas, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá, Madrid, Spain
| | - Isabel Liste
- Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Mª Carmen González-Caballero
- Environmental Toxicology Unit, Centro Nacional de Sanidad Ambiental (CNSA), Instituto de Salud Carlos III (ISCIII), Ctra. Majadahonda-Pozuelo Km. 2,2., Majadahonda, Madrid, 28220, Spain.
| |
Collapse
|
4
|
van Kleef RGDM, Embry MR, Mitchell CA, Westerink RHS. Neuroactivity screening of botanical extracts using microelectrode array (MEA) recordings. Food Chem Toxicol 2024; 184:114438. [PMID: 38191119 DOI: 10.1016/j.fct.2024.114438] [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: 10/17/2023] [Revised: 12/19/2023] [Accepted: 01/04/2024] [Indexed: 01/10/2024]
Abstract
Toxicity testing of botanicals is challenging because of their chemical complexity and variability. Since botanicals may affect many different modes of action involved in neuronal function, we used microelectrode array (MEA) recordings of primary rat cortical cultures to screen 16 different botanical extracts for their effects on cell viability and neuronal network function in vitro. Our results demonstrate that extract materials (50 μg/mL) derived from goldenseal, milk thistle, tripterygium, and yohimbe decrease mitochondrial activity following 7 days exposure, indicative of cytotoxicity. Importantly, most botanical extracts alter neuronal network function following acute exposure. Extract materials (50 μg/mL) derived from aristolochia, ephedra, green tea, milk thistle, tripterygium, and usnea inhibit neuronal activity. Extracts of kava, kratom and yohimbe are particularly potent and induce a profound inhibition of neuronal activity at the low dose of 5 μg/mL. Extracts of blue cohosh, goldenseal and oleander cause intensification of the bursts. Aconite extract (5 μg/mL) evokes a clear hyperexcitation with a marked increase in the number of spikes and (network) bursts. The distinct activity patterns suggest that botanical extracts have diverse modes of action. Our combined data also highlight the applicability of MEA recordings for hazard identification and potency ranking of botanicals.
Collapse
Affiliation(s)
- 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.
| | - Michelle R Embry
- Health and Environmental Sciences Institute, Washington, DC, USA.
| | | | - 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
|
5
|
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
|
6
|
van Melis LVJ, Heusinkveld HJ, Langendoen C, Peters A, Westerink RHS. Organophosphate insecticides disturb neuronal network development and function via non-AChE mediated mechanisms. Neurotoxicology 2023; 94:35-45. [PMID: 36347328 DOI: 10.1016/j.neuro.2022.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/06/2022]
Abstract
Exposure to organophosphate (OP) insecticides has been related to several adverse health effects, including neurotoxicity. The primary insecticidal mode of action of OP insecticides relies on (irreversible) binding to acetylcholine esterase (AChE), with -oxon metabolites having a much higher potency for AChE inhibition than the parent compounds. However, OP insecticides can also have non-AChE-mediated effects, including changes in gene expression, neuroendocrine effects, disruption of neurite outgrowth and disturbance of the intracellular calcium (Ca2+) homeostasis. Since Ca2+ is involved in neurotransmission and neuronal development, our research aimed to assess the effects of two widely used OP insecticides, chlorpyrifos (CPF) and diazinon (DZ) and their respective -oxon metabolites, on intracellular Ca2+ homeostasis in human SH-SY5Y cells and rat primary cortical cultures. Furthermore, we assessed the acute and chronic effects of exposure to these compounds on neuronal network maturation and function in rat primary cortical cultures using microelectrode array (MEA) recordings. While inhibition of AChE appears to be the primary mode of action of oxon-metabolites, our data indicate that both parent OP insecticides (CPF and DZ) inhibit depolarization-evoked Ca2+ influx and neuronal activity at concentrations far below their sensitivity for AChE inhibition, indicating that inhibition of voltage-gated calcium channels is a common mode of action of OP insecticides. Notably, parent compounds were more potent than their oxon metabolites, with exposure to diazinon-oxon (DZO) having no effect on both neuronal activity and Ca2+ influx. Human SH-SY5Y cells were more sensitive to OP-induced inhibition of depolarization-evoked Ca2+ influx than rat cortical cells. Acute exposure to OP insecticides had more potent effects on neuronal activity than on Ca2+ influx, suggesting that neuronal activity parameters are especially sensitive to OP exposure. Interestingly, the effects of DZ and chlorpyrifos-oxon (CPO) on neuronal activity lessened after 48 h of exposure, while the potency of CPF did not differ over time. This suggests that neurotoxicity after exposure to different OPs has different effects over time and occurs at levels that are close to human exposure levels. In line with these results, chronic exposure to CPF during 10 days impaired neuronal network development, illustrating the need to investigate possible links between early-life OP exposure and neurodevelopmental disorders in children and highlighting the importance of non-AChE mediated mechanisms of neurotoxicity after OP exposure.
Collapse
Affiliation(s)
- Lennart V J van Melis
- 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
| | - Harm J Heusinkveld
- 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; Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Celine Langendoen
- 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
| | - Anouk Peters
- 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
| | - 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
|
7
|
Kreir M, Floren W, Policarpo R, De Bondt A, Van den Wyngaert I, Teisman A, Gallacher DJ, Lu HR. Is the forming of neuronal network activity in human-induced pluripotent stem cells important for the detection of drug-induced seizure risks? Eur J Pharmacol 2022; 931:175189. [PMID: 35987255 DOI: 10.1016/j.ejphar.2022.175189] [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: 03/08/2022] [Revised: 08/03/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Functional network activity is a characteristic for neuronal cells, and the complexity of the network activity represents the necessary substrate to support complex brain functions. Drugs that drastically increase the neuronal network activity may have a potential higher risk for seizures in human. Although there has been some recent considerable progress made using cultures from different types of human-induced pluripotent stem cell (hiPSC) derived neurons, one of the primary limitations is the lack of - or very low - network activity. METHOD In the present study, we investigated whether the limited neuronal network activity in commercial hiPSC-neurons (CNS.4U®) is capable of detecting drug-induced potential seizure risks. Therefore, we compared the hiPSC-results to those in rat primary neurons with known high neuronal network activity in vitro. RESULTS Gene expression and electrical activity from in vitro developing neuronal networks were assessed at multiple time-points. Transcriptomes of 7, 28, and 50 days in vitro were analyzed and compared to those from human brain tissues. Data from measurements of electrical activity using multielectrode arrays (MEAs) indicate that neuronal networks matured gradually over time, albeit in hiPSC this developed slower than rat primary cultures. The response of neuronal networks to neuronal active reference drugs modulating glutamatergic, acetylcholinergic and GABAergic pathways could be detected in both hiPSC-neurons and rat primary neurons. However, in comparison, GABAergic responses were limited in hiPSC-neurons. CONCLUSION Overall, despite a slower network development and lower network activity, CNS.4U® hiPSC-neurons can be used to detect drug induced changes in neuronal network activity, as shown by well-known seizurogenic drugs (affecting e.g., the Glycine receptor and Na+ channel). However, lower sensitivity to GABA antagonists has been observed.
Collapse
Affiliation(s)
- Mohamed Kreir
- Global Safety Pharmacology, Predictive & Investigative Translational Toxicology, Nonclinical Safety, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium.
| | - Wim Floren
- Global Safety Pharmacology, Predictive & Investigative Translational Toxicology, Nonclinical Safety, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Rafaela Policarpo
- Neuroscience Therapeutic Area, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Belgium
| | - An De Bondt
- High Dimensional & Computational Biology, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Ilse Van den Wyngaert
- High Dimensional & Computational Biology, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Ard Teisman
- Global Safety Pharmacology, Predictive & Investigative Translational Toxicology, Nonclinical Safety, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - David J Gallacher
- Global Safety Pharmacology, Predictive & Investigative Translational Toxicology, Nonclinical Safety, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Hua Rong Lu
- Global Safety Pharmacology, Predictive & Investigative Translational Toxicology, Nonclinical Safety, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| |
Collapse
|
8
|
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]
|
9
|
Crofton KM, Bassan A, Behl M, Chushak YG, Fritsche E, Gearhart JM, Marty MS, Mumtaz M, Pavan M, Ruiz P, Sachana M, Selvam R, Shafer TJ, Stavitskaya L, Szabo DT, Szabo ST, Tice RR, Wilson D, Woolley D, Myatt GJ. Current status and future directions for a neurotoxicity hazard assessment framework that integrates in silico approaches. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 22:100223. [PMID: 35844258 PMCID: PMC9281386 DOI: 10.1016/j.comtox.2022.100223] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
Neurotoxicology is the study of adverse effects on the structure or function of the developing or mature adult nervous system following exposure to chemical, biological, or physical agents. The development of more informative alternative methods to assess developmental (DNT) and adult (NT) neurotoxicity induced by xenobiotics is critically needed. The use of such alternative methods including in silico approaches that predict DNT or NT from chemical structure (e.g., statistical-based and expert rule-based systems) is ideally based on a comprehensive understanding of the relevant biological mechanisms. This paper discusses known mechanisms alongside the current state of the art in DNT/NT testing. In silico approaches available today that support the assessment of neurotoxicity based on knowledge of chemical structure are reviewed, and a conceptual framework for the integration of in silico methods with experimental information is presented. Establishing this framework is essential for the development of protocols, namely standardized approaches, to ensure that assessments of NT and DNT based on chemical structures are generated in a transparent, consistent, and defendable manner.
Collapse
Affiliation(s)
| | - Arianna Bassan
- Innovatune srl, Via Giulio Zanon 130/D, 35129 Padova,
Italy
| | - Mamta Behl
- Division of the National Toxicology Program, National
Institutes of Environmental Health Sciences, Durham, NC 27709, USA
| | - Yaroslav G. Chushak
- Henry M Jackson Foundation for the Advancement of Military
Medicine, Wright-Patterson AFB, OH 45433, USA
| | - Ellen Fritsche
- IUF – Leibniz Research Institute for Environmental
Medicine & Medical Faculty Heinrich-Heine-University, Düsseldorf,
Germany
| | - Jeffery M. Gearhart
- Henry M Jackson Foundation for the Advancement of Military
Medicine, Wright-Patterson AFB, OH 45433, USA
| | | | - Moiz Mumtaz
- Agency for Toxic Substances and Disease Registry, US
Department of Health and Human Services, Atlanta, GA, USA
| | - Manuela Pavan
- Innovatune srl, Via Giulio Zanon 130/D, 35129 Padova,
Italy
| | - Patricia Ruiz
- Agency for Toxic Substances and Disease Registry, US
Department of Health and Human Services, Atlanta, GA, USA
| | - Magdalini Sachana
- Environment Health and Safety Division, Environment
Directorate, Organisation for Economic Co-Operation and Development (OECD), 75775
Paris Cedex 16, France
| | - Rajamani Selvam
- Office of Clinical Pharmacology, Office of Translational
Sciences, Center for Drug Evaluation and Research (CDER), U.S. Food and Drug
Administration (FDA), Silver Spring, MD 20993, USA
| | - Timothy J. Shafer
- Biomolecular and Computational Toxicology Division, Center
for Computational Toxicology and Exposure, US EPA, Research Triangle Park, NC,
USA
| | - Lidiya Stavitskaya
- Office of Clinical Pharmacology, Office of Translational
Sciences, Center for Drug Evaluation and Research (CDER), U.S. Food and Drug
Administration (FDA), Silver Spring, MD 20993, USA
| | | | | | | | - Dan Wilson
- The Dow Chemical Company, Midland, MI 48667, USA
| | | | - Glenn J. Myatt
- Instem, Columbus, OH 43215, USA
- Corresponding author.
(G.J. Myatt)
| |
Collapse
|
10
|
Liu X, Wei Q, Yang X, Wang X, Zhang J, Xu R, Zhang H, Wang S, Wan X, Jiang L, He Y, Li S, Chen R, Wang Y, Chen Y, Qin F, Chen Y, Dai Y, Li H, Zhao Y, Zhang H, Bu Q, Wang H, Tian J, Zhao Y, Cen X. Lipidomics Reveals Dysregulated Glycerophospholipid Metabolism in the Corpus Striatum of Mice Treated with Cefepime. ACS Chem Neurosci 2021; 12:4449-4464. [PMID: 34762393 DOI: 10.1021/acschemneuro.1c00608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Cefepime exhibits a broad spectrum of antimicrobial activity and thus is a widely used treatment for severe bacterial infections. Adverse effects on the central nervous system (CNS) have been reported in patients treated with cefepime. Current explanation for the adverse neurobehavioral effect of cefepime is mainly attributed to its ability to cross the blood-brain barrier and competitively bind to the GABAergic receptor; however, the underlying mechanism is largely unknown. In this study, mice were intraperitoneally administered 80 mg/kg cefepime for different periods, followed by neurobehavioral tests and a brain lipidomic analysis. LC/MS-MS-based metabolomics was used to investigate the effect of cefepime on the brain lipidomic profile and metabolic pathways. Repeated cefepime treatment time-dependently caused anxiety-like behaviors, which were accompanied by reduced locomotor activity in the open field test. Cefepime profoundly altered the lipid profile, acyl chain length, and unsaturation of fatty acids in the corpus striatum, and glycerophospholipids accounted for a large proportion of those significantly modified lipids. In addition, cefepime treatment caused obvious alteration in the lipid-enriched membrane structure, neurites, mitochondria, and synaptic vesicles of primary cultured striatal neurons; moreover, the spontaneous electrical activity of striatal neurons was significantly reduced. Collectively, cefepime reprograms glycerophospholipid metabolism in the corpus striatum, which may interfere with neuronal structure and activity, eventually leading to aberrant neurobehaviors in mice.
Collapse
Affiliation(s)
- Xiaocong Liu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Qingfan Wei
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Xiaowei Yang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Xiaojie Wang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Jiamei Zhang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Rui Xu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Haoluo Zhang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Shaomin Wang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Xuemei Wan
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Linhong Jiang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Yuman He
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Shu Li
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Rong Chen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Yonghai Wang
- Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, People’s Republic of China
| | - Yaxing Chen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Feng Qin
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Yuanyuan Chen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Yanping Dai
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Hongchun Li
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Ying Zhao
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Huaqin Zhang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Qian Bu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Hongbo Wang
- Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, People’s Republic of China
| | - Jingwei Tian
- Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, People’s Republic of China
| | - Yinglan Zhao
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| | - Xiaobo Cen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 1 Keyuan Road, Gaopeng Street, High-Tech Development Zone, Chengdu 610041, People’s Republic of China
| |
Collapse
|
11
|
Kumar D, Baligar P, Srivastav R, Narad P, Raj S, Tandon C, Tandon S. Stem Cell Based Preclinical Drug Development and Toxicity Prediction. Curr Pharm Des 2021; 27:2237-2251. [PMID: 33076801 DOI: 10.2174/1381612826666201019104712] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/22/2020] [Indexed: 01/09/2023]
Abstract
Stem cell based toxicity prediction plays a very important role in the development of the drug. Unexpected adverse effects of the drugs during clinical trials are a major reason for the termination or withdrawal of drugs. Methods for predicting toxicity employ in vitro as well as in vivo models; however, the major drawback seen in the data derived from these animal models is the lack of extrapolation, owing to interspecies variations. Due to these limitations, researchers have been striving to develop more robust drug screening platforms based on stem cells. The application of stem cells based toxicity testing has opened up robust methods to study the impact of new chemical entities on not only specific cell types, but also organs. Pluripotent stem cells, as well as cells derived from them, can be evaluated for modulation of cell function in response to drugs. Moreover, the combination of state-of-the -art techniques such as tissue engineering and microfluidics to fabricate organ- on-a-chip, has led to assays which are amenable to high throughput screening to understand the adverse and toxic effects of chemicals and drugs. This review summarizes the important aspects of the establishment of the embryonic stem cell test (EST), use of stem cells, pluripotent, induced pluripotent stem cells and organoids for toxicity prediction and drug development.
Collapse
Affiliation(s)
- Dhruv Kumar
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida, Uttar Pradesh 201313, India
| | - Prakash Baligar
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida, Uttar Pradesh 201313, India
| | - Rajpal Srivastav
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201313, India
| | - Priyanka Narad
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201313, India
| | - Sibi Raj
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida, Uttar Pradesh 201313, India
| | - Chanderdeep Tandon
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201313, India
| | - Simran Tandon
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida, Uttar Pradesh 201313, India
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Maiorov SA, Zinchenko VP, Gaidin SG, Kosenkov AM. Potential mechanism of GABA secretion in response to the activation of GluK1-containing kainate receptors. Neurosci Res 2021; 171:27-33. [PMID: 33785410 DOI: 10.1016/j.neures.2021.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/08/2021] [Accepted: 03/17/2021] [Indexed: 11/26/2022]
Abstract
Hippocampal GABAergic neurons are subdivided into more than 20 subtypes that are distinguished by features and functions. We have previously described the subpopulation of GABAergic neurons, which can be identified in hippocampal cell culture by the calcium response to the application of domoic acid (DoA), an agonist of kainate receptors (KARs). Here, we investigate the features of DoA-sensitive neurons and their GABA release mechanism in response to KARs activation. We demonstrate that DoA-sensitive GABAergic neurons express GluK1-containing KARs because ATPA, a selective agonist of GluK1-containing receptors, induces the calcium response exclusively in these GABAergic neurons. Our experiments also show that NASPM, previously considered a selective antagonist of calcium-permeable AMPARs, blocks calcium-permeable KARs. We established using NASPM that GluK1-containing receptors of the studied population of GABAergic neurons are calcium-permeable, and their activation is required for GABA release, at least in particular synapses. Notably, GABA release occurs even in the presence of tetrodotoxin, indicating that propagation of the depolarizing stimulus is not required for GABA release in this case. Thus, our data demonstrate that the activation of GluK1-containing calcium-permeable KARs mediates the GABA release by the studied subpopulation of GABAergic neurons.
Collapse
Affiliation(s)
- S A Maiorov
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290, Pushchino, Russia
| | - V P Zinchenko
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290, Pushchino, Russia
| | - S G Gaidin
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290, Pushchino, Russia.
| | - A M Kosenkov
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290, Pushchino, Russia.
| |
Collapse
|
15
|
A human stem cell-derived test system for agents modifying neuronal N-methyl-D-aspartate-type glutamate receptor Ca 2+-signalling. Arch Toxicol 2021; 95:1703-1722. [PMID: 33713149 PMCID: PMC8113295 DOI: 10.1007/s00204-021-03024-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/04/2021] [Indexed: 12/12/2022]
Abstract
Methods to assess neuronal receptor functions are needed in toxicology and for drug development. Human-based test systems that allow studies on glutamate signalling are still scarce. To address this issue, we developed and characterized pluripotent stem cell (PSC)-based neural cultures capable of forming a functional network. Starting from a stably proliferating neuroepithelial stem cell (NESC) population, we generate “mixed cortical cultures” (MCC) within 24 days. Characterization by immunocytochemistry, gene expression profiling and functional tests (multi-electrode arrays) showed that MCC contain various functional neurotransmitter receptors, and in particular, the N-methyl-d-aspartate subtype of ionotropic glutamate receptors (NMDA-R). As this important receptor is found neither on conventional neural cell lines nor on most stem cell-derived neurons, we focused here on the characterization of rapid glutamate-triggered Ca2+ signalling. Changes of the intracellular free calcium ion concentration ([Ca2+]i) were measured by fluorescent imaging as the main endpoint, and a method to evaluate and quantify signals in hundreds of cells at the same time was developed. We observed responses to glutamate in the low µM range. MCC responded to kainate and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), and a subpopulation of 50% had functional NMDA-R. The receptor was modulated by Mg2+, Zn2+ and Pb2+ in the expected ways, and various toxicologically relevant agonists (quinolinic acid, ibotenic acid, domoic acid) triggered [Ca2+]i responses in MCC. Antagonists, such as phencyclidine, ketamine and dextromethorphan, were also readily identified. Thus, the MCC developed here may fill an important gap in the panel of test systems available to characterize the effects of chemicals on neurotransmitter receptors.
Collapse
|
16
|
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
|
17
|
Domínguez HJ, Cabrera-García D, Cuadrado C, Novelli A, Fernández-Sánchez MT, Fernández JJ, Daranas AH. Prorocentroic Acid, a Neuroactive Super-Carbon-Chain Compound from the Dinoflagellate Prorocentrum hoffmannianum. Org Lett 2020; 23:13-18. [DOI: 10.1021/acs.orglett.0c03437] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Humberto J. Domínguez
- Instituto Universitario de Bio-Orgánica Antonio González, University of La Laguna, 38206, Tenerife, Spain
| | - David Cabrera-García
- Department of Biochemistry and Molecular Biology and University Institute of Biotechnology of Asturias (IUBA), Campus “El Cristo”, University of Oviedo, 33006 Oviedo, Oviedo, Spain
| | - Cristina Cuadrado
- Instituto de Productos Naturales y Agrobiología, Consejo Superior de Investigaciones Científicas (IPNA-CSIC), La Laguna, 38206, Tenerife, Spain
| | - Antonello Novelli
- Department of Psychology and University Institute of Biotechnology of Asturias (IUBA), Campus “El Cristo”, University of Oviedo; Institute for Sanitary Research of the Princedom of Asturias (ISPA), 33006 Oviedo, Oviedo, Spain
| | - M. Teresa Fernández-Sánchez
- Department of Biochemistry and Molecular Biology and University Institute of Biotechnology of Asturias (IUBA), Campus “El Cristo”, University of Oviedo, 33006 Oviedo, Oviedo, Spain
| | - José J. Fernández
- Instituto Universitario de Bio-Orgánica Antonio González, University of La Laguna, 38206, Tenerife, Spain
| | - Antonio Hernández Daranas
- Instituto de Productos Naturales y Agrobiología, Consejo Superior de Investigaciones Científicas (IPNA-CSIC), La Laguna, 38206, Tenerife, Spain
| |
Collapse
|
18
|
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
|
19
|
Lovett ML, Nieland TJ, Dingle YTL, Kaplan DL. Innovations in 3-Dimensional Tissue Models of Human Brain Physiology and Diseases. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1909146. [PMID: 34211358 PMCID: PMC8240470 DOI: 10.1002/adfm.201909146] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Indexed: 05/04/2023]
Abstract
3-dimensional (3D) laboratory tissue cultures have emerged as an alternative to traditional 2-dimensional (2D) culture systems that do not recapitulate native cell behavior. The discrepancy between in vivo and in vitro tissue-cell-molecular responses impedes understanding of human physiology in general and creates roadblocks for the discovery of therapeutic solutions. Two parallel approaches have emerged for the design of 3D culture systems. The first is biomedical engineering methodology, including bioengineered materials, bioprinting, microfluidics and bioreactors, used alone or in combination, to mimic the microenvironments of native tissues. The second approach is organoid technology, in which stem cells are exposed to chemical and/or biological cues to activate differentiation programs that are reminiscent of human (prenatal) development. This review article describes recent technological advances in engineering 3D cultures that more closely resemble the human brain. The contributions of in vitro 3D tissue culture systems to new insights in neurophysiology, neurological diseases and regenerative medicine are highlighted. Perspectives on designing improved tissue models of the human brain are offered, focusing on an integrative approach merging biomedical engineering tools with organoid biology.
Collapse
Affiliation(s)
- Michael L. Lovett
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155
| | - Thomas J.F. Nieland
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155
| | - Yu-Ting L. Dingle
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155
| |
Collapse
|
20
|
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
|
21
|
Badman RP, Moore SL, Killian JL, Feng T, Cleland TA, Hu F, Wang MD. Dextran-coated iron oxide nanoparticle-induced nanotoxicity in neuron cultures. Sci Rep 2020; 10:11239. [PMID: 32641693 PMCID: PMC7343881 DOI: 10.1038/s41598-020-67724-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 02/27/2020] [Indexed: 11/09/2022] Open
Abstract
Recent technological advances have introduced diverse engineered nanoparticles (ENPs) into our air, water, medicine, cosmetics, clothing, and food. However, the health and environmental effects of these increasingly common ENPs are still not well understood. In particular, potential neurological effects are one of the most poorly understood areas of nanoparticle toxicology (nanotoxicology), in that low-to-moderate neurotoxicity can be subtle and difficult to measure. Culturing primary neuron explants on planar microelectrode arrays (MEAs) has emerged as one of the most promising in vitro techniques with which to study neuro-nanotoxicology, as MEAs enable the fluorescent tracking of nanoparticles together with neuronal electrical activity recording at the submillisecond time scale, enabling the resolution of individual action potentials. Here we examine the dose-dependent neurotoxicity of dextran-coated iron oxide nanoparticles (dIONPs), a common type of functionalized ENP used in biomedical applications, on cultured primary neurons harvested from postnatal day 0-1 mouse brains. A range of dIONP concentrations (5-40 µg/ml) were added to neuron cultures, and cells were plated either onto well plates for live cell, fluorescent reactive oxidative species (ROS) and viability observations, or onto planar microelectrode arrays (MEAs) for electrophysiological measurements. Below 10 µg/ml, there were no dose-dependent cellular ROS increases or effects in MEA bursting behavior at sub-lethal dosages. However, above 20 µg/ml, cell death was obvious and widespread. Our findings demonstrate a significant dIONP toxicity in cultured neurons at concentrations previously reported to be safe for stem cells and other non-neuronal cell types.
Collapse
Affiliation(s)
- Ryan P Badman
- Department of Physics and LASSP, Cornell University, Ithaca, NY, 14853, USA.,Center for Brain Science, RIKEN, Saitama, 351-0198, Japan
| | - Shanna L Moore
- Department of Physics and LASSP, Cornell University, Ithaca, NY, 14853, USA.,Howard Hughes Medical Institute, Cornell University, Ithaca, NY, 14853, USA
| | - Jessica L Killian
- Department of Physics and LASSP, Cornell University, Ithaca, NY, 14853, USA.,Howard Hughes Medical Institute, Cornell University, Ithaca, NY, 14853, USA.,Quantum Biosystems, Menlo Park, CA, 94025, USA
| | - Tuancheng Feng
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA.,Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Thomas A Cleland
- Department of Psychology, Cornell University, Ithaca, NY, 14853, USA
| | - Fenghua Hu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA.,Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Michelle D Wang
- Department of Physics and LASSP, Cornell University, Ithaca, NY, 14853, USA. .,Howard Hughes Medical Institute, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
22
|
Shafer TJ, Brown JP, Lynch B, Davila-Montero S, Wallace K, Friedman KP. Evaluation of Chemical Effects on Network Formation in Cortical Neurons Grown on Microelectrode Arrays. Toxicol Sci 2020; 169:436-455. [PMID: 30816951 DOI: 10.1093/toxsci/kfz052] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Thousands of chemicals to which humans are potentially exposed have not been evaluated for potential developmental neurotoxicity (DNT), driving efforts to develop a battery of in vitro screening approaches for DNT hazard. Here, 136 unique chemicals were evaluated for potential DNT hazard using a network formation assay (NFA) in cortical cells grown on microelectrode arrays. The effects of chemical exposure from 2 h postplating through 12 days in vitro (DIV) on network formation were evaluated at DIV 5, 7, 9, and 12, with cell viability assessed at DIV 12. Only 82 chemicals altered at least 1 network development parameter. Assay results were reproducible; 10 chemicals tested as biological replicates yielded qualitative results that were 100% concordant, with consistent potency values. Toxicological tipping points were determined for 58 chemicals and were similar to or lower than the lowest 50% effect concentrations (EC50) for all parameters. When EC50 and tipping point values from the NFA were compared to the range of potencies observed in ToxCast assays, the NFA EC50 values were less than the lower quartile for ToxCast assay potencies for a subset of chemicals, many of which are acutely neurotoxic in vivo. For 13 chemicals with available in vivo DNT data, estimated administered equivalent doses based on NFA results were similar to or lower than administered doses in vivo. Collectively, these results indicate that the NFA is sensitive to chemicals acting on nervous system function and will be a valuable contribution to an in vitro DNT screening battery.
Collapse
Affiliation(s)
- Timothy J Shafer
- Integrated Systems Toxicology Division, NHEERL, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - Jasmine P Brown
- Integrated Systems Toxicology Division, NHEERL, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711.,Graduate Program in Public Health, University of Michigan, Ann Arbor, MI
| | - Brittany Lynch
- Tandon School of Engineering, New York University, Brooklyn, New York 11201
| | - Sylmarie Davila-Montero
- Department of Electrical and Computer Engineering, Michigan State University, E. Lansing, Michigan 48824
| | - Kathleen Wallace
- Integrated Systems Toxicology Division, NHEERL, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - Katie Paul Friedman
- National Center for Computational Toxicology, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| |
Collapse
|
23
|
Characterization and application of electrically active neuronal networks established from human induced pluripotent stem cell-derived neural progenitor cells for neurotoxicity evaluation. Stem Cell Res 2020; 45:101761. [PMID: 32244191 DOI: 10.1016/j.scr.2020.101761] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 02/20/2020] [Accepted: 03/05/2020] [Indexed: 12/13/2022] Open
Abstract
Neurotoxicity is mediated by a variety of modes-of-actions leading to disturbance of neuronal function. In order to screen larger numbers of compounds for their neurotoxic potential, in vitro functional neuronal networks (NN) might be helpful tools. We established and characterized human NN (hNN) from hiPSC-derived neural progenitor cells by comparing hNN formation with two different differentiation media: in presence (CINDA) and absence (neural differentiation medium (NDM)) of maturation-supporting factors. As a NN control we included differentiating rat NN (rNN) in the study. Gene/protein expression and electrical activity from in vitro developing NN were assessed at multiple time points. Transcriptomes of 5, 14 and 28 days in vitro CINDA-grown hNN were compared to gene expression profiles of in vivo human developing brains. Molecular expression analyses as well as measures of electrical activity indicate that NN mature into neurons of different subtypes and astrocytes over time. In contrast to rNN, hNN are less electrically active within the same period of differentiation time, yet hNN grown in CINDA medium develop higher firing rates than hNN without supplements. Challenge of NN with neuronal receptor stimulators and inhibitors demonstrate presence of inhibitory, GABAergic neurons, whereas glutamatergic responses are limited. hiPSC-derived GABAergic hNN grown in CINDA medium might be a useful tool as part of an in vitro battery for assessing neurotoxicity.
Collapse
|
24
|
Simultaneous electrophysiological and morphological assessment of functional damage to neural networks in vitro after 30-300 g impacts. Sci Rep 2019; 9:14994. [PMID: 31628381 PMCID: PMC6802386 DOI: 10.1038/s41598-019-51541-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 09/26/2019] [Indexed: 11/08/2022] Open
Abstract
An enigma of mild traumatic brain injury are observations of substantial behavior and performance deficits in the absence of bleeding or other observable structural damage. Altered behavior and performance reflect changes in action potential (AP) patterns within neuronal networks, which could result from subtle subcellular responses that affect synaptic efficacy and AP production. The aim of this study was to investigate and quantify network activity changes after simulated concussions in vitro and therewith develop a platform for simultaneous and direct observations of morphological and electrophysiological changes in neural networks. We used spontaneously active networks grown on microelectrode arrays (MEAs) to allow long-term multisite monitoring with simultaneous optical observations before and after impacts delivered by a ballistic pendulum (30 to 300 g accelerations). The monitoring of AP waveshape templates for long periods before and after impact provided an internal control for cell death or loss of cell-electrode coupling in the observed set of neurons. Network activity patterns were linked in real-time to high power phase contrast microscopy. There was no overt loss of glial or neuronal adhesion, even at high-g impacts. All recording experiments showed repeatable spike production responses: a loss of activity with recovery to near reference in 1 hr, followed by a slow activity decay to a stable, level plateau approximately 30–40% below reference. The initial recovery occurred in two steps: a rapid return of activity to an average 24% below reference, forming a level plateau lasting from 5 to 20 min, followed by a climb to within 10% of reference where a second plateau was established for 1 to 2 hrs. Cross correlation profiles revealed changes in firing hierarchy as well as in Phase 1 in spontaneous network oscillations that were reduced by as much as 20% 6–8 min post impact with only a partial recovery at 30 min. We also observed that normally stable nuclei developed irregular rotational motion after impact in 27 out of 30 networks. The evolution of network activity deficits and recovery can be linked with microscopically observable changes in the very cells that are generating the activity. The repeatable electrophysiological impact response profiles and oscillation changes can provide a quantitative basis for systematic evaluations of pharmacological intervention strategies. Future expansion to include fluorescent microscopy should allow detailed investigations of damage mechanisms on the subcellular level.
Collapse
|
25
|
Differential effects of fluoxetine and venlafaxine in the neural embryonic stem cell test (ESTn) revealed by a cell lineage map. Neurotoxicology 2019; 76:1-9. [PMID: 31593710 DOI: 10.1016/j.neuro.2019.09.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/21/2019] [Accepted: 09/22/2019] [Indexed: 01/21/2023]
Abstract
There is a need for in vitro tests for the evaluation of chemicals and pharmaceuticals that may cause developmental neurotoxicity (DNT) in humans. The neural embryonic stem cell test (ESTn) is such an in vitro test that mimics early neural differentiation. The aim of this study was to define the biological domain of ESTn based on the expression of selective markers for certain cell types, and to investigate the effects of two antidepressants, fluoxetine (FLX) and venlafaxine (VNX), on neural differentiation. A cell lineage map was made to track neural differentiation and the effects of FLX and VNX in ESTn. Whole transcriptome analysis revealed differentiation from an embryonic stem cell population to a mixed culture of neural progenitors, neurons and neural crest cells 7 days into differentiation. Maturing neurons, astrocytes and oligodendrocytes were present after 13 days. Exposure to FLX or VNX led to different expression patterns between compounds at both time points. On day 7, both compounds upregulated most of the stem cell- and immature neuron markers, but had distinct effects on neural subtype markers. FLX downregulated glycinergic markers and upregulated cholinergic markers, while VNX had the opposite effect. On day 13, FLX and VNX affected their specific therapeutic targets, represented by mainly serotonergic markers by FLX- and dopaminergic and noradrenergic markers in VNX-exposed cultures, as well as oligodendrocyte and glycinergic neuron markers. This proof of concept study shows the added value of assessing DNT in ESTn through a cell lineage map and gives mechanistic insight in the potential neurodevelopmental effects of FLX and VNX. More compounds should be tested to further evaluate the use of the cell lineage map.
Collapse
|
26
|
Fritsche E, Barenys M, Klose J, Masjosthusmann S, Nimtz L, Schmuck M, Wuttke S, Tigges J. Current Availability of Stem Cell-Based In Vitro Methods for Developmental Neurotoxicity (DNT) Testing. Toxicol Sci 2019; 165:21-30. [PMID: 29982830 DOI: 10.1093/toxsci/kfy178] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
There is evidence that chemical exposure during development can cause irreversible impairments of the human developing nervous system. Therefore, testing compounds for their developmentally neurotoxic potential has high priority for different stakeholders: academia, industry, and regulatory bodies. Due to the resource-intensity of current developmental neurotoxicity (DNT) in vivo guidelines, alternative methods that are scientifically valid and have a high predictivity for humans are especially desired by regulators. Here, we review availability of stem-/progenitor cell-based in vitro methods for DNT evaluation that is based on the concept of neurodevelopmental process assessment. These test methods are assembled into a DNT in vitro testing battery. Gaps in this testing battery addressing research needs are also pointed out.
Collapse
Affiliation(s)
| | - Marta Barenys
- IUF - Leibniz Research Institute for Environmental Medicine 40225, Düsseldorf, Germany
| | - Jördis Klose
- IUF - Leibniz Research Institute for Environmental Medicine 40225, Düsseldorf, Germany
| | - Stefan Masjosthusmann
- IUF - Leibniz Research Institute for Environmental Medicine 40225, Düsseldorf, Germany
| | - Laura Nimtz
- IUF - Leibniz Research Institute for Environmental Medicine 40225, Düsseldorf, Germany
| | - Martin Schmuck
- IUF - Leibniz Research Institute for Environmental Medicine 40225, Düsseldorf, Germany
| | - Saskia Wuttke
- IUF - Leibniz Research Institute for Environmental Medicine 40225, Düsseldorf, Germany
| | - Julia Tigges
- IUF - Leibniz Research Institute for Environmental Medicine 40225, Düsseldorf, Germany
| |
Collapse
|
27
|
Domoic acid suppresses hyperexcitation in the network due to activation of kainate receptors of GABAergic neurons. Arch Biochem Biophys 2019; 671:52-61. [DOI: 10.1016/j.abb.2019.06.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/12/2019] [Accepted: 06/15/2019] [Indexed: 01/01/2023]
|
28
|
Abstract
Substrate-integrated multielectrode arrays (MEAs) enable multisite, long-term, and label-free sensing and actuation of neuronal electrical signals in reduced cell culture models for network electrophysiology. Conventional, thin-film fabricated passive MEAs typically provide a few tens of electrode sites. New generations of active CMOS-based high-resolution arrays provide the capabilities of simultaneous recordings from thousands of neurons over fields of view of several square millimeters, yet allowing extracellular electrical imaging to be achieved down to the subcellular scale. In turn, such advancement in chip-based electrical readouts can significantly complement recently developed biotechnological and bimolecular techniques for neurobiology applications. Here, we describe (1) a simple method to fabricate passive MEAs and (2) protocols for preparing and growing primary rat hippocampal neuronal cultures and human iPS-derived neurons on MEAs. The aim is to provide reliable protocols for initiating the reader to this technology and for stimulating their further development and experimental use in neurobiology.
Collapse
|
29
|
SPICODYN: A Toolbox for the Analysis of Neuronal Network Dynamics and Connectivity from Multi-Site Spike Signal Recordings. Neuroinformatics 2019; 16:15-30. [PMID: 28988388 DOI: 10.1007/s12021-017-9343-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We implemented an automated and efficient open-source software for the analysis of multi-site neuronal spike signals. The software package, named SPICODYN, has been developed as a standalone windows GUI application, using C# programming language with Microsoft Visual Studio based on .NET framework 4.5 development environment. Accepted input data formats are HDF5, level 5 MAT and text files, containing recorded or generated time series spike signals data. SPICODYN processes such electrophysiological signals focusing on: spiking and bursting dynamics and functional-effective connectivity analysis. In particular, for inferring network connectivity, a new implementation of the transfer entropy method is presented dealing with multiple time delays (temporal extension) and with multiple binary patterns (high order extension). SPICODYN is specifically tailored to process data coming from different Multi-Electrode Arrays setups, guarantying, in those specific cases, automated processing. The optimized implementation of the Delayed Transfer Entropy and the High-Order Transfer Entropy algorithms, allows performing accurate and rapid analysis on multiple spike trains from thousands of electrodes.
Collapse
|
30
|
Pérez-Gómez A, Cabrera-García D, Warm D, Marini AM, Salas Puig J, Fernández-Sánchez MT, Novelli A. From the Cover: Selective Enhancement of Domoic Acid Toxicity in Primary Cultures of Cerebellar Granule Cells by Lowering Extracellular Na+ Concentration. Toxicol Sci 2019; 161:103-114. [PMID: 29029261 DOI: 10.1093/toxsci/kfx201] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Domoic acid (DOM) is an excitatory amino acid analog of kainic acid (KA) that acts through glutamic acid (GLU) receptors, inducing a fast and potent neurotoxic response. Here, we present evidence for an enhancement of excitotoxicity following exposure of cultured cerebellar granule cells to DOM in the presence of lower than physiological Na+ concentrations. The concentration of DOM that reduced by 50% neuronal survival was approximately 3 µM in Na+-free conditions and 16 µM in presence of a physiological concentration of extracellular Na+. The enhanced neurotoxic effect of DOM was fully prevented by AMPA/KA receptor antagonist, while N-methyl-D-aspartate-receptor-mediated neurotoxicity did not seem to be involved, as the absence of extracellular Na+ failed to potentiate GLU excitotoxicity under the same experimental conditions. Lowering of extracellular Na+ concentration to 60 mM eliminated extracellular recording of spontaneous electrophysiological activity from cultured neurons grown on a multi electrode array and prevented DOM stimulation of the electrical activity. Although changes in the extracellular Na+ concentration did not alter the magnitude of the rapid increase in intracellular Ca2+ levels associated to DOM exposure, they did change significantly the contribution of voltage-sensitive calcium channels (VScaCs) and the recovery time to baseline. The prevention of Ca2+ influx via VSCaCs by nifedipine failed to prevent DOM toxicity at any extracellular Na+ concentration, while the reduction of extracellular Ca2+ concentration ameliorated DOM toxicity only in the absence of extracellular Na+, enhancing it in physiological conditions. Our data suggest a crucial role for extracellular Na+ concentration in determining excitotoxicity by DOM.
Collapse
Affiliation(s)
- Anabel Pérez-Gómez
- Department of Biochemistry and Molecular Biology, University of Oviedo, Oviedo, Spain
| | - David Cabrera-García
- Department of Biochemistry and Molecular Biology, University of Oviedo, Oviedo, Spain
| | - Davide Warm
- Department of Biochemistry and Molecular Biology, University of Oviedo, Oviedo, Spain
| | - Ann M Marini
- Department of Neurology and Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814
| | - Javier Salas Puig
- Unit of Epilepsy, Vall d'Hebrón Hospital, Barcelona, Spain.,Department of Medicine, University Autonoma of Barcelona, Barcelona, Spain
| | - Maria Teresa Fernández-Sánchez
- Department of Biochemistry and Molecular Biology, University of Oviedo, Oviedo, Spain.,University Institute of Biotechnology
| | - Antonello Novelli
- University Institute of Biotechnology.,Department of Psychology, University of Oviedo, Oviedo, Spain
| |
Collapse
|
31
|
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
|
32
|
Frank CL, Brown JP, Wallace K, Wambaugh JF, Shah I, Shafer TJ. Defining toxicological tipping points in neuronal network development. Toxicol Appl Pharmacol 2018; 354:81-93. [DOI: 10.1016/j.taap.2018.01.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 01/22/2018] [Indexed: 12/18/2022]
|
33
|
Bal-Price A, Pistollato F, Sachana M, Bopp SK, Munn S, Worth A. Strategies to improve the regulatory assessment of developmental neurotoxicity (DNT) using in vitro methods. Toxicol Appl Pharmacol 2018; 354:7-18. [PMID: 29476865 PMCID: PMC6095942 DOI: 10.1016/j.taap.2018.02.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/05/2018] [Accepted: 02/13/2018] [Indexed: 01/23/2023]
Abstract
Currently, the identification of chemicals that have the potential to induce developmental neurotoxicity (DNT) is based on animal testing. Since at the regulatory level, systematic testing of DNT is not a standard requirement within the EU or USA chemical legislation safety assessment, DNT testing is only performed in higher tiered testing triggered based on chemical structure activity relationships or evidence of neurotoxicity in systemic acute or repeated dose toxicity studies. However, these triggers are rarely used and, in addition, do not always serve as reliable indicators of DNT, as they are generally based on observations in adult rodents. Therefore, there is a pressing need for developing alternative methodologies that can reliably support identification of DNT triggers, and more rapidly and cost-effectively support the identification and characterization of chemicals with DNT potential. We propose to incorporate mechanistic knowledge and data derived from in vitro studies to support various regulatory applications including: (a) the identification of potential DNT triggers, (b) initial chemical screening and prioritization, (c) hazard identification and characterization, (d) chemical biological grouping, and (e) assessment of exposure to chemical mixtures. Ideally, currently available cellular neuronal/glial models derived from human induced pluripotent stem cells (hiPSCs) should be used as they allow evaluation of chemical impacts on key neurodevelopmental processes, by reproducing different windows of exposure during human brain development. A battery of DNT in vitro test methods derived from hiPSCs could generate valuable mechanistic data, speeding up the evaluation of thousands of compounds present in industrial, agricultural and consumer products that lack safety data on DNT potential.
Collapse
Affiliation(s)
- Anna Bal-Price
- European Commission, Joint Research Centre (JRC), Ispra, Italy.
| | | | - Magdalini Sachana
- Organisation for Economic Co-operation and Development (OECD), 2 rue André Pascal, 75775 Paris, Cedex 16, France
| | | | - Sharon Munn
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Andrew Worth
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| |
Collapse
|
34
|
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
|
35
|
Electrophysiological evidence of RML12 mosquito cell line towards neuronal differentiation by 20-hydroxyecdysdone. Sci Rep 2018; 8:10109. [PMID: 29973702 PMCID: PMC6031678 DOI: 10.1038/s41598-018-28357-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 06/07/2018] [Indexed: 01/06/2023] Open
Abstract
Continuous cell lines from insect larval tissues are widely used in different research domains, such as virology, insect immunity, gene expression, and bio pharmacology. Previous study showed that introduction of 20-hydroxyecdysone to Spodoptera cell line induced a neuron-like morphology with neurite extensions. Despite some results suggesting potential presence of neuro-receptors, no study so far has shown that these neuron-induced cells were functional. Here, using microelectrode arrays, we showed that the mosquito cell line, RML12, differentiated with 20-hydroxyecdysone, displays spontaneous electrophysiological activity. Results showed that these cells can be stimulated by GABAergic antagonist as well as nicotinic agonist. These results provide new evidence of neuron-like functionality of 20-hydroxyecdysone induced differentiated mosquito cell line. Finally, we used this new model to test the effects of two insecticides, temephos and permethrin. Our analysis revealed significant changes in the spiking activity after the introduction of these insecticides with prolonged effect on the neuronal activity. We believe that this differentiated mosquito neuronal cell model can be used for high-throughput screening of new pesticides on insect nervous system instead of primary neurons or in vivo studies.
Collapse
|
36
|
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: 84] [Impact Index Per Article: 12.0] [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
|
37
|
Baskar MK, Murthy PB. Acute in vitro neurotoxicity of some pyrethroids using microelectrode arrays. Toxicol In Vitro 2018; 47:165-177. [DOI: 10.1016/j.tiv.2017.11.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 10/10/2017] [Accepted: 11/15/2017] [Indexed: 12/23/2022]
|
38
|
Bal-Price A, Hogberg HT, Crofton KM, Daneshian M, FitzGerald RE, Fritsche E, Heinonen T, Hougaard Bennekou S, Klima S, Piersma AH, Sachana M, Shafer TJ, Terron A, Monnet-Tschudi F, Viviani B, Waldmann T, Westerink RHS, Wilks MF, Witters H, Zurich MG, Leist M. Recommendation on test readiness criteria for new approach methods in toxicology: Exemplified for developmental neurotoxicity. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2018; 35:306-352. [PMID: 29485663 DOI: 10.14573/altex.1712081] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 01/29/2018] [Indexed: 01/06/2023]
Abstract
Multiple non-animal-based test methods have never been formally validated. In order to use such new approach methods (NAMs) in a regulatory context, criteria to define their readiness are necessary. The field of developmental neurotoxicity (DNT) testing is used to exemplify the application of readiness criteria. The costs and number of untested chemicals are overwhelming for in vivo DNT testing. Thus, there is a need for inexpensive, high-throughput NAMs, to obtain initial information on potential hazards, and to allow prioritization for further testing. A background on the regulatory and scientific status of DNT testing is provided showing different types of test readiness levels, depending on the intended use of data from NAMs. Readiness criteria, compiled during a stakeholder workshop, uniting scientists from academia, industry and regulatory authorities are presented. An important step beyond the listing of criteria, was the suggestion for a preliminary scoring scheme. On this basis a (semi)-quantitative analysis process was assembled on test readiness of 17 NAMs with respect to various uses (e.g. prioritization/screening, risk assessment). The scoring results suggest that several assays are currently at high readiness levels. Therefore, suggestions are made on how DNT NAMs may be assembled into an integrated approach to testing and assessment (IATA). In parallel, the testing state in these assays was compiled for more than 1000 compounds. Finally, a vision is presented on how further NAM development may be guided by knowledge of signaling pathways necessary for brain development, DNT pathophysiology, and relevant adverse outcome pathways (AOP).
Collapse
Affiliation(s)
- Anna Bal-Price
- European Commission, Joint Research Centre (EC JRC), Ispra (VA), Italy
| | - Helena T Hogberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Baltimore, MD, USA
| | - Kevin M Crofton
- National Centre for Computational Toxicology, US EPA, RTP, Washington, NC, USA
| | - Mardas Daneshian
- Center for Alternatives to Animal Testing, CAAT-Europe, University of Konstanz, Konstanz, Germany
| | - Rex E FitzGerald
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland
| | - Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine & Heinrich-Heine-University, Düsseldorf, Germany
| | - Tuula Heinonen
- Finnish Centre for Alternative Methods (FICAM), University of Tampere, Tampere, Finland
| | | | - Stefanie Klima
- In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Aldert H Piersma
- RIVM, National Institute for Public Health and the Environment, Bilthoven, and Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Magdalini Sachana
- Organisation for Economic Co-operation and Development (OECD), Paris, France
| | - Timothy J Shafer
- National Centre for Computational Toxicology, US EPA, RTP, Washington, NC, USA
| | | | - Florianne Monnet-Tschudi
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland.,Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Barbara Viviani
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Tanja Waldmann
- In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Remco H S Westerink
- Neurotoxicology Research Group, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Martin F Wilks
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland
| | - Hilda Witters
- VITO, Flemish Institute for Technological Research, Unit Environmental Risk and Health, Mol, Belgium
| | - Marie-Gabrielle Zurich
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland.,Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Marcel Leist
- Center for Alternatives to Animal Testing, CAAT-Europe, University of Konstanz, Konstanz, Germany.,In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
39
|
Pamies D, Block K, Lau P, Gribaldo L, Pardo CA, Barreras P, Smirnova L, Wiersma D, Zhao L, Harris G, Hartung T, Hogberg HT. Rotenone exerts developmental neurotoxicity in a human brain spheroid model. Toxicol Appl Pharmacol 2018; 354:101-114. [PMID: 29428530 DOI: 10.1016/j.taap.2018.02.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 01/22/2018] [Accepted: 02/02/2018] [Indexed: 12/21/2022]
Abstract
Growing concern suggests that some chemicals exert (developmental) neurotoxicity (DNT and NT) and are linked to the increase in incidence of autism, attention deficit and hyperactivity disorders. The high cost of routine tests for DNT and NT assessment make it difficult to test the high numbers of existing chemicals. Thus, more cost effective neurodevelopmental models are needed. The use of induced pluripotent stem cells (iPSC) in combination with the emerging human 3D tissue culture platforms, present a novel tool to predict and study human toxicity. By combining these technologies, we generated multicellular brain spheroids (BrainSpheres) from human iPSC. The model has previously shown to be reproducible and recapitulates several neurodevelopmental features. Our results indicate, rotenone's toxic potency varies depending on the differentiation status of the cells, showing higher reactive oxygen species (ROS) and higher mitochondrial dysfunction during early than later differentiation stages. Immuno-fluorescence morphology analysis after rotenone exposure indicated dopaminergic-neuron selective toxicity at non-cytotoxic concentrations (1 μM), while astrocytes and other neuronal cell types were affected at (general) cytotoxic concentrations (25 μM). Omics analysis showed changes in key pathways necessary for brain development, indicating rotenone as a developmental neurotoxicant and show a possible link between previously shown effects on neurite outgrowth and presently observed effects on Ca2+ reabsorption, synaptogenesis and PPAR pathway disruption. In conclusion, our BrainSpheres model has shown to be a reproducible and novel tool to study neurotoxicity and developmental neurotoxicity. Results presented here support the idea that rotenone can potentially be a developmental neurotoxicant.
Collapse
Affiliation(s)
- David Pamies
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Katharina Block
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Pierre Lau
- European Commission, Joint Research Centre, European Reference Laboratory - European Centre for the Validation of Alternative Methods (EURL ECVAM), Via Enrico Fermi 2749, Ispra, VA 21027, Italy
| | - Laura Gribaldo
- European Commission, Joint Research Centre, European Reference Laboratory - European Centre for the Validation of Alternative Methods (EURL ECVAM), Via Enrico Fermi 2749, Ispra, VA 21027, Italy
| | - Carlos A Pardo
- Department of Neurology, Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD 21287, USA
| | - Paula Barreras
- Department of Neurology, Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD 21287, USA
| | - Lena Smirnova
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Daphne Wiersma
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Liang Zhao
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA; Bloomberg-Kimmel Institute for Cancer Immunotherapy, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, 650 Orleans Street, CRB1, Rm 464, Baltimore, MD 21287, USA
| | - Georgina Harris
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Thomas Hartung
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA; University of Konstanz, CAAT-Europe, Universitätsstr. 10, Konstanz 78464, Germany
| | - Helena T Hogberg
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|
40
|
An S, Zhao YF, Lü XY, Wang ZG. Quantitative evaluation of extrinsic factors influencing electrical excitability in neuronal networks: Voltage Threshold Measurement Method (VTMM). Neural Regen Res 2018; 13:1026-1035. [PMID: 29926830 PMCID: PMC6022462 DOI: 10.4103/1673-5374.233446] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The electrical excitability of neural networks is influenced by different environmental factors. Effective and simple methods are required to objectively and quantitatively evaluate the influence of such factors, including variations in temperature and pharmaceutical dosage. The aim of this paper was to introduce ‘the voltage threshold measurement method’, which is a new method using microelectrode arrays that can quantitatively evaluate the influence of different factors on the electrical excitability of neural networks. We sought to verify the feasibility and efficacy of the method by studying the effects of acetylcholine, ethanol, and temperature on hippocampal neuronal networks and hippocampal brain slices. First, we determined the voltage of the stimulation pulse signal that elicited action potentials in the two types of neural networks under normal conditions. Second, we obtained the voltage thresholds for the two types of neural networks under different concentrations of acetylcholine, ethanol, and different temperatures. Finally, we obtained the relationship between voltage threshold and the three influential factors. Our results indicated that the normal voltage thresholds of the hippocampal neuronal network and hippocampal slice preparation were 56 and 31 mV, respectively. The voltage thresholds of the two types of neural networks were inversely proportional to acetylcholine concentration, and had an exponential dependency on ethanol concentration. The curves of the voltage threshold and the temperature of the medium for the two types of neural networks were U-shaped. The hippocampal neuronal network and hippocampal slice preparations lost their excitability when the temperature of the medium decreased below 34 and 33°C or increased above 42 and 43°C, respectively. These results demonstrate that the voltage threshold measurement method is effective and simple for examining the performance/excitability of neuronal networks.
Collapse
Affiliation(s)
- Shuai An
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, Jiangsu Province, China
| | - Yong-Fang Zhao
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, Jiangsu Province, China
| | - Xiao-Ying Lü
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Zhi-Gong Wang
- Institute of RF- & OE-ICs, Southeast University, Nanjing; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
41
|
Simpson DA, Morrisroe E, McCoey JM, Lombard AH, Mendis DC, Treussart F, Hall LT, Petrou S, Hollenberg LCL. Non-Neurotoxic Nanodiamond Probes for Intraneuronal Temperature Mapping. ACS NANO 2017; 11:12077-12086. [PMID: 29111670 DOI: 10.1021/acsnano.7b04850] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Optical biomarkers have been used extensively for intracellular imaging with high spatial and temporal resolution. Extending the modality of these probes is a key driver in cell biology. In recent years, the nitrogen-vacancy (NV) center in nanodiamond has emerged as a promising candidate for bioimaging and biosensing with low cytotoxicity and stable photoluminescence. Here we study the electrophysiological effects of this quantum probe in primary cortical neurons. Multielectrode array recordings across five replicate studies showed no statistically significant difference in 25 network parameters when nanodiamonds are added at varying concentrations over various time periods, 12-36 h. The physiological validation motivates the second part of the study, which demonstrates how the quantum properties of these biomarkers can be used to report intracellular information beyond their location and movement. Using the optically detected magnetic resonance from the nitrogen-vacancy defects within the nanodiamonds we demonstrate enhanced signal-to-noise imaging and temperature mapping from thousands of nanodiamond probes simultaneously. This work establishes nanodiamonds as viable multifunctional intraneuronal sensors with nanoscale resolution, which may ultimately be used to detect magnetic and electrical activity at the membrane level in excitable cellular systems.
Collapse
Affiliation(s)
- David A Simpson
- School of Physics, University of Melbourne , Parkville, 3010, Australia
- Centre for Neural Engineering, University of Melbourne , Parkville, 3010, Australia
| | - Emma Morrisroe
- Florey Neuroscience Institute, University of Melbourne , Parkville, 3010, Australia
| | - Julia M McCoey
- School of Physics, University of Melbourne , Parkville, 3010, Australia
| | - Alain H Lombard
- Laboratoire Aimé Cotton, CNRS, Université Paris-Sud, ENS Paris-Saclay, Université Paris-Saclay , 91405 Orsay, France
| | - Dulini C Mendis
- Department of Mechanical Engineering, University of Melbourne , Parkville, VIC 3010, Australia
| | - François Treussart
- Laboratoire Aimé Cotton, CNRS, Université Paris-Sud, ENS Paris-Saclay, Université Paris-Saclay , 91405 Orsay, France
| | - Liam T Hall
- School of Physics, University of Melbourne , Parkville, 3010, Australia
| | - Steven Petrou
- Centre for Neural Engineering, University of Melbourne , Parkville, 3010, Australia
- Florey Neuroscience Institute, University of Melbourne , Parkville, 3010, Australia
- Centre for Integrated Brain Function, University of Melbourne , Parkville, 3010, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne , Parkville, 3010, Australia
| | - Lloyd C L Hollenberg
- School of Physics, University of Melbourne , Parkville, 3010, Australia
- Centre for Neural Engineering, University of Melbourne , Parkville, 3010, Australia
- Centre for Quantum Computation and Communication Technology, University of Melbourne , Parkville, 3052, Australia
| |
Collapse
|
42
|
Attoff K, Gliga A, Lundqvist J, Norinder U, Forsby A. Whole genome microarray analysis of neural progenitor C17.2 cells during differentiation and validation of 30 neural mRNA biomarkers for estimation of developmental neurotoxicity. PLoS One 2017; 12:e0190066. [PMID: 29261810 PMCID: PMC5738075 DOI: 10.1371/journal.pone.0190066] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/07/2017] [Indexed: 01/01/2023] Open
Abstract
Despite its high relevance, developmental neurotoxicity (DNT) is one of the least studied forms of toxicity. Current guidelines for DNT testing are based on in vivo testing and they require extensive resources. Transcriptomic approaches using relevant in vitro models have been suggested as a useful tool for identifying possible DNT-generating compounds. In this study, we performed whole genome microarray analysis on the murine progenitor cell line C17.2 following 5 and 10 days of differentiation. We identified 30 genes that are strongly associated with neural differentiation. The C17.2 cell line can be differentiated into a co-culture of both neurons and neuroglial cells, giving a more relevant picture of the brain than using neuronal cells alone. Among the most highly upregulated genes were genes involved in neurogenesis (CHRDL1), axonal guidance (BMP4), neuronal connectivity (PLXDC2), axonogenesis (RTN4R) and astrocyte differentiation (S100B). The 30 biomarkers were further validated by exposure to non-cytotoxic concentrations of two DNT-inducing compounds (valproic acid and methylmercury) and one neurotoxic chemical possessing a possible DNT activity (acrylamide). Twenty-eight of the 30 biomarkers were altered by at least one of the neurotoxic substances, proving the importance of these biomarkers during differentiation. These results suggest that gene expression profiling using a predefined set of biomarkers could be used as a sensitive tool for initial DNT screening of chemicals. Using a predefined set of mRNA biomarkers, instead of the whole genome, makes this model affordable and high-throughput. The use of such models could help speed up the initial screening of substances, possibly indicating alerts that need to be further studied in more sophisticated models.
Collapse
Affiliation(s)
- Kristina Attoff
- Department of Neurochemistry, Stockholm University, Stockholm, Sweden
- * E-mail:
| | - Anda Gliga
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jessica Lundqvist
- Department of Neurochemistry, Stockholm University, Stockholm, Sweden
- Swetox, Karolinska Institutet, Unit of Toxicology Sciences, Södertälje, Sweden
| | - Ulf Norinder
- Swetox, Karolinska Institutet, Unit of Toxicology Sciences, Södertälje, Sweden
| | - Anna Forsby
- Department of Neurochemistry, Stockholm University, Stockholm, Sweden
- Swetox, Karolinska Institutet, Unit of Toxicology Sciences, Södertälje, Sweden
| |
Collapse
|
43
|
Hong HJ, Koom WS, Koh WG. Cell Microarray Technologies for High-Throughput Cell-Based Biosensors. SENSORS (BASEL, SWITZERLAND) 2017; 17:E1293. [PMID: 28587242 PMCID: PMC5492771 DOI: 10.3390/s17061293] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 05/24/2017] [Accepted: 05/31/2017] [Indexed: 12/27/2022]
Abstract
Due to the recent demand for high-throughput cellular assays, a lot of efforts have been made on miniaturization of cell-based biosensors by preparing cell microarrays. Various microfabrication technologies have been used to generate cell microarrays, where cells of different phenotypes are immobilized either on a flat substrate (positional array) or on particles (solution or suspension array) to achieve multiplexed and high-throughput cell-based biosensing. After introducing the fabrication methods for preparation of the positional and suspension cell microarrays, this review discusses the applications of the cell microarray including toxicology, drug discovery and detection of toxic agents.
Collapse
Affiliation(s)
- Hye Jin Hong
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-749, Korea.
| | - Woong Sub Koom
- Department of Radiation Oncology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-749, Korea.
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-749, Korea.
| |
Collapse
|
44
|
Han Y, Li H, Lang Y, Zhao Y, Sun H, Zhang P, Ma X, Han J, Wang Q, Zhou J, Wang C. The Effects of Acute GABA Treatment on the Functional Connectivity and Network Topology of Cortical Cultures. Neurochem Res 2017; 42:1394-1402. [PMID: 28290133 DOI: 10.1007/s11064-017-2190-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/05/2016] [Accepted: 01/20/2017] [Indexed: 02/05/2023]
Abstract
γ-Aminobutyric acid (GABA) is an inhibitory transmitter, acting on receptor channels to reduce neuronal excitability in matured neural systems. However, electrophysiological responses of whole neuronal ensembles to the exposure to GABA are still unclear. We used micro-electrode arrays (MEAs) to study the effects of the increasing amount of GABA on functional network of cortical neural cultures. Then the recorded data were analyzed by the cross-covariance analysis and graph theory. Results showed that after the GABA treatment, the activity parameters of firing rate, bursting rate, bursting duration and network burst frequency in neural cultures decreased as expected. In addition, the functional connectivity also decreased in similarity, network density, and the size of the largest component. However, small-worldness was not found to be influenced by the acute GABA treatment. Our results support the position that using graph theory to evaluate the functional connectivity of neural cultures may enhance understanding of the pharmacological impact of neurotransmitters on neuronal networks.
Collapse
Affiliation(s)
- Yao Han
- Department of advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Hong Li
- Department of advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Yiran Lang
- Department of advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Yuwei Zhao
- Department of advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Hongji Sun
- Department of advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Peng Zhang
- Neural Interface& Rehabilitation Technology Research Center, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xuan Ma
- Neural Interface& Rehabilitation Technology Research Center, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jiuqi Han
- Department of advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Qiyu Wang
- Department of advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Jin Zhou
- Department of advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, People's Republic of China.
| | - Changyong Wang
- Department of advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, People's Republic of China.
| |
Collapse
|
45
|
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]
|
46
|
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
|
47
|
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]
|
48
|
Caputo L, Souza LF, Alloisio S, Cornara L, De Feo V. Coriandrum sativum and Lavandula angustifolia Essential Oils: Chemical Composition and Activity on Central Nervous System. Int J Mol Sci 2016; 17:ijms17121999. [PMID: 27916876 PMCID: PMC5187799 DOI: 10.3390/ijms17121999] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/04/2016] [Accepted: 11/17/2016] [Indexed: 11/16/2022] Open
Abstract
The aims of this study are to determine the chemical composition of Lavandula angustifolia Mill. and Coriandrum sativum L. essential oils, to evaluate their cytotoxic effects in SH-SY5Y human neuroblastoma cells, to investigate whether an alteration of adenylate cyclase 1 (ADCY1) and of extracellular signal-regulated kinase (ERK) expression can take part in the molecular mechanisms of the essential oils, and to study their possible neuronal electrophysiological effects. The essential oils were obtained by hydrodistillation, and studied by GC and GC-MS. In the oils from L. angustifolia and C. sativum, linalool was the main component (33.1% and 67.8%, respectively). SH-SY5Y cells were incubated with different concentrations of essential oils and of linalool. Cell viability and effects on ADCY1 and ERK expression were analyzed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide MTT and Western blotting, respectively. Variation in cellular electrophysiology was studied in primary cultures of rat cortical neurons with a multi-electrode array (MEA)-based approach. The essential oils and linalool revealed different cytotoxic activities. Linalool inhibited ADCY1 and ERK expression. Neuronal networks subjected to L. angustifolia and C. sativum essential oils showed a concentration-dependent inhibition of spontaneous electrical activity.
Collapse
Affiliation(s)
- Lucia Caputo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano (Salerno), Italy.
| | - Lucéia Fátima Souza
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano (Salerno), Italy.
- Department of Agronomy, University of Rio Grande do Sul (UFRGS), 91501-970 Porto Alegre, Brazil.
| | | | - Laura Cornara
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, University of Genoa, Corso Europa 26, 16132 Genoa, Italy.
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano (Salerno), Italy.
| |
Collapse
|
49
|
Wanke E, Gullo F, Dossi E, Valenza G, Becchetti A. Neuron-glia cross talk revealed in reverberating networks by simultaneous extracellular recording of spikes and astrocytes' glutamate transporter and K+ currents. J Neurophysiol 2016; 116:2706-2719. [PMID: 27683885 PMCID: PMC5133298 DOI: 10.1152/jn.00509.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 09/22/2016] [Indexed: 01/11/2023] Open
Abstract
In neocortex networks, we simultaneously captured spikes and the slower astrocytes' K+and glutamate transporter (GluT) currents with the use of individual MEA electrodes. Inward and outward K+currents in different regions of the glial syncytium suggested that spatial buffering was operant. Moreover, in organotypic slices from ventral tegmental area and prefrontal cortex, the large GluT current amplitudes allowed to measure transporter currents with a single electrode. Our method allows direct study of the dynamic interplay of different cell types in excitable and nonexcitable tissue. Astrocytes uptake synaptically released glutamate with electrogenic transporters (GluT) and buffer the spike-dependent extracellular K+ excess with background K+ channels. We studied neuronal spikes and the slower astrocytic signals on reverberating neocortical cultures and organotypic slices from mouse brains. Spike trains and glial responses were simultaneously captured from individual sites of multielectrode arrays (MEA) by splitting the recorded traces into appropriate filters and reconstructing the original signal by deconvolution. GluT currents were identified by using dl-threo-β-benzyloxyaspartate (TBOA). K+ currents were blocked by 30 μM Ba2+, suggesting a major contribution of inwardly rectifying K+ currents. Both types of current were tightly correlated with the spike rate, and their astrocytic origin was tested in primary cultures by blocking glial proliferation with cytosine β-d-arabinofuranoside (AraC). The spike-related, time-locked inward and outward K+ currents in different regions of the astrocyte syncytium were consistent with the assumptions of the spatial K+ buffering model. In organotypic slices from ventral tegmental area and prefrontal cortex, the GluT current amplitudes exceeded those observed in primary cultures by several orders of magnitude, which allowed to directly measure transporter currents with a single electrode. Simultaneously measuring cell signals displaying widely different amplitudes and kinetics will help clarify the neuron-glia interplay and make it possible to follow the cross talk between different cell types in excitable as well as nonexcitable tissue.
Collapse
Affiliation(s)
- Enzo Wanke
- Department of Biotechnologies and Biosciences and Milan Center For Neuroscience (NeuroMI), University of Milano-Bicocca, Milan, Italy; and
| | - Francesca Gullo
- Department of Biotechnologies and Biosciences and Milan Center For Neuroscience (NeuroMI), University of Milano-Bicocca, Milan, Italy; and
| | - Elena Dossi
- Department of Biotechnologies and Biosciences and Milan Center For Neuroscience (NeuroMI), University of Milano-Bicocca, Milan, Italy; and
| | - Gaetano Valenza
- Research Centre "E. Piaggio" and Department of Information Engineering, School of Engineering, University of Pisa, Pisa, Italy
| | - Andrea Becchetti
- Department of Biotechnologies and Biosciences and Milan Center For Neuroscience (NeuroMI), University of Milano-Bicocca, Milan, Italy; and
| |
Collapse
|
50
|
Hiolski EM, Ito S, Beggs JM, Lefebvre KA, Litke AM, Smith DR. Domoic acid disrupts the activity and connectivity of neuronal networks in organotypic brain slice cultures. Neurotoxicology 2016; 56:215-224. [PMID: 27506300 DOI: 10.1016/j.neuro.2016.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/04/2016] [Accepted: 08/05/2016] [Indexed: 12/22/2022]
Abstract
Domoic acid is a neurotoxin produced by algae and is found in seafood during harmful algal blooms. As a glutamate agonist, domoic acid inappropriately stimulates excitatory activity in neurons. At high doses, this leads to seizures and brain lesions, but it is unclear how lower, asymptomatic exposures disrupt neuronal activity. Domoic acid has been detected in an increasing variety of species across a greater geographical range than ever before, making it critical to understand the potential health impacts of low-level exposure on vulnerable marine mammal and human populations. To determine whether prolonged domoic acid exposure altered neuronal activity in hippocampal networks, we used a custom-made 512 multi-electrode array with high spatial and temporal resolution to record extracellular potentials (spikes) in mouse organotypic brain slice cultures. We identified individual neurons based on spike waveform and location, and measured the activity and functional connectivity within the neuronal networks of brain slice cultures. Domoic acid exposure significantly altered neuronal spiking activity patterns, and increased functional connectivity within exposed cultures, in the absence of overt cellular or neuronal toxicity. While the overall spiking activity of neurons in domoic acid-exposed cultures was comparable to controls, exposed neurons spiked significantly more often in bursts. We also identified a subset of neurons that were electrophysiologically silenced in exposed cultures, and putatively identified those neurons as fast-spiking inhibitory neurons. These results provide evidence that domoic acid affects neuronal activity in the absence of cytotoxicity, and suggest that neurodevelopmental exposure to domoic acid may alter neurological function in the absence of clinical symptoms.
Collapse
Affiliation(s)
- E M Hiolski
- Department of Microbiology & Environmental Toxicology, University of California, Santa Cruz, CA, USA
| | - S Ito
- Santa Cruz Institute for Particle Physics, University of California, Santa Cruz, CA, USA
| | - J M Beggs
- Department of Physics, Indiana University, Bloomington, IN, USA
| | - K A Lefebvre
- Northwest Fisheries Science Center, NOAA Fisheries, Seattle, WA, USA
| | - A M Litke
- Santa Cruz Institute for Particle Physics, University of California, Santa Cruz, CA, USA
| | - D R Smith
- Department of Microbiology & Environmental Toxicology, University of California, Santa Cruz, CA, USA
| |
Collapse
|