1
|
Meng C, Lu Y, Huang Y, Lü X. Electrical excitability of neuronal networks based on the voltage threshold of electrical stimulation. Sci Rep 2024; 14:31573. [PMID: 39738192 DOI: 10.1038/s41598-024-78142-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/29/2024] [Indexed: 01/01/2025] Open
Abstract
Microelectrode arrays (MEAs) have been widely used in studies on the electrophysiological features of neuronal networks. In classic MEA experiments, spike or burst rates and spike waveforms are the primary characteristics used to evaluate the neuronal network excitability. Here, we introduced a new method to assess the excitability using the voltage threshold of electrical stimulation. We tested the stability of the voltage threshold during the experiment and demonstrated the reliability of our method by examining the effect of Ni2+ on neocortical neuronal networks of acute brain slices from rats. Moreover, we compared our new method with the spontaneous activity analysis, which is one of the most commonly used methods in protocols for large-scale drug screening with MEA; our new method performed better in the experiments investigating the neocortical neuronal network excitability after the application of Ni2+. Based on the results from our study, our new method has great potential for use in large-scale screening of drugs.
Collapse
Affiliation(s)
- Chen Meng
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yang Lu
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yan Huang
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xiaoying Lü
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
2
|
Ku RY, Bansal A, Dutta DJ, Yamashita S, Peloquin J, Vu DN, Shen Y, Uchida T, Torii M, Hashimoto-Torii K. Evaluating chemical effects on human neural cells through calcium imaging and deep learning. iScience 2024; 27:111298. [PMID: 39634567 PMCID: PMC11616611 DOI: 10.1016/j.isci.2024.111298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/24/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024] Open
Abstract
New substances intended for human consumption must undergo extensive preclinical safety pharmacology testing prior to approval. These tests encompass the evaluation of effects on the central nervous system, which is highly sensitive to chemical substances. With the growing understanding of the species-specific characteristics of human neural cells and advancements in machine learning technology, the development of effective and efficient methods for the initial screening of chemical effects on human neural function using machine learning platforms is anticipated. In this study, we employed a deep learning model to analyze calcium dynamics in human-induced pluripotent stem cell-derived neural progenitor cells, which were exposed to various concentrations of four representative chemicals. We report that this approach offers a reliable and concise method for quantitatively classifying the effects of chemical exposures and predicting potential harm to human neural cells.
Collapse
Affiliation(s)
- Ray Yueh Ku
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Hospital, Washington, DC 20010, USA
| | - Ankush Bansal
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Hospital, Washington, DC 20010, USA
| | - Dipankar J. Dutta
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Hospital, Washington, DC 20010, USA
| | - Satoshi Yamashita
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Hospital, Washington, DC 20010, USA
| | - John Peloquin
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Hospital, Washington, DC 20010, USA
| | - Diana N. Vu
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Hospital, Washington, DC 20010, USA
| | - Yubing Shen
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Hospital, Washington, DC 20010, USA
| | - Tomoki Uchida
- Novel Business Development Department, Suntory Global Innovation Center Limited, 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan
| | - Masaaki Torii
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Hospital, Washington, DC 20010, USA
- Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children’s Research Institute, Children’s National Hospital, Washington, DC 20010, USA
- Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| |
Collapse
|
3
|
van Melis LVJ, Peerdeman AM, González CA, van Kleef RGDM, Wopken JP, Westerink RHS. Effects of chronic insecticide exposure on neuronal network development in vitro in rat cortical cultures. Arch Toxicol 2024; 98:3837-3857. [PMID: 39162819 PMCID: PMC11489184 DOI: 10.1007/s00204-024-03840-0] [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: 05/16/2024] [Accepted: 08/08/2024] [Indexed: 08/21/2024]
Abstract
Developmental exposure to carbamates, organophosphates, and pyrethroids has been associated with impaired neurodevelopmental outcomes. Sex-specific differences following chronic insecticide exposure are rather common in vivo. Therefore, we assessed the chronic effects of in vitro exposure to different carbamates (carbaryl, methomyl and aldicarb), organophosphates [chlorpyrifos (CPF), chlorpyrifos-oxon (CPO), and 3,5,6,trichloropyridinol (TCP)], and pyrethroids [permethrin, alpha-cypermethrin and 3-phenoxy benzoic acid (3-PBA)] on neuronal network development in sex-separated rat primary cortical cultures using micro-electrode array (MEA) recordings. Our results indicate that exposure for 1 week to carbaryl inhibited neurodevelopment in male cultures, while a hyperexcitation was observed in female cultures. Methomyl and aldicarb evoked a hyperexcitation after 2 weeks of exposure, which was more pronounced in female cultures. In contrast to acute MEA results, exposure to ≥ 10 µM CPF caused hyperexcitation in both sexes after 10 days. Interestingly, exposure to 10 µM CPO induced a clear hyperexcitation after 10 days of exposure in male but not female cultures. Exposure to 100 µM CPO strongly inhibited neuronal development. Exposure to the type I pyrethroid permethrin resulted in a hyperexcitation at 10 µM and a decrease in neuronal development at 100 µM. In comparison, exposure to ≥ 10 µM of the type II pyrethroid alpha-cypermethrin decreased neuronal development. In female but not in male cultures, exposure to 1 and 10 µM permethrin changed (network) burst patterns, with female cultures having shorter (network) bursts with fewer spikes per (network) burst. Together, these results show that MEA recordings are suitable for measuring sex-specific developmental neurotoxicity in vitro. Additionally, pyrethroid exposure induced effects on neuronal network development at human-relevant concentrations. Finally, chronic exposure has different effects on neuronal functioning compared to acute exposure, highlighting the value of both exposure paradigms.
Collapse
Affiliation(s)
- Lennart V J van Melis
- Neurotoxicology Research Group, Division of Toxicology, Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences (IRAS), Utrecht University, P.O. Box 80.177, NL-3508 TD, Utrecht, The Netherlands
| | - Anneloes M Peerdeman
- Neurotoxicology Research Group, Division of Toxicology, Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences (IRAS), Utrecht University, P.O. Box 80.177, NL-3508 TD, Utrecht, The Netherlands
| | - Celia Arenas González
- Neurotoxicology Research Group, Division of Toxicology, Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences (IRAS), Utrecht University, P.O. Box 80.177, NL-3508 TD, Utrecht, The Netherlands
| | - Regina G D M van Kleef
- Neurotoxicology Research Group, Division of Toxicology, Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences (IRAS), Utrecht University, P.O. Box 80.177, NL-3508 TD, Utrecht, The Netherlands
| | - J Pepijn Wopken
- Neurotoxicology Research Group, Division of Toxicology, Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences (IRAS), Utrecht University, P.O. Box 80.177, NL-3508 TD, Utrecht, The Netherlands
| | - Remco H S Westerink
- Neurotoxicology Research Group, Division of Toxicology, Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences (IRAS), Utrecht University, P.O. Box 80.177, NL-3508 TD, Utrecht, The Netherlands.
| |
Collapse
|
4
|
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
|
5
|
Malepathirana T, Senanayake D, Gautam V, Engel M, Balez R, Lovelace MD, Sundaram G, Heng B, Chow S, Marquis C, Guillemin GJ, Brew B, Jagadish C, Ooi L, Halgamuge S. Visualization of incrementally learned projection trajectories for longitudinal data. Sci Rep 2024; 14:13558. [PMID: 38866809 PMCID: PMC11169470 DOI: 10.1038/s41598-024-63511-z] [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: 09/27/2023] [Accepted: 05/29/2024] [Indexed: 06/14/2024] Open
Abstract
Longitudinal studies that continuously generate data enable the capture of temporal variations in experimentally observed parameters, facilitating the interpretation of results in a time-aware manner. We propose IL-VIS (incrementally learned visualizer), a new machine learning pipeline that incrementally learns and visualizes a progression trajectory representing the longitudinal changes in longitudinal studies. At each sampling time point in an experiment, IL-VIS generates a snapshot of the longitudinal process on the data observed thus far, a new feature that is beyond the reach of classical static models. We first verify the utility and correctness of IL-VIS using simulated data, for which the true progression trajectories are known. We find that it accurately captures and visualizes the trends and (dis)similarities between high-dimensional progression trajectories. We then apply IL-VIS to longitudinal multi-electrode array data from brain cortical organoids when exposed to different levels of quinolinic acid, a metabolite contributing to many neuroinflammatory diseases including Alzheimer's disease, and its blocking antibody. We uncover valuable insights into the organoids' electrophysiological maturation and response patterns over time under these conditions.
Collapse
Affiliation(s)
- Tamasha Malepathirana
- Department of Mechanical Engineering, University of Melbourne, Melbourne, 3010, VIC, Australia
| | - Damith Senanayake
- Department of Mechanical Engineering, University of Melbourne, Melbourne, 3010, VIC, Australia
| | - Vini Gautam
- School of Chemical and Biomedical Engineering, University of Melbourne, Melbourne, 3010, VIC, Australia
- Centre for Nano Science and Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Martin Engel
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, 2522, NSW, Australia
| | - Rachelle Balez
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, 2522, NSW, Australia
| | - Michael D Lovelace
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St. Vincent's Centre for Applied Medical Research, 405 Liverpool St., Darlinghurst, Sydney, 2010, NSW, Australia
- School of Clinical Medicine, UNSW Medicine and Health, St. Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, 2010, NSW, Australia
| | | | - Benjamin Heng
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, 2109, NSW, Australia
| | - Sharron Chow
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, 2109, NSW, Australia
| | - Christopher Marquis
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, 2052, NSW, Australia
| | - Gilles J Guillemin
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St. Vincent's Centre for Applied Medical Research, 405 Liverpool St., Darlinghurst, Sydney, 2010, NSW, Australia
- IPB University, Bogor, Indonesia
| | - Bruce Brew
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St. Vincent's Centre for Applied Medical Research, 405 Liverpool St., Darlinghurst, Sydney, 2010, NSW, Australia
- School of Clinical Medicine, UNSW Medicine and Health, St. Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, 2010, NSW, Australia
- Departments of Neurology and Immunology, St. Vincent's Hospital, Sydney, 2010, NSW, Australia
| | - Chennupati Jagadish
- Research School of Physics, Australian National University, Canberra, 2601, ACT, Australia
| | - Lezanne Ooi
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, 2522, NSW, Australia.
| | - Saman Halgamuge
- Department of Mechanical Engineering, University of Melbourne, Melbourne, 3010, VIC, Australia.
| |
Collapse
|
6
|
Martin MM, Carpenter AF, Shafer TJ, Paul Friedman K, Carstens KE. Chemical effects on neural network activity: Comparison of acute versus network formation exposure in microelectrode array assays. Toxicology 2024; 505:153842. [PMID: 38788893 DOI: 10.1016/j.tox.2024.153842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/08/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
New approach methodologies (NAMs) can address information gaps on potential neurotoxicity or developmental neurotoxicity hazard for data-poor chemicals. Two assays have been previously developed using microelectrode arrays (MEA), a technology which measures neural activity. The MEA acute network function assay (AcN) uses dissociated rat cortical cells cultured at postnatal day 0 and evaluates network activity during a 40-minute chemical exposure on day in vitro (DIV)13 or 15. In contrast, the MEA network formation assay (NFA) uses a developmental exposure paradigm spanning DIV0 through DIV12. Measures of network activity over time at DIV5, 7, 9, and 12 in the NFA are reduced to an estimated area under the curve to facilitate concentration-response evaluation. Here, we evaluated the hypothesis that chemicals with effects in the AcN also perturb the NFA by examining quantitative and qualitative concordance between assays. Out of 243 chemicals screened in both assays, we observed 70.3% concordance between the AcN and NFA after eliminating activity inferred to be cytotoxic (selective activity), with the majority of discordance explained by chemicals that altered selective activity in the AcN but not NFA. The NFA detected more active chemicals when evaluating activity associated with cytotoxicity. Median potency values were lower in the NFA compared to the AcN, but within-chemical potency values were not uniformly lower in the NFA than the AcN. Lastly, the AcN and NFA captured unique bioactivity fingerprints; the AcN was more informative for identifying chemicals with a shared mode of action, while the NFA provided information relevant to developmental exposure. Taken together, this analysis provides a rationale for using both approaches for chemical evaluation with consideration of the context of use, such as screening/ prioritization, hazard identification, or to address questions regarding biological mechanism or function.
Collapse
Affiliation(s)
- Melissa M Martin
- Computational Toxicology & Bioinformatics Branch, Biomolecular and Computational Toxicology Division, CCTE/ORD, US. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Amy F Carpenter
- Computational Toxicology & Bioinformatics Branch, Biomolecular and Computational Toxicology Division, CCTE/ORD, US. Environmental Protection Agency, Research Triangle Park, NC 27711, USA; Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Timothy J Shafer
- Computational Toxicology & Bioinformatics Branch, Biomolecular and Computational Toxicology Division, CCTE/ORD, US. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Katie Paul Friedman
- Computational Toxicology & Bioinformatics Branch, Biomolecular and Computational Toxicology Division, CCTE/ORD, US. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Kelly E Carstens
- Computational Toxicology & Bioinformatics Branch, Biomolecular and Computational Toxicology Division, CCTE/ORD, US. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| |
Collapse
|
7
|
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
|
8
|
Gerber LS, de Leijer DCA, Rujas Arranz A, Lehmann JMML, Verheul ME, Cassee FR, Westerink RHS. In vitro neurotoxicity of particles from diesel and biodiesel fueled engines following direct and simulated inhalation exposure. ENVIRONMENT INTERNATIONAL 2024; 184:108481. [PMID: 38330748 DOI: 10.1016/j.envint.2024.108481] [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: 11/24/2023] [Revised: 01/17/2024] [Accepted: 02/02/2024] [Indexed: 02/10/2024]
Abstract
Combustion-derived particulate matter (PM) is a major source of air pollution. Efforts to reduce diesel engine emission include the application of biodiesel. However, while urban PM exposure has been linked to adverse brain effects, little is known about the direct effects of PM from regular fossil diesel (PMDEP) and biodiesel (PMBIO) on neuronal function. Furthermore, it is unknown to what extent the PM-induced effects in the lung (e.g., inflammation) affect the brain. This in vitro study investigates direct and indirect toxicity of PMDEP and PMBIO on the lung and brain and compared it with effects of clean carbon particles (CP). PM were generated using a common rail diesel engine. CP was sampled from a spark generator. First, effects of 48 h exposure to PM and CP (1.2-3.9 µg/cm2) were assessed in an in vitro lung model (air-liquid interface co-culture of Calu-3 and THP1 cells) by measuring cell viability, cytotoxicity, barrier function, inflammation, and oxidative and cell stress. None of the exposures caused clear adverse effects and only minor changes in gene expression were observed. Next, the basal medium was collected for subsequent simulated inhalation exposure of rat primary cortical cells. Neuronal activity, recorded using microelectrode arrays (MEA), was increased after acute (0.5 h) simulated inhalation exposure. In contrast, direct exposure to PMDEP and PMBIO (1-100 µg/mL; 1.2-119 µg/cm2) reduced neuronal activity after 24 h with lowest observed effect levels of respectively 10 µg/mL and 30 µg/mL, indicating higher neurotoxic potency of PMDEP, whereas neuronal activity remained unaffected following CP exposure. These findings indicate that combustion-derived PM potently inhibit neuronal function following direct exposure, while the lung serves as a protective barrier. Furthermore, PMDEP exhibit a higher direct neurotoxic potency than PMBIO, and the data suggest that the neurotoxic effects is caused by adsorbed chemicals rather than the pure carbon core.
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
| | - Dirk C A de Leijer
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Andrea Rujas Arranz
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Jonas M M L Lehmann
- 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
| | - Meike E Verheul
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, 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 H S Westerink
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
9
|
Zips S, Huang B, Hotte S, Hiendlmeier L, Wang C, Rajamani K, Buriez O, Al Boustani G, Chen Y, Wolfrum B, Yamada A. Aerosol Jet-Printed High-Aspect Ratio Micro-Needle Electrode Arrays Applied for Human Cerebral Organoids and 3D Neurospheroid Networks. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37469180 DOI: 10.1021/acsami.3c06210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
The human brain is a complex and poorly accessible organ. Thus, new tools are required for studying the neural function in a controllable environment that preserves multicellular interaction and neuronal wiring. In particular, high-throughput methods that alleviate the need for animal experiments are essential for future studies. Recent developments of induced pluripotent stem cell technologies have enabled in vitro modeling of the human brain by creating three-dimensional brain tissue mimic structures. To leverage these new technologies, a systematic and versatile approach for evaluating neuronal activity at larger tissue depths within the regime of tens to hundreds of micrometers is required. Here, we present an aerosol-jet- and inkjet-printing-based method to fabricate microelectrode arrays, equipped with high-aspect ratio μ-needle electrodes that penetrate 3D neural network assemblies. The arrays have been successfully applied for electrophysiological recordings on interconnected neurospheroids formed on an engineered substrate and on cerebral organoids, both derived from human induced pluripotent stem cells.
Collapse
Affiliation(s)
- Sabine Zips
- Neuroelectronics─Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Boltzmannstr. 11, 85748 Garching, Germany
| | - Boxin Huang
- PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Salammbô Hotte
- PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Lukas Hiendlmeier
- Neuroelectronics─Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Boltzmannstr. 11, 85748 Garching, Germany
| | - Chen Wang
- Neuroelectronics─Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Boltzmannstr. 11, 85748 Garching, Germany
| | - Karthyayani Rajamani
- PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Olivier Buriez
- PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - George Al Boustani
- Neuroelectronics─Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Boltzmannstr. 11, 85748 Garching, Germany
| | - Yong Chen
- PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Bernhard Wolfrum
- Neuroelectronics─Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Boltzmannstr. 11, 85748 Garching, Germany
| | - Ayako Yamada
- PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| |
Collapse
|
10
|
Pergolizzi J, Varrassi G. The Emerging Role of Sigma Receptors in Pain Medicine. Cureus 2023; 15:e42626. [PMID: 37641763 PMCID: PMC10460634 DOI: 10.7759/cureus.42626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/25/2023] [Indexed: 08/31/2023] Open
Abstract
Sigma receptors are protein chaperones with the unexpected characteristic of being activated by ligand binding. As such, they represent intriguing new targets for potential drug development. As a protein chaperone, these "receptors" escort proteins from the endoplasmic reticulum to their destinations and act to correct misfolded proteins. The two subtypes of sigma receptors, named σ1 and σ2, are markedly distinct from each other. Agonists and antagonists at these receptors show promise as new drug targets, addressing a range of diseases including neurodegenerative disorders, cancer, and cardiac disorders, and may also be analgesic agents and rehabilitation drugs for opioid use disorder. As an analgesic, sigma receptors seem to be more effective in treating neuropathic than nociceptive pain. New bifunctional compounds are being developed with opioids, because agents targeting sigma receptors may have an opioid-sparing effect. The pipeline of agents based on the sigma receptors is long and may treat things from Fragile X syndrome to Parkinson's disease to Huntington's disease to cancer. A novel agent ADV502 acts as a high-affinity σ1 antagonist and partial agonist at the µ-opioid receptor and may be an important agent both for the treatment of neuropathic cancer pain and for rehabilitation of opioid use disorder. Since there has been little recent innovation in pain medicine regarding new compounds and drug targets, drugs that affect the sigma receptor system seem promising and encouraging.
Collapse
|
11
|
Poggio F, Brofiga M, Callegari F, Tedesco M, Massobrio P. Developmental conditions and culture medium influence the neuromodulated response of in vitro cortical networks. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38083479 DOI: 10.1109/embc40787.2023.10340933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Goal of this work is to show how the developmental conditions of in vitro neuronal networks influence the effect of drug delivery. The proposed experimental neuronal model consists of dissociated cortical neurons plated to Micro-Electrode Arrays (MEAs) and grown according to different conditions (i.e., by varying both the adopted culture medium and the number of days needed to let the network grow before performing the chemical modulation). We delivered rising amount of bicuculline (BIC), a competitive antagonist of GABAA receptors, and we computed the firing rate dose-response curve for each culture. We found that networks matured in BrainPhys for 18 days in vitro exhibited a decreasing firing trend as a function of the BIC concentration, quantified by an average IC50 (i.e., half maximal inhibitory concentration) of 4.64 ± 4.02 µM. On the other hand, both cultures grown in the same medium for 11 days, and ones matured in Neurobasal for 18 days displayed an increasing firing rate when rising amounts of BIC were delivered, characterized by average EC50 values (i.e., half maximal excitatory concentration) of 0.24 ± 0.05 µM and 0.59 ± 0.46 µM, respectively.Clinical Relevance- This research proves the relevance of the experimental factors that can influence the network development as key variables when developing a neuronal model to conduct drug delivery in vitro, simulating the in vivo environment. Our findings suggest that not considering the consequences of the chosen growing conditions when performing in vitro pharmacological studies could lead to incomplete predictions of the chemically induced alterations.
Collapse
|
12
|
McDonald M, Sebinger D, Brauns L, Gonzalez-Cano L, Menuchin-Lasowski Y, Mierzejewski M, Psathaki OE, Stumpf A, Wickham J, Rauen T, Schöler H, Jones PD. A mesh microelectrode array for non-invasive electrophysiology within neural organoids. Biosens Bioelectron 2023; 228:115223. [PMID: 36931193 DOI: 10.1016/j.bios.2023.115223] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/13/2023]
Abstract
Organoids are emerging in vitro models of human physiology. Neural models require the evaluation of functional activity of single cells and networks, which is commonly measured by microelectrode arrays. The characteristics of organoids clash with existing in vitro or in vivo microelectrode arrays. With inspiration from implantable mesh electronics and growth of organoids on polymer scaffolds, we fabricated suspended hammock-like mesh microelectrode arrays for neural organoids. We have demonstrated the growth of organoids enveloping these meshes and the culture of organoids on meshes for up to one year. Furthermore, we present proof-of-principle recordings of spontaneous electrical activity across the volume of an organoid. Our concept enables a new class of microelectrode arrays for in vitro models of three-dimensional electrically active tissue.
Collapse
Affiliation(s)
- Matthew McDonald
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770, Reutlingen, Germany; Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany
| | - David Sebinger
- Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany
| | - Lisa Brauns
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770, Reutlingen, Germany; Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany
| | - Laura Gonzalez-Cano
- Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany
| | | | - Michael Mierzejewski
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770, Reutlingen, Germany; Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany
| | - Olympia-Ekaterini Psathaki
- Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany; University of Osnabrück, CellNanOs (Center of Cellular Nanoanalytics), Integrated Bioimaging Facility iBiOs, Barbarastr. 11, 49076, Osnabrück, Germany
| | - Angelika Stumpf
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770, Reutlingen, Germany
| | - Jenny Wickham
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770, Reutlingen, Germany
| | - Thomas Rauen
- Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany.
| | - Hans Schöler
- Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149, Münster, Germany
| | - Peter D Jones
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770, Reutlingen, Germany.
| |
Collapse
|
13
|
Girardin S, Ihle SJ, Menghini A, Krubner M, Tognola L, Duru J, Fruh I, Müller M, Ruff T, Vörös J. Engineering circuits of human iPSC-derived neurons and rat primary glia. Front Neurosci 2023; 17:1103437. [PMID: 37250404 PMCID: PMC10213452 DOI: 10.3389/fnins.2023.1103437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 04/18/2023] [Indexed: 05/31/2023] Open
Abstract
Novel in vitro platforms based on human neurons are needed to improve early drug testing and address the stalling drug discovery in neurological disorders. Topologically controlled circuits of human induced pluripotent stem cell (iPSC)-derived neurons have the potential to become such a testing system. In this work, we build in vitro co-cultured circuits of human iPSC-derived neurons and rat primary glial cells using microfabricated polydimethylsiloxane (PDMS) structures on microelectrode arrays (MEAs). Our PDMS microstructures are designed in the shape of a stomach, which guides axons in one direction and thereby facilitates the unidirectional flow of information. Such circuits are created by seeding either dissociated cells or pre-aggregated spheroids at different neuron-to-glia ratios. Furthermore, an antifouling coating is developed to prevent axonal overgrowth in undesired locations of the microstructure. We assess the electrophysiological properties of different types of circuits over more than 50 days, including their stimulation-induced neural activity. Finally, we demonstrate the inhibitory effect of magnesium chloride on the electrical activity of our iPSC circuits as a proof-of-concept for screening of neuroactive compounds.
Collapse
Affiliation(s)
- Sophie Girardin
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Department of Electrical Engineering and Information Technology, University and ETH Zürich, Zürich, Switzerland
| | - Stephan J. Ihle
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Department of Electrical Engineering and Information Technology, University and ETH Zürich, Zürich, Switzerland
| | - Arianna Menghini
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Department of Electrical Engineering and Information Technology, University and ETH Zürich, Zürich, Switzerland
| | - Magdalena Krubner
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Department of Electrical Engineering and Information Technology, University and ETH Zürich, Zürich, Switzerland
| | - Leonardo Tognola
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Department of Electrical Engineering and Information Technology, University and ETH Zürich, Zürich, Switzerland
| | - Jens Duru
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Department of Electrical Engineering and Information Technology, University and ETH Zürich, Zürich, Switzerland
| | - Isabelle Fruh
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Matthias Müller
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Tobias Ruff
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Department of Electrical Engineering and Information Technology, University and ETH Zürich, Zürich, Switzerland
| | - János Vörös
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Department of Electrical Engineering and Information Technology, University and ETH Zürich, Zürich, Switzerland
| |
Collapse
|
14
|
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
|
15
|
Hartmann J, Henschel N, Bartmann K, Dönmez A, Brockerhoff G, Koch K, Fritsche E. Molecular and Functional Characterization of Different BrainSphere Models for Use in Neurotoxicity Testing on Microelectrode Arrays. Cells 2023; 12:cells12091270. [PMID: 37174670 PMCID: PMC10177384 DOI: 10.3390/cells12091270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/14/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
The currently accepted methods for neurotoxicity (NT) testing rely on animal studies. However, high costs and low testing throughput hinder their application for large numbers of chemicals. To overcome these limitations, in vitro methods are currently being developed based on human-induced pluripotent stem cells (hiPSC) that allow higher testing throughput at lower costs. We applied six different protocols to generate 3D BrainSphere models for acute NT evaluation. These include three different media for 2D neural induction and two media for subsequent 3D differentiation resulting in self-organized, organotypic neuron/astrocyte microtissues. All induction protocols yielded nearly 100% NESTIN-positive hiPSC-derived neural progenitor cells (hiNPCs), though with different gene expression profiles concerning regional patterning. Moreover, gene expression and immunocytochemistry analyses revealed that the choice of media determines neural differentiation patterns. On the functional level, BrainSpheres exhibited different levels of electrical activity on microelectrode arrays (MEA). Spike sorting allowed BrainSphere functional characterization with the mixed cultures consisting of GABAergic, glutamatergic, dopaminergic, serotonergic, and cholinergic neurons. A test method for acute NT testing, the human multi-neurotransmitter receptor (hMNR) assay, was proposed to apply such MEA-based spike sorting. These models are promising tools not only in toxicology but also for drug development and disease modeling.
Collapse
Affiliation(s)
- Julia Hartmann
- IUF-Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
| | - Noah Henschel
- IUF-Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
| | - Kristina Bartmann
- IUF-Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
- DNTOX GmbH, Gurlittstraße 53, 40223 Düsseldorf, Germany
| | - Arif Dönmez
- IUF-Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
- DNTOX GmbH, Gurlittstraße 53, 40223 Düsseldorf, Germany
| | - Gabriele Brockerhoff
- IUF-Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
| | - Katharina Koch
- IUF-Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
- DNTOX GmbH, Gurlittstraße 53, 40223 Düsseldorf, Germany
| | - Ellen Fritsche
- IUF-Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
- DNTOX GmbH, Gurlittstraße 53, 40223 Düsseldorf, Germany
- Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany
| |
Collapse
|
16
|
Enterovirus D-68 Infection of Primary Rat Cortical Neurons: Entry, Replication, and Functional Consequences. mBio 2023; 14:e0024523. [PMID: 36877033 PMCID: PMC10127580 DOI: 10.1128/mbio.00245-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Enterovirus D68 (EV-D68) is an emerging pathogen associated with mild to severe respiratory disease. Since 2014, EV-D68 is also linked to acute flaccid myelitis (AFM), causing paralysis and muscle weakness in children. However, it remains unclear whether this is due to an increased pathogenicity of contemporary EV-D68 clades or increased awareness and detection of this virus. Here, we describe an infection model of primary rat cortical neurons to study the entry, replication, and functional consequences of different EV-D68 strains, including historical and contemporary strains. We demonstrate that sialic acids are important (co)receptors for infection of both neurons and respiratory epithelial cells. Using a collection of glycoengineered isogenic HEK293 cell lines, we show that sialic acids on either N-glycans or glycosphingolipids can be used for infection. Additionally, we show that both excitatory glutamatergic and inhibitory GABA-ergic neurons are susceptible and permissive to historical and contemporary EV-D68 strains. EV-D68 infection of neurons leads to the reorganization of the Golgi-endomembranes forming replication organelles, first in the soma and later in the processes. Finally, we demonstrate that the spontaneous neuronal activity of EV-D68-infected neuronal network cultured on microelectrode arrays (MEA) is decreased, independent of the virus strain. Collectively, our findings provide novel insights into neurotropism and -pathology of different EV-D68 strains, and argue that it is unlikely that increased neurotropism is a recently acquired phenotype of a specific genetic lineage. IMPORTANCE Acute flaccid myelitis (AFM) is a serious neurological illness characterized by muscle weakness and paralysis in children. Since 2014, outbreaks of AFM have emerged worldwide, and they appear to be caused by nonpolio enteroviruses, particularly enterovirus-D68 (EV-D68), an unusual enterovirus that is known to mainly cause respiratory disease. It is unknown whether these outbreaks reflect a change of EV-D68 pathogenicity or are due to increased detection and awareness of this virus in recent years. To gain more insight herein, it is crucial to define how historical and circulating EV-D68 strains infect and replicate in neurons and how they affect their physiology. This study compares the entry and replication in neurons and the functional consequences on the neural network upon infection with an old "historical" strain and contemporary "circulating" strains of EV-D68.
Collapse
|
17
|
Goshi N, Kim H, Girardi G, Gardner A, Seker E. Electrophysiological Activity of Primary Cortical Neuron-Glia Mixed Cultures. Cells 2023; 12:cells12050821. [PMID: 36899957 PMCID: PMC10001406 DOI: 10.3390/cells12050821] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/17/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023] Open
Abstract
Neuroinflammation plays a central role in many neurological disorders, ranging from traumatic brain injuries to neurodegeneration. Electrophysiological activity is an essential measure of neuronal function, which is influenced by neuroinflammation. In order to study neuroinflammation and its electrophysiological fingerprints, there is a need for in vitro models that accurately capture the in vivo phenomena. In this study, we employed a new tri-culture of primary rat neurons, astrocytes, and microglia in combination with extracellular electrophysiological recording techniques using multiple electrode arrays (MEAs) to determine the effect of microglia on neural function and the response to neuroinflammatory stimuli. Specifically, we established the tri-culture and its corresponding neuron-astrocyte co-culture (lacking microglia) counterpart on custom MEAs and monitored their electrophysiological activity for 21 days to assess culture maturation and network formation. As a complementary assessment, we quantified synaptic puncta and averaged spike waveforms to determine the difference in excitatory to inhibitory neuron ratio (E/I ratio) of the neurons. The results demonstrate that the microglia in the tri-culture do not disrupt neural network formation and stability and may be a better representation of the in vivo rat cortex due to its more similar E/I ratio as compared to more traditional isolated neuron and neuron-astrocyte co-cultures. In addition, only the tri-culture displayed a significant decrease in both the number of active channels and spike frequency following pro-inflammatory lipopolysaccharide exposure, highlighting the critical role of microglia in capturing electrophysiological manifestations of a representative neuroinflammatory insult. We expect the demonstrated technology to assist in studying various brain disease mechanisms.
Collapse
Affiliation(s)
- Noah Goshi
- Department of Biomedical Engineering, University of California—Davis, Davis, CA 95616, USA
| | - Hyehyun Kim
- Department of Biomedical Engineering, University of California—Davis, Davis, CA 95616, USA
| | - Gregory Girardi
- Department of Biomedical Engineering, University of California—Davis, Davis, CA 95616, USA
| | - Alexander Gardner
- Department of Electrical and Computer Engineering, University of California—Davis, Davis, CA 95616, USA
| | - Erkin Seker
- Department of Electrical and Computer Engineering, University of California—Davis, Davis, CA 95616, USA
- Correspondence:
| |
Collapse
|
18
|
Kim DS, Pessah IN, Santana CM, Purnell BS, Li R, Buchanan GF, Rumbeiha WK. Investigations into hydrogen sulfide-induced suppression of neuronal activity in vivo and calcium dysregulation in vitro. Toxicol Sci 2023; 192:kfad022. [PMID: 36882182 PMCID: PMC10109532 DOI: 10.1093/toxsci/kfad022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Abstract
Acute exposure to high concentrations of hydrogen sulfide (H2S) leads to sudden death and, if survived, lingering neurological disorders. Clinical signs include seizures, loss of consciousness, and dyspnea. The proximate mechanisms underlying H2S-induced acute toxicity and death have not been clearly elucidated. We investigated electrocerebral, cardiac and respiratory activity during H2S exposure using electroencephalogram (EEG), electrocardiogram (EKG) and plethysmography. H2S suppressed electrocerebral activity and disrupted breathing. Cardiac activity was comparatively less affected. To test whether Ca2+ dysregulation contributes to H2S-induced EEG suppression, we developed an in vitro real-time rapid throughput assay measuring patterns of spontaneous synchronized Ca2+ oscillations in cultured primary cortical neuronal networks loaded with the indicator Fluo-4 using the fluorescent imaging plate reader (FLIPR-Tetra®). Sulfide >5 ppm dysregulated synchronous calcium oscillation (SCO) patterns in a dose-dependent manner. Inhibitors of NMDA and AMPA receptors magnified H2S-induced SCO suppression. Inhibitors of L-type voltage gated Ca2+ channels and transient receptor potential channels prevented H2S-induced SCO suppression. Inhibitors of T-type voltage gated Ca2+ channels, ryanodine receptors, and sodium channels had no measurable influence on H2S-induced SCO suppression. Exposures to > 5 ppm sulfide also suppressed neuronal electrical activity in primary cortical neurons measured by multi-electrode array (MEA), an effect alleviated by pretreatment with the nonselective transient receptor potential channel inhibitor, 2-APB. 2-APB also reduced primary cortical neuronal cell death from sulfide exposure. These results improve our understanding of the role of different Ca2+ channels in acute H2S-induced neurotoxicity and identify transient receptor potential channel modulators as novel structures with potential therapeutic benefits.
Collapse
Affiliation(s)
- Dong-Suk Kim
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California 95616, USA
| | - Isaac N Pessah
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California 95616, USA
| | - Cristina M Santana
- VDPAM, College of Veterinary Medicine, Iowa State University, Ames, Iowa 50011, USA
- MRIGlobal, Kansas City, Missouri 64110, USA
| | - Benton S Purnell
- Department of Neurology, Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52246, USA
- Department of Nerosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Rui Li
- Department of Neurology, Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52246, USA
| | - Gordon F Buchanan
- Department of Neurology, Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52246, USA
| | - Wilson K Rumbeiha
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California 95616, USA
| |
Collapse
|
19
|
Kuroda T, Matsuda N, Ishibashi Y, Suzuki I. Detection of astrocytic slow oscillatory activity and response to seizurogenic compounds using planar microelectrode array. Front Neurosci 2023; 16:1050150. [PMID: 36703996 PMCID: PMC9872017 DOI: 10.3389/fnins.2022.1050150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/14/2022] [Indexed: 01/12/2023] Open
Abstract
Since the development of the planar microelectrode array (MEA), it has become popular to evaluate compounds based on the electrical activity of rodent and human induced pluripotent stem cell (iPSC)-derived neurons. However, there are no reports recording spontaneous human astrocyte activity from astrocyte-only culture sample by MEA. It is becoming clear that astrocytes play an important role in various neurological diseases, and astrocytes are expected to be excellent candidates for targeted therapeutics for the treatment of neurological diseases. Therefore, measuring astrocyte activity is very important for drug development for astrocytes. Recently, astrocyte activity has been found to be reflected in the low-frequency band < 1 Hz, which is much lower than the frequency band for recording neural activity. Here, we separated the signals obtained from human primary astrocytes cultured on MEA into seven frequency bands and successfully recorded the extracellular electrical activity of human astrocytes. The slow waveforms of spontaneous astrocyte activity were observed most clearly in direct current potentials < 1 Hz. We established nine parameters to assess astrocyte activity and evaluated five seizurogenic drug responses in human primary astrocytes and human iPSC-derived astrocytes. Astrocytes demonstrated the most significant dose-dependent changes in pilocarpine. Furthermore, in a principal component analysis using those parameter sets, the drug responses to each seizurogenic compound were separated. In this paper, we report the spontaneous electrical activity measurement of astrocytes alone using MEA for the first time and propose that the MEA measurement focusing on the low-frequency band could be useful as one of the methods to assess drug response in vitro.
Collapse
|
20
|
Brofiga M, Poggio F, Callegari F, Tedesco M, Massobrio P. Modularity and neuronal heterogeneity: Two properties that influence in vitro neuropharmacological experiments. Front Cell Neurosci 2023; 17:1147381. [PMID: 37020847 PMCID: PMC10067731 DOI: 10.3389/fncel.2023.1147381] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/02/2023] [Indexed: 04/07/2023] Open
Abstract
Introduction The goal of this work is to prove the relevance of the experimental model (in vitro neuronal networks in this study) when drug-delivery testing is performed. Methods We used dissociated cortical and hippocampal neurons coupled to Micro-Electrode Arrays (MEAs) arranged in different configurations characterized by modularity (i.e., the presence of interconnected sub-networks) and heterogeneity (i.e., the co-existence of neurons coming from brain districts). We delivered increasing concentrations of bicuculline (BIC), a neuromodulator acting on the GABAergic system, and we extracted the IC50 values (i.e., the effective concentration yielding a reduction in the response by 50%) of the mean firing rate for each configuration. Results We found significant lower values of the IC50 computed for modular cortical-hippocampal ensembles than isolated cortical or hippocampal ones. Discussion Although tested with a specific neuromodulator, this work aims at proving the relevance of ad hoc experimental models to perform neuropharmacological experiments to avoid errors of overestimation/underestimation leading to biased information in the characterization of the effects of a drug on neuronal networks.
Collapse
Affiliation(s)
- Martina Brofiga
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genova, Genova, Italy
- ScreenNeuroPharm S.r.l., Sanremo, Italy
| | - Fabio Poggio
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genova, Genova, Italy
| | - Francesca Callegari
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genova, Genova, Italy
| | | | - Paolo Massobrio
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genova, Genova, Italy
- National Institute for Nuclear Physics (INFN), Genova, Italy
- MNESYS Extended Partnership Neuroscience and Neuropharmacology, Genova, Italy
- *Correspondence: Paolo Massobrio,
| |
Collapse
|
21
|
Choo SS, Keever JY, Brown J, Strickland JD, Shafer TJ. Assaying Spontaneous Network Activity and Cellular Viability Using Multi-Well Microelectrode Arrays. Methods Mol Biol 2023; 2644:133-154. [PMID: 37142920 DOI: 10.1007/978-1-0716-3052-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Microelectrode array (MEA) technology is a neurophysiological method that allows for the measurement of spontaneous or evoked neural activity to determine chemical effects thereon. Following assessment of compound effects on multiple endpoints that evaluate network function, a cell viability endpoint in the same well is determined using a multiplexed approach. Recently, it has become possible to measure electrical impedance of cells attached to the electrodes, where greater impedance indicates greater number of cells attached. This would allow rapid and repeated assessments of cell health as the neural network develops in longer exposure assays without impacting cell health. Typically, the lactate dehydrogenase (LDH) assay for cytotoxity and CellTiter-Blue® (CTB) assay for cell viability are only performed at the end of the chemical exposure period because these assays involve lysing of the cells. Procedures describing the multiplexed methods in acute and network formation screening are included in this chapter.
Collapse
Affiliation(s)
- Seline S Choo
- Oak Ridge Institute for Science and Engineering, Oak Ridge, TN, USA
- Rapid Assay Development Branch, Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Jackson Y Keever
- Rapid Assay Development Branch, Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC, USA
- Oak Ridge Associated Universities Student Contractor, Oak Ridge, TN, USA
| | - Jasmine Brown
- Rapid Assay Development Branch, Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| | - Jenna D Strickland
- Axion Biosystems, Atlanta, GA, USA
- LabCorp Drug Development, Madison, WI, USA
| | - Timothy J Shafer
- Rapid Assay Development Branch, Biomolecular and Computational Toxicology Division, Center for Computational Toxicology and Exposure, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC, USA.
| |
Collapse
|
22
|
Keil-Stietz K, Lein PJ. Gene×environment interactions in autism spectrum disorders. Curr Top Dev Biol 2022; 152:221-284. [PMID: 36707213 PMCID: PMC10496028 DOI: 10.1016/bs.ctdb.2022.11.001] [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] [Indexed: 12/24/2022]
Abstract
There is credible evidence that environmental factors influence individual risk and/or severity of autism spectrum disorders (hereafter referred to as autism). While it is likely that environmental chemicals contribute to the etiology of autism via multiple mechanisms, identifying specific environmental factors that confer risk for autism and understanding how they contribute to the etiology of autism has been challenging, in part because the influence of environmental chemicals likely varies depending on the genetic substrate of the exposed individual. Current research efforts are focused on elucidating the mechanisms by which environmental chemicals interact with autism genetic susceptibilities to adversely impact neurodevelopment. The goal is to not only generate insights regarding the pathophysiology of autism, but also inform the development of screening platforms to identify specific environmental factors and gene×environment (G×E) interactions that modify autism risk. Data from such studies are needed to support development of intervention strategies for mitigating the burden of this neurodevelopmental condition on individuals, their families and society. In this review, we discuss environmental chemicals identified as putative autism risk factors and proposed mechanisms by which G×E interactions influence autism risk and/or severity using polychlorinated biphenyls (PCBs) as an example.
Collapse
Affiliation(s)
- Kimberly Keil-Stietz
- Department of Comparative Biosciences, University of Wisconsin-Madison, School of Veterinary Medicine, Madison, WI, United States
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA, United States.
| |
Collapse
|
23
|
Zhang Z, Meng C, Hou K, Wang Z, Huang Y, Lü X. The cytological and electrophysiological effects of silver nanoparticles on neuron-like PC12 cells. PLoS One 2022; 17:e0277942. [PMID: 36512588 PMCID: PMC9746933 DOI: 10.1371/journal.pone.0277942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/27/2022] [Indexed: 12/15/2022] Open
Abstract
The aim of this study was to investigate the toxic effects and mechanism of silver nanoparticles (SNPs) on the cytological and electrophysiological properties of rat adrenal pheochromocytoma (PC12) cells. Different concentrations of SNPs (20 nm) were prepared, and the effects of different application durations on the cell viability and electrical excitability of PC12 quasi-neuronal networks were investigated. The effects of 200 μM SNPs on the neurite length, cell membrane potential (CMP) difference, intracellular Ca2+ content, mitochondrial membrane potential (MMP) difference, adenosine triphosphate (ATP) content, and reactive oxygen species (ROS) content of networks were then investigated. The results showed that 200 μM SNPs produced grade 1 cytotoxicity at 48 h of interaction, and the other concentrations of SNPs were noncytotoxic. Noncytotoxic 5 μM SNPs significantly increased electrical excitability, and noncytotoxic 100 μM SNPs led to an initial increase followed by a significant decrease in electrical excitability. Cytotoxic SNPs (200 μM) significantly decreased electrical excitability. SNPs (200 μM) led to decreases in neurite length, MMP difference and ATP content and increases in CMP difference and intracellular Ca2+ and ROS levels. The results revealed that not only cell viability but also electrophysiological properties should be considered when evaluating nanoparticle-induced neurotoxicity. The SNP-induced cytotoxicity mainly originated from its effects on ATP content, cytoskeletal structure and ROS content. The decrease in electrical excitability was mainly due to the decrease in ATP content. ATP content may thus be an important indicator of both cell viability and electrical excitability in PC12 quasi-neuronal networks.
Collapse
Affiliation(s)
- Zequn Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu Province, China
| | - Chen Meng
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu Province, China
| | - Kun Hou
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu Province, China
| | - Zhigong Wang
- Institute of RF- & OE-ICs, Southeast University, Nanjing, Jiangsu Province, China
- Coinnovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
- * E-mail: (ZW); (YH); (XL)
| | - Yan Huang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu Province, China
- * E-mail: (ZW); (YH); (XL)
| | - Xiaoying Lü
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu Province, China
- Coinnovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
- * E-mail: (ZW); (YH); (XL)
| |
Collapse
|
24
|
Sharfstein ST. Bio-hybrid electronic and photonic devices. Exp Biol Med (Maywood) 2022; 247:2128-2141. [PMID: 36533579 PMCID: PMC9837307 DOI: 10.1177/15353702221144087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Bio-hybrid devices, combining electronic and photonic components with cells, tissues, and organs, hold potential for advancing our understanding of biology, physiology, and pathologies and for treating a wide range of conditions and diseases. In this review, I describe the devices, materials, and technologies that enable bio-hybrid devices and provide examples of their utilization at multiple biological scales ranging from the subcellular to whole organs. Finally, I describe the outcomes of a National Science Foundation (NSF)-funded workshop envisioning potential applications of these technologies to improve health outcomes and quality of life.
Collapse
|
25
|
Wlodkowic D, Jansen M. High-throughput screening paradigms in ecotoxicity testing: Emerging prospects and ongoing challenges. CHEMOSPHERE 2022; 307:135929. [PMID: 35944679 DOI: 10.1016/j.chemosphere.2022.135929] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 06/09/2022] [Accepted: 07/31/2022] [Indexed: 06/15/2023]
Abstract
The rapidly increasing number of new production chemicals coupled with stringent implementation of global chemical management programs necessities a paradigm shift towards boarder uses of low-cost and high-throughput ecotoxicity testing strategies as well as deeper understanding of cellular and sub-cellular mechanisms of ecotoxicity that can be used in effective risk assessment. The latter will require automated acquisition of biological data, new capabilities for big data analysis as well as computational simulations capable of translating new data into in vivo relevance. However, very few efforts have been so far devoted into the development of automated bioanalytical systems in ecotoxicology. This is in stark contrast to standardized and high-throughput chemical screening and prioritization routines found in modern drug discovery pipelines. As a result, the high-throughput and high-content data acquisition in ecotoxicology is still in its infancy with limited examples focused on cell-free and cell-based assays. In this work we outline recent developments and emerging prospects of high-throughput bioanalytical approaches in ecotoxicology that reach beyond in vitro biotests. We discuss future importance of automated quantitative data acquisition for cell-free, cell-based as well as developments in phytotoxicity and in vivo biotests utilizing small aquatic model organisms. We also discuss recent innovations such as organs-on-a-chip technologies and existing challenges for emerging high-throughput ecotoxicity testing strategies. Lastly, we provide seminal examples of the small number of successful high-throughput implementations that have been employed in prioritization of chemicals and accelerated environmental risk assessment.
Collapse
Affiliation(s)
- Donald Wlodkowic
- The Neurotox Lab, School of Science, RMIT University, Melbourne, VIC, 3083, Australia.
| | - Marcus Jansen
- LemnaTec GmbH, Nerscheider Weg 170, 52076, Aachen, Germany
| |
Collapse
|
26
|
Lam D, Sebastian A, Bogguri C, Hum NR, Ladd A, Cadena J, Valdez CA, Fischer NO, Loots GG, Enright HA. Dose-dependent consequences of sub-chronic fentanyl exposure on neuron and glial co-cultures. FRONTIERS IN TOXICOLOGY 2022; 4:983415. [PMID: 36032789 PMCID: PMC9403314 DOI: 10.3389/ftox.2022.983415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Fentanyl is one of the most common opioid analgesics administered to patients undergoing surgery or for chronic pain management. While the side effects of chronic fentanyl abuse are recognized (e.g., addiction, tolerance, impairment of cognitive functions, and inhibit nociception, arousal, and respiration), it remains poorly understood what and how changes in brain activity from chronic fentanyl use influences the respective behavioral outcome. Here, we examined the functional and molecular changes to cortical neural network activity following sub-chronic exposure to two fentanyl concentrations, a low (0.01 μM) and high (10 μM) dose. Primary rat co-cultures, containing cortical neurons, astrocytes, and oligodendrocyte precursor cells, were seeded in wells on either a 6-well multi-electrode array (MEA, for electrophysiology) or a 96-well tissue culture plate (for serial endpoint bulk RNA sequencing analysis). Once networks matured (at 28 days in vitro), co-cultures were treated with 0.01 or 10 μM of fentanyl for 4 days and monitored daily. Only high dose exposure to fentanyl resulted in a decline in features of spiking and bursting activity as early as 30 min post-exposure and sustained for 4 days in cultures. Transcriptomic analysis of the complex cultures after 4 days of fentanyl exposure revealed that both the low and high dose induced gene expression changes involved in synaptic transmission, inflammation, and organization of the extracellular matrix. Collectively, the findings of this in vitro study suggest that while neuroadaptive changes to neural network activity at a systems level was detected only at the high dose of fentanyl, transcriptomic changes were also detected at the low dose conditions, suggesting that fentanyl rapidly elicits changes in plasticity.
Collapse
Affiliation(s)
- Doris Lam
- Biosciences and Biotechnology Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Aimy Sebastian
- Biosciences and Biotechnology Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Chandrakumar Bogguri
- Biosciences and Biotechnology Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Nicholas R. Hum
- Biosciences and Biotechnology Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Alexander Ladd
- Computational Engineering Division, Engineering Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Jose Cadena
- Computational Engineering Division, Engineering Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Carlos A. Valdez
- Nuclear and Chemical Sciences Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Nicholas O. Fischer
- Biosciences and Biotechnology Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Gabriela G. Loots
- Biosciences and Biotechnology Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Heather A. Enright
- Biosciences and Biotechnology Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
- *Correspondence: Heather A. Enright,
| |
Collapse
|
27
|
Wlodkowic D, Bownik A, Leitner C, Stengel D, Braunbeck T. Beyond the behavioural phenotype: Uncovering mechanistic foundations in aquatic eco-neurotoxicology. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 829:154584. [PMID: 35306067 DOI: 10.1016/j.scitotenv.2022.154584] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 06/14/2023]
Abstract
During the last decade, there has been an increase in awareness of how anthropogenic pollution can alter behavioural traits of diverse aquatic organisms. Apart from understanding profound ecological implications, alterations in neuro-behavioural indices have emerged as sensitive and physiologically integrative endpoints in chemical risk assessment. Accordingly, behavioural ecotoxicology and broader eco-neurotoxicology are becoming increasingly popular fields of research that span a plethora of fundamental laboratory experimentations as well as applied field-based studies. Despite mounting interest in aquatic behavioural ecotoxicology studies, there is, however, a considerable paucity in deciphering the mechanistic foundations underlying behavioural alterations upon exposure to pollutants. The behavioural phenotype is indeed the highest-level integrative neurobiological phenomenon, but at its core lie myriads of intertwined biochemical, cellular, and physiological processes. Therefore, the mechanisms that underlie changes in behavioural phenotypes can stem among others from dysregulation of neurotransmitter pathways, electrical signalling, and cell death of discrete cell populations in the central and peripheral nervous systems. They can, however, also be a result of toxicity to sensory organs and even metabolic dysfunctions. In this critical review, we outline why behavioural phenotyping should be the starting point that leads to actual discovery of fundamental mechanisms underlying actions of neurotoxic and neuromodulating contaminants. We highlight potential applications of the currently existing and emerging neurobiology and neurophysiology analytical strategies that should be embraced and more broadly adopted in behavioural ecotoxicology. Such strategies can provide new mechanistic discoveries instead of only observing the end sum phenotypic effects.
Collapse
Affiliation(s)
- Donald Wlodkowic
- The Neurotox Laboratory, School of Science, RMIT University, Melbourne, Australia.
| | - Adam Bownik
- Department of Hydrobiology and Protection of Ecosystems, Faculty of Environmental Biology, University of Life Sciences, Lublin, Poland
| | - Carola Leitner
- Aquatic Ecology and Toxicology, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, D-69120 Heidelberg, Germany
| | - Daniel Stengel
- Aquatic Ecology and Toxicology, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, D-69120 Heidelberg, Germany
| | - Thomas Braunbeck
- Aquatic Ecology and Toxicology, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, D-69120 Heidelberg, Germany
| |
Collapse
|
28
|
Dimensionality reduction for visualizing high-dimensional biological data. Biosystems 2022; 220:104749. [DOI: 10.1016/j.biosystems.2022.104749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 07/02/2022] [Accepted: 07/24/2022] [Indexed: 01/04/2023]
|
29
|
Schröter M, Wang C, Terrigno M, Hornauer P, Huang Z, Jagasia R, Hierlemann A. Functional imaging of brain organoids using high-density microelectrode arrays. MRS BULLETIN 2022; 47:530-544. [PMID: 36120104 PMCID: PMC9474390 DOI: 10.1557/s43577-022-00282-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/02/2022] [Indexed: 05/31/2023]
Abstract
ABSTRACT Studies have provided evidence that human cerebral organoids (hCOs) recapitulate fundamental milestones of early brain development, but many important questions regarding their functionality and electrophysiological properties persist. High-density microelectrode arrays (HD-MEAs) represent an attractive analysis platform to perform functional studies of neuronal networks at the cellular and network scale. Here, we use HD-MEAs to derive large-scale electrophysiological recordings from sliced hCOs. We record the activity of hCO slices over several weeks and probe observed neuronal dynamics pharmacologically. Moreover, we present results on how the obtained recordings can be spike-sorted and subsequently studied across scales. For example, we show how to track single neurons across several days on the HD-MEA and how to infer axonal action potential velocities. We also infer putative functional connectivity from hCO recordings. The introduced methodology will contribute to a better understanding of developing neuronal networks in brain organoids and provide new means for their functional characterization. IMPACT STATEMENT Human cerebral organoids (hCOs) represent an attractive in vitro model system to study key physiological mechanisms underlying early neuronal network formation in tissue with healthy or disease-related genetic backgrounds. Despite remarkable advances in the generation of brain organoids, knowledge on the functionality of their neuronal circuits is still scarce. Here, we used complementary metal-oxide-semiconductor (CMOS)-based high-density microelectrode arrays (HD-MEAs) to perform large-scale recordings from sliced hCOs over several weeks and quantified their activity across scales. Using single-cell and network metrics, we were able to probe aspects of hCO neurophysiology that are more difficult to obtain with other techniques, such as patch clamping (lower yield) and calcium imaging (lower temporal resolution). These metrics included, for example, extracellular action potential (AP) waveform features and axonal AP velocity at the cellular level, as well as functional connectivity at the network level. Analysis was enabled by the large sensing area and the high spatiotemporal resolution provided by HD-MEAs, which allowed recordings from hundreds of neurons and spike sorting of their activity. Our results demonstrate that HD-MEAs provide a multi-purpose platform for the functional characterization of hCOs, which will be key in improving our understanding of this model system and assessing its relevance for translational research. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1557/s43577-022-00282-w.
Collapse
Affiliation(s)
- Manuel Schröter
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Congwei Wang
- NRD, F. Hoffmann-La Roche Ltd., Roche Innovation Center Basel, Basel, Switzerland
| | - Marco Terrigno
- NRD, F. Hoffmann-La Roche Ltd., Roche Innovation Center Basel, Basel, Switzerland
| | - Philipp Hornauer
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Ziqiang Huang
- EMBL Imaging Centre, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Ravi Jagasia
- NRD, F. Hoffmann-La Roche Ltd., Roche Innovation Center Basel, Basel, Switzerland
| | - Andreas Hierlemann
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| |
Collapse
|
30
|
Miny L, Maisonneuve BGC, Quadrio I, Honegger T. Modeling Neurodegenerative Diseases Using In Vitro Compartmentalized Microfluidic Devices. Front Bioeng Biotechnol 2022; 10:919646. [PMID: 35813998 PMCID: PMC9263267 DOI: 10.3389/fbioe.2022.919646] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/31/2022] [Indexed: 01/27/2023] Open
Abstract
The human brain is a complex organ composed of many different types of cells interconnected to create an organized system able to efficiently process information. Dysregulation of this delicately balanced system can lead to the development of neurological disorders, such as neurodegenerative diseases (NDD). To investigate the functionality of human brain physiology and pathophysiology, the scientific community has been generated various research models, from genetically modified animals to two- and three-dimensional cell culture for several decades. These models have, however, certain limitations that impede the precise study of pathophysiological features of neurodegeneration, thus hindering therapeutical research and drug development. Compartmentalized microfluidic devices provide in vitro minimalistic environments to accurately reproduce neural circuits allowing the characterization of the human central nervous system. Brain-on-chip (BoC) is allowing our capability to improve neurodegeneration models on the molecular and cellular mechanism aspects behind the progression of these troubles. This review aims to summarize and discuss the latest advancements of microfluidic models for the investigations of common neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Louise Miny
- NETRI, Lyon, France
- BIORAN Team, Lyon Neuroscience Research Center, CNRS UMR 5292, INSERM U1028, Lyon 1 University, Bron, France
| | | | - Isabelle Quadrio
- BIORAN Team, Lyon Neuroscience Research Center, CNRS UMR 5292, INSERM U1028, Lyon 1 University, Bron, France
- Laboratory of Neurobiology and Neurogenetics, Department of Biochemistry and Molecular Biology, Lyon University Hospital, Bron, France
| | | |
Collapse
|
31
|
Mai P, Hampl J, Baca M, Brauer D, Singh S, Weise F, Borowiec J, Schmidt A, Küstner JM, Klett M, Gebinoga M, Schroeder IS, Markert UR, Glahn F, Schumann B, Eckstein D, Schober A. MatriGrid® Based Biological Morphologies: Tools for 3D Cell Culturing. Bioengineering (Basel) 2022; 9:bioengineering9050220. [PMID: 35621498 PMCID: PMC9138054 DOI: 10.3390/bioengineering9050220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 02/06/2023] Open
Abstract
Recent trends in 3D cell culturing has placed organotypic tissue models at another level. Now, not only is the microenvironment at the cynosure of this research, but rather, microscopic geometrical parameters are also decisive for mimicking a tissue model. Over the years, technologies such as micromachining, 3D printing, and hydrogels are making the foundation of this field. However, mimicking the topography of a particular tissue-relevant substrate can be achieved relatively simply with so-called template or morphology transfer techniques. Over the last 15 years, in one such research venture, we have been investigating a micro thermoforming technique as a facile tool for generating bioinspired topographies. We call them MatriGrid®s. In this research account, we summarize our learning outcome from this technique in terms of the influence of 3D micro morphologies on different cell cultures that we have tested in our laboratory. An integral part of this research is the evolution of unavoidable aspects such as possible label-free sensing and fluidic automatization. The development in the research field is also documented in this account.
Collapse
Affiliation(s)
- Patrick Mai
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Jörg Hampl
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
- Correspondence: (J.H.); (A.S.); Tel.: +49-3677-6933387 (A.S.)
| | - Martin Baca
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Dana Brauer
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Sukhdeep Singh
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Frank Weise
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Justyna Borowiec
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - André Schmidt
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany; (A.S.); (U.R.M.)
| | - Johanna Merle Küstner
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Maren Klett
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Michael Gebinoga
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Insa S. Schroeder
- Biophysics Division, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany;
| | - Udo R. Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany; (A.S.); (U.R.M.)
| | - Felix Glahn
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Berit Schumann
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Diana Eckstein
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Andreas Schober
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
- Correspondence: (J.H.); (A.S.); Tel.: +49-3677-6933387 (A.S.)
| |
Collapse
|
32
|
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
|
33
|
Hou K, Meng C, Huang Y, Zhang Z, Wang Z, Lü X. A Research on the Role and Mechanism of N-Methyl-D-Aspartate Receptors in the Effects of Silver Nanoparticles on the Electrical Excitability of Hippocampal Neuronal Networks. J Biomed Nanotechnol 2022. [DOI: 10.1166/jbn.2022.3357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The purpose of this paper is to explore the role and mechanism of N-Methyl-D-Aspartate (NMDA) receptors in the effects of silver nanoparticles (SNPs) on the electrical excitability of hippocampal neuronal networks. First, the cytotoxicity of different concentrations of SNPs was evaluated
and screened by MTT experiment, then the Voltage Threshold Measurement Method (VTMM) was employed to study the effects of SNPs on the electrical excitability of hippocampal neuronal networks under non-cytotoxic (5 μM) and cytotoxic (100 μM) concentrations after different
action times. The role of NMDA receptors in the effects of SNPs on the electrical excitability of hippocampal neuronal networks was investigated through the NMDA receptor antagonist MK-801. Then, the effects of SNPs on the number of NMDA receptors and the Ca2+ content in hippocampal
neurons were further investigated, and the relationship between these changes and neuronal networks electrical excitability was discussed. The results of voltage threshold (VTh) test showed that non-cytotoxic 5 μM SNPs has an excitatory effect on hippocampal neuronal
networks, while the effect of cytotoxic 100 μM SNPs gradually changed from excitatory to inhibitory with the extension of action time. It was found that SNPs could increase the electrical excitability of neuronal networks by activating NMDA receptors through the experiments with
MK-801 antagonists. Moreover, the fluorescent staining experiments showed that the activation of NMDA receptors by SNPs can lead to an increase in the intracellular Ca2+ content, and then trigger a negative feedback regulation mechanism of neurons between the number of NMDA receptors
and intracellular Ca2+ content. The high Ca2+ content in neurons can also decrease neurons’ cell viability, which in turn leads to changes in the electrical excitability of the neuronal networks.
Collapse
Affiliation(s)
- Kun Hou
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, PR China
| | - Chen Meng
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, PR China
| | - Yan Huang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, PR China
| | - Zequn Zhang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, PR China
| | - Zhigong Wang
- Institute of RF- & OE-ICs, Southeast University, Nanjing, 210096, PR China
| | - Xiaoying Lü
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, PR China
| |
Collapse
|
34
|
Bryson A, Mendis D, Morrisroe E, Reid CA, Halgamuge S, Petrou S. Classification of antiseizure drugs in cultured neuronal networks using multielectrode arrays and unsupervised learning. Epilepsia 2022; 63:1693-1703. [PMID: 35460272 DOI: 10.1111/epi.17268] [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: 02/09/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Antiseizure drugs (ASDs) modulate synaptic and ion channel function to prevent abnormal hypersynchronous or excitatory activity arising in neuronal networks, but the relationship between ASDs with respect to their impact on network activity is poorly defined. In this study, we first investigated whether different ASD classes exert differential impact upon network activity, and we then sought to classify ASDs according to their impact on network activity. METHODS We used multielectrode arrays (MEAs) to record the network activity of cultured cortical neurons after applying ASDs from two classes: sodium channel blockers (SCBs) and γ-aminobutyric acid type A receptor-positive allosteric modulators (GABA PAMs). A two-dimensional representation of changes in network features was then derived, and the ability of this low-dimensional representation to classify ASDs with different molecular targets was assessed. RESULTS A two-dimensional representation of network features revealed a separation between the SCB and GABA PAM drug classes, and could classify several test compounds known to act through these molecular targets. Interestingly, several ASDs with novel targets, such as cannabidiol and retigabine, had closer similarity to the SCB class with respect to their impact upon network activity. SIGNIFICANCE These results demonstrate that the molecular target of two common classes of ASDs is reflected through characteristic changes in network activity of cultured neurons. Furthermore, a low-dimensional representation of network features can be used to infer an ASDs molecular target. This approach may allow for drug screening to be performed based on features extracted from MEA recordings.
Collapse
Affiliation(s)
- Alexander Bryson
- Ion Channels and Diseases Group, Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Department of Neurology, Austin Health, Heidelberg, Victoria, Australia
| | | | - Emma Morrisroe
- Ion Channels and Diseases Group, Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Christopher A Reid
- Ion Channels and Diseases Group, Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Saman Halgamuge
- Department of Mechanical Engineering, School of Electrical, Mechanical, and Infrastructure Engineering, University of Melbourne, Parkville, Victoria, Australia
| | - Steven Petrou
- Ion Channels and Diseases Group, Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| |
Collapse
|
35
|
Tomasello DL, Wlodkowic D. Noninvasive Electrophysiology: Emerging Prospects in Aquatic Neurotoxicity Testing. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:4788-4794. [PMID: 35196004 DOI: 10.1021/acs.est.1c08471] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The significance of neurotoxicological risks associated with anthropogenic pollution is gaining increasing recognition worldwide. In this regard, perturbations in behavioral traits upon exposure to environmentally relevant concentrations of neurotoxic and neuro-modulating contaminants have been linked to diminished ecological fitness of many aquatic species. Despite an increasing interest in behavioral testing in aquatic ecotoxicology there is, however, a notable gap in understanding of the neurophysiological foundations responsible for the altered behavioral phenotypes. One of the canonical approaches to explain the mechanisms of neuro-behavioral changes is functional analysis of neuronal transmission. In aquatic animals it requires, however, invasive, complex, and time-consuming electrophysiology techniques. In this perspective, we highlight emerging prospects of noninvasive, in situ electrophysiology based on multielectrode arrays (MEAs). This technology has only recently been pioneered for the detection and analysis of transient electrical signals in the central nervous system of small model organisms such as zebrafish. The analysis resembles electroencephalography (EEG) applications and provides an appealing strategy for mechanistic explorative studies as well as routine neurotoxicity risk assessment. We outline the prospective future applications and existing challenges of this emerging analytical strategy that is poised to bring new vistas for aquatic ecotoxicology such as greater mechanistic understanding of eco-neurotoxicity and thus more robust risk assessment protocols.
Collapse
Affiliation(s)
- Danielle L Tomasello
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, United States
| | - Donald Wlodkowic
- The Neurotox Lab, School of Science, RMIT University, Melbourne, Victoria 3083, Australia
| |
Collapse
|
36
|
Dobreniecki S, Mendez E, Lowit A, Freudenrich TM, Wallace K, Carpenter A, Wetmore BA, Kreutz A, Korol-Bexell E, Friedman KP, Shafer TJ. Integration of toxicodynamic and toxicokinetic new approach methods into a weight-of-evidence analysis for pesticide developmental neurotoxicity assessment: A case-study with DL- and L-glufosinate. Regul Toxicol Pharmacol 2022; 131:105167. [PMID: 35413399 DOI: 10.1016/j.yrtph.2022.105167] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/14/2022] [Accepted: 04/06/2022] [Indexed: 01/13/2023]
Abstract
DL-glufosinate ammonium (DL-GLF) is a registered herbicide for which a guideline Developmental Neurotoxicity (DNT) study has been conducted. Offspring effects included altered brain morphometrics, decreased body weight, and increased motor activity. Guideline DNT studies are not available for its enriched isomers L-GLF acid and L-GLF ammonium; conducting one would be time consuming, resource-intensive, and possibly redundant given the existing DL-GLF DNT. To support deciding whether to request a guideline DNT study for the L-GLF isomers, DL-GLF and the L-GLF isomers were screened using in vitro assays for network formation and neurite outgrowth. DL-GLF and L-GLF isomers were without effects in both assays. DL-GLF and L-GLF (1-100 μM) isomers increased mean firing rate of mature networks to 120-140% of baseline. In vitro toxicokinetic assessments were used to derive administered equivalent doses (AEDs) for the in vitro testing concentrations. The AED for L-GLF was ∼3X higher than the NOAEL from the DL-GLF DNT indicating that the available guideline study would be protective of potential DNT due to L-GLF exposure. Based in part on the results of these in vitro studies, EPA is not requiring L-GLF isomer guideline DNT studies, thereby providing a case study for a useful application of DNT screening assays.
Collapse
Affiliation(s)
| | | | - Anna Lowit
- Office of Pesticide Programs USEPA, Washington, DC, USA
| | - Theresa M Freudenrich
- Center for Computational Toxicology and Exposure, Office of Research and Development. US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Kathleen Wallace
- Center for Computational Toxicology and Exposure, Office of Research and Development. US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Amy Carpenter
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | - Barbara A Wetmore
- Center for Computational Toxicology and Exposure, Office of Research and Development. US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Anna Kreutz
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
| | | | - Katie Paul Friedman
- Center for Computational Toxicology and Exposure, Office of Research and Development. US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Timothy J Shafer
- Center for Computational Toxicology and Exposure, Office of Research and Development. US Environmental Protection Agency, Research Triangle Park, NC, USA.
| |
Collapse
|
37
|
Zhang H, Rong G, Bian S, Sawan M. Lab-on-Chip Microsystems for Ex Vivo Network of Neurons Studies: A Review. Front Bioeng Biotechnol 2022; 10:841389. [PMID: 35252149 PMCID: PMC8888888 DOI: 10.3389/fbioe.2022.841389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Increasing population is suffering from neurological disorders nowadays, with no effective therapy available to treat them. Explicit knowledge of network of neurons (NoN) in the human brain is key to understanding the pathology of neurological diseases. Research in NoN developed slower than expected due to the complexity of the human brain and the ethical considerations for in vivo studies. However, advances in nanomaterials and micro-/nano-microfabrication have opened up the chances for a deeper understanding of NoN ex vivo, one step closer to in vivo studies. This review therefore summarizes the latest advances in lab-on-chip microsystems for ex vivo NoN studies by focusing on the advanced materials, techniques, and models for ex vivo NoN studies. The essential methods for constructing lab-on-chip models are microfluidics and microelectrode arrays. Through combination with functional biomaterials and biocompatible materials, the microfluidics and microelectrode arrays enable the development of various models for ex vivo NoN studies. This review also includes the state-of-the-art brain slide and organoid-on-chip models. The end of this review discusses the previous issues and future perspectives for NoN studies.
Collapse
Affiliation(s)
| | | | - Sumin Bian
- CenBRAIN Lab, School of Engineering, Westlake University, Hangzhou, China
| | - Mohamad Sawan
- CenBRAIN Lab, School of Engineering, Westlake University, Hangzhou, China
| |
Collapse
|
38
|
McCready FP, Gordillo-Sampedro S, Pradeepan K, Martinez-Trujillo J, Ellis J. Multielectrode Arrays for Functional Phenotyping of Neurons from Induced Pluripotent Stem Cell Models of Neurodevelopmental Disorders. BIOLOGY 2022; 11:316. [PMID: 35205182 PMCID: PMC8868577 DOI: 10.3390/biology11020316] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 12/11/2022]
Abstract
In vitro multielectrode array (MEA) systems are increasingly used as higher-throughput platforms for functional phenotyping studies of neurons in induced pluripotent stem cell (iPSC) disease models. While MEA systems generate large amounts of spatiotemporal activity data from networks of iPSC-derived neurons, the downstream analysis and interpretation of such high-dimensional data often pose a significant challenge to researchers. In this review, we examine how MEA technology is currently deployed in iPSC modeling studies of neurodevelopmental disorders. We first highlight the strengths of in vitro MEA technology by reviewing the history of its development and the original scientific questions MEAs were intended to answer. Methods of generating patient iPSC-derived neurons and astrocytes for MEA co-cultures are summarized. We then discuss challenges associated with MEA data analysis in a disease modeling context, and present novel computational methods used to better interpret network phenotyping data. We end by suggesting best practices for presenting MEA data in research publications, and propose that the creation of a public MEA data repository to enable collaborative data sharing would be of great benefit to the iPSC disease modeling community.
Collapse
Affiliation(s)
- Fraser P. McCready
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; (F.P.M.); (S.G.-S.)
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Sara Gordillo-Sampedro
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; (F.P.M.); (S.G.-S.)
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Kartik Pradeepan
- Department of Physiology and Pharmacology, Department of Psychiatry, Robarts Research and Brain and Mind Institutes, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5B7, Canada; (K.P.); (J.M.-T.)
| | - Julio Martinez-Trujillo
- Department of Physiology and Pharmacology, Department of Psychiatry, Robarts Research and Brain and Mind Institutes, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5B7, Canada; (K.P.); (J.M.-T.)
| | - James Ellis
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; (F.P.M.); (S.G.-S.)
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| |
Collapse
|
39
|
Matsuda N, Odawara A, Kinoshita K, Okamura A, Shirakawa T, Suzuki I. Raster plots machine learning to predict the seizure liability of drugs and to identify drugs. Sci Rep 2022; 12:2281. [PMID: 35145132 PMCID: PMC8831568 DOI: 10.1038/s41598-022-05697-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 01/03/2022] [Indexed: 11/17/2022] Open
Abstract
In vitro microelectrode array (MEA) assessment using human induced pluripotent stem cell (iPSC)-derived neurons holds promise as a method of seizure and toxicity evaluation. However, there are still issues surrounding the analysis methods used to predict seizure and toxicity liability as well as drug mechanisms of action. In the present study, we developed an artificial intelligence (AI) capable of predicting the seizure liability of drugs and identifying drugs using deep learning based on raster plots of neural network activity. The seizure liability prediction AI had a prediction accuracy of 98.4% for the drugs used to train it, classifying them correctly based on their responses as either seizure-causing compounds or seizure-free compounds. The AI also made concentration-dependent judgments of the seizure liability of drugs that it was not trained on. In addition, the drug identification AI implemented using the leave-one-sample-out scheme could distinguish among 13 seizure-causing compounds as well as seizure-free compound responses, with a mean accuracy of 99.9 ± 0.1% for all drugs. These AI prediction models are able to identify seizure liability concentration-dependence, rank the level of seizure liability based on the seizure liability probability, and identify the mechanism of the action of compounds. This holds promise for the future of in vitro MEA assessment as a powerful, high-accuracy new seizure liability prediction method.
Collapse
Affiliation(s)
- N Matsuda
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - A Odawara
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - K Kinoshita
- Drug Safety Research Labs, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan
| | - A Okamura
- Drug Safety Research Labs, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan
| | - T Shirakawa
- Drug Safety Research Labs, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan
| | - I Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan.
| |
Collapse
|
40
|
Smith AS, Kim JH, Chun C, Gharai A, Moon HW, Kim EY, Nam SH, Ha N, Song JY, Chung KW, Doo HM, Hesson J, Mathieu J, Bothwell M, Choi BO, Kim DH. HDAC6 Inhibition Corrects Electrophysiological and Axonal Transport Deficits in a Human Stem Cell-Based Model of Charcot-Marie-Tooth Disease (Type 2D). Adv Biol (Weinh) 2022; 6:e2101308. [PMID: 34958183 PMCID: PMC8849597 DOI: 10.1002/adbi.202101308] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Indexed: 02/03/2023]
Abstract
Charcot-Marie-Tooth disease type 2D (CMT2D), is a hereditary peripheral neuropathy caused by mutations in the gene encoding glycyl-tRNA synthetase (GARS1). Here, human induced pluripotent stem cell (hiPSC)-based models of CMT2D bearing mutations in GARS1 and their use for the identification of predictive biomarkers amenable to therapeutic efficacy screening is described. Cultures containing spinal cord motor neurons generated from this line exhibit network activity marked by significant deficiencies in spontaneous action potential firing and burst fire behavior. This result matches clinical data collected from a patient bearing a GARS1P724H mutation and is coupled with significant decreases in acetylated α-tubulin levels and mitochondrial movement within axons. Treatment with histone deacetylase 6 inhibitors, tubastatin A and CKD504, improves mitochondrial movement and α-tubulin acetylation in these cells. Furthermore, CKD504 treatment enhances population-level electrophysiological activity, highlighting its potential as an effective treatment for CMT2D.
Collapse
Affiliation(s)
| | | | - Changho Chun
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Ava Gharai
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Hyo Won Moon
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eun Young Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Soo Hyun Nam
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Nina Ha
- CKD Research Institute, Yongin, 16995, Republic of Korea
| | - Ju Yong Song
- CKD Research Institute, Yongin, 16995, Republic of Korea
| | - Ki Wha Chung
- Department of Biological Sciences, Kongju National University, Gongju 32588, Republic of Korea
| | - Hyun Myung Doo
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jennifer Hesson
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA.,Department of Comparative Medicine, University of Washington, Seattle, WA 98195, USA
| | - Julie Mathieu
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA.,Department of Comparative Medicine, University of Washington, Seattle, WA 98195, USA
| | - Mark Bothwell
- Department of Physiology and Biophysics, University of Washington, Seattle WA 98195, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Byung-Ok Choi
- Authors share corresponding authorship: To whom correspondence should be addressed: Dr. Deok-Ho Kim, Department of Biomedical Engineering, The Johns Hopkins University, Ross Research Building, 724B, 720 Rutland Avenue, Baltimore, MD 21205, , Dr. Byung-Ok Choi, Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea,
| | - Deok-Ho Kim
- Authors share corresponding authorship: To whom correspondence should be addressed: Dr. Deok-Ho Kim, Department of Biomedical Engineering, The Johns Hopkins University, Ross Research Building, 724B, 720 Rutland Avenue, Baltimore, MD 21205, , Dr. Byung-Ok Choi, Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea,
| |
Collapse
|
41
|
Woo H, Kim S, Nam H, Choi W, Shin K, Kim K, Yoon S, Kim GH, Kim J, Lim G. Au Hierarchical Nanostructure-Based Surface Modification of Microelectrodes for Improved Neural Signal Recording. Anal Chem 2021; 93:11765-11774. [PMID: 34387479 DOI: 10.1021/acs.analchem.1c02168] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Microelectrodes are widely used for neural signal analysis because they can record high-resolution signals. In general, the smaller the size of the microelectrode for obtaining a high-resolution signal, the higher the impedance and noise value of the electrodes. Therefore, to improve the signal-to-noise ratio (SNR) of neural signals, it is important to develop microelectrodes with low impedance and noise. In this research, an Au hierarchical nanostructure (AHN) was deposited to improve the electrochemical surface area (ECSA) of a microelectrode. Au nanostructures on different scales were deposited on the electrode surface in a hierarchical structure using an electrochemical deposition method. The AHN-modified microelectrode exhibited an average of 80% improvement in impedance compared to a bare microelectrode. Through electrochemical impedance spectroscopy analysis and impedance equivalent circuit modeling, the increase in the ECSA due to the AHN was confirmed. After evaluating the cell cytotoxicity of the AHN-modified microelectrode through an in vitro test, neural signals from rats were obtained in in vivo experiments. The AHN-modified microelectrode exhibited an approximate 9.79 dB improvement in SNR compared to the bare microelectrode. This surface modification technology is a post-treatment strategy used for existing fabricated electrodes, so it can be applied to microelectrode arrays and nerve electrodes made from various structures and materials.
Collapse
Affiliation(s)
| | | | | | - Wonsuk Choi
- Center for Bionics, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Kumjae Shin
- Construction Equipment Technology Center, Korea Institute of Industrial Technology (KITECH), 288-1, Daehak-ri, Hayang-eup, Gyeongsan-si, Gyeongsangbuk-do 38408, Republic of Korea
| | | | | | - Geon Hwee Kim
- School of Mechanical Engineering, Chungbuk National University (CBNU), 1, Chungdae-ro, Seowon-gu, Cheongju-si, Chungcheongbuk-do 28644, Republic of Korea
| | - Jinseok Kim
- Center for Bionics, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | | |
Collapse
|
42
|
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
|
43
|
Tao Y, Hu B, Ma Z, Li H, Du E, Wang G, Xing B, Ma J, Song Z. Intravitreous delivery of melatonin affects the retinal neuron survival and visual signal transmission: in vivo and ex vivo study. Drug Deliv 2021; 27:1386-1396. [PMID: 33016801 PMCID: PMC7580852 DOI: 10.1080/10717544.2020.1818882] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Intravitreal delivery can maximize the intensity of therapeutic agents and extend their residence time within ocular tissue. Melatonin is a lipophilic molecule that crosses freely biological barriers and cell membranes. This study intends to investigate the effects of intravitreally delivered melatonin on mouse retina. The visual function of administered mice is assessed by electrophysiological and behavior examinations three weeks after intravitreal delivery. Moreover, multi-electrode array (MEA) was used to assess the electrical activities of retinal ganglion cells (RGCs). We found that intravitreal delivery of high dosage melatonin (400-500 µg/kg) destroyed the retinal architecture and impaired the visual function of mice. Conversely, the melatonin administration at low dose (100-300 µg/kg) did not have any significant effects on the photoreceptor survival or visual function. As shown in the MEA recording, the photoreceptors activity of the central region was more severely disturbed by the high dose melatonin. A pronounced augment of the spontaneous firing frequency was recorded in these mice received high dosage melatonin, indicating that intravitreal delivery of high dosage melatonin would affect the electrical activity of RGCs. Immunostaining assay showed that the vitality of cone photoreceptor was impaired by high dose melatonin. These findings suggest that intravitreal melatonin is not always beneficial for ocular tissues, especially when it is administered at high dosage. These data add new perspectives to current knowledge about melatonin delivery at the ocular level. Further therapeutic strategies should take into consideration of these risks that caused by delivery approach.
Collapse
Affiliation(s)
- Ye Tao
- Department of Ophthalmology, People's hospital of Zhengzhou University, Zhengzhou, PR China.,Department of physiology and neuroscience, Basic college of medicine, Zhengzhou University Zhengzhou, PR China
| | - Bang Hu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Zhao Ma
- Department of Neurosurgery, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wu Han, PR China
| | - Haijun Li
- Department of Ophthalmology, People's hospital of Zhengzhou University, Zhengzhou, PR China.,Department of physiology and neuroscience, Basic college of medicine, Zhengzhou University Zhengzhou, PR China
| | - Enming Du
- Department of Ophthalmology, People's hospital of Zhengzhou University, Zhengzhou, PR China.,Department of physiology and neuroscience, Basic college of medicine, Zhengzhou University Zhengzhou, PR China
| | - Gang Wang
- Department of Ophthalmology, People's hospital of Zhengzhou University, Zhengzhou, PR China.,Department of physiology and neuroscience, Basic college of medicine, Zhengzhou University Zhengzhou, PR China
| | - Biao Xing
- Department of Neurosurgery, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wu Han, PR China
| | - Jie Ma
- Department of Neurosurgery, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wu Han, PR China
| | - Zongming Song
- Department of Ophthalmology, People's hospital of Zhengzhou University, Zhengzhou, PR China.,Department of physiology and neuroscience, Basic college of medicine, Zhengzhou University Zhengzhou, PR China
| |
Collapse
|
44
|
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
|
45
|
Bownik A, Wlodkowic D. Applications of advanced neuro-behavioral analysis strategies in aquatic ecotoxicology. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 772:145577. [PMID: 33770877 DOI: 10.1016/j.scitotenv.2021.145577] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 06/12/2023]
Abstract
Despite mounting evidence of pleiotropic ecological risks, the understanding of the eco-neurotoxic impact of most industrially relevant chemicals is still very limited. In particularly the acute and chronic exposures to industrial pollutants on nervous systems and thus potential alterations in ecological fitness remain profoundly understudied. Since the behavioral phenotype is the highest-level and functional manifestation of integrated neurological functions, the alterations in neuro-behavioral traits have been postulated as very sensitive and physiologically integrative endpoints to assess eco-neurotoxicological risks associated with industrial pollutants. Due to a considerable backlog of risk assessments of existing and new production chemicals there is a need for a paradigm shift from high cost, low throughput ecotoxicity test models to next generation systems amenable to higher throughput. In this review we concentrate on emerging aspects of laboratory-based neuro-behavioral phenotyping approaches that can be amenable for rapid prioritizing pipelines. We outline the importance of development and applications of innovative neuro-behavioral assays utilizing small aquatic biological indicators and demonstrate emerging concepts of high-throughput chemo-behavioral phenotyping. We also discuss new analytical approaches to effectively and rapidly evaluate the impact of pollutants on higher behavioral functions such as sensory-motor assays, decision-making and cognitive behaviors using innovative model organisms. Finally, we provide a snapshot of most recent analytical approaches that can be applied to elucidate mechanistic rationale that underlie the observed neuro-behavioral alterations upon exposure to pollutants. This review is intended to outline the emerging opportunities for innovative multidisciplinary research and highlight the existing challenges as well barriers to future development.
Collapse
Affiliation(s)
- Adam Bownik
- Department of Hydrobiology and Protection of Ecosystems, Faculty of Environmental Biology, University of Life Sciences, Lublin, Poland
| | | |
Collapse
|
46
|
Xu D, Mo J, Xie X, Hu N. In-Cell Nanoelectronics: Opening the Door to Intracellular Electrophysiology. NANO-MICRO LETTERS 2021; 13:127. [PMID: 34138366 PMCID: PMC8124030 DOI: 10.1007/s40820-021-00655-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/13/2021] [Indexed: 05/07/2023]
Abstract
Establishing a reliable electrophysiological recording platform is crucial for cardiology and neuroscience research. Noninvasive and label-free planar multitransistors and multielectrode arrays are conducive to perform the large-scale cellular electrical activity recordings, but the signal attenuation limits these extracellular devices to record subthreshold activities. In recent decade, in-cell nanoelectronics have been rapidly developed to open the door to intracellular electrophysiology. With the unique three-dimensional nanotopography and advanced penetration strategies, high-throughput and high-fidelity action potential like signal recordings is expected to be realized. This review summarizes in-cell nanoelectronics from versatile nano-biointerfaces, penetration strategies, active/passive nanodevices, systematically analyses the applications in electrogenic cells and especially evaluates the influence of nanodevices on the high-quality intracellular electrophysiological signals. Further, the opportunities, challenges and broad prospects of in-cell nanoelectronics are prospected, expecting to promote the development of in-cell electrophysiological platforms to meet the demand of theoretical investigation and clinical application.
Collapse
Affiliation(s)
- Dongxin Xu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Jingshan Mo
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
- The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Ning Hu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China.
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China.
| |
Collapse
|
47
|
Passaro AP, Aydin O, Saif MTA, Stice SL. Development of an objective index, neural activity score (NAS), reveals neural network ontogeny and treatment effects on microelectrode arrays. Sci Rep 2021; 11:9110. [PMID: 33907294 PMCID: PMC8079414 DOI: 10.1038/s41598-021-88675-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 02/16/2021] [Indexed: 02/06/2023] Open
Abstract
Microelectrode arrays (MEAs) are valuable tools for electrophysiological analysis, providing assessment of neural network health and development. Analysis can be complex, however, requiring intensive processing of large data sets consisting of many activity parameters, leading to information loss as studies subjectively report relatively few metrics in the interest of simplicity. In screening assays, many groups report simple overall activity (i.e. firing rate) but omit network connectivity changes (e.g. burst characteristics and synchrony) that may not be evident from basic parameters. Our goal was to develop an objective process to capture most of the valuable information gained from MEAs in neural development and toxicity studies. We implemented principal component analysis (PCA) to reduce the high dimensionality of MEA data. Upon analysis, we found the first principal component was strongly correlated to time, representing neural culture development; therefore, factor loadings were used to create a single index score-named neural activity score (NAS)-reflecting neural maturation. For validation, we applied NAS to studies analyzing various treatments. In all cases, NAS accurately recapitulated expected results, suggesting viability of NAS to measure network health and development. This approach may be adopted by other researchers using MEAs to analyze complicated treatment effects and multicellular interactions.
Collapse
Affiliation(s)
- Austin P. Passaro
- grid.213876.90000 0004 1936 738XRegenerative Bioscience Center, University of Georgia, Athens, GA USA ,grid.213876.90000 0004 1936 738XDivision of Neuroscience, Biomedical Health and Sciences Institute, University of Georgia, Athens, GA USA
| | - Onur Aydin
- grid.35403.310000 0004 1936 9991Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL USA
| | - M. Taher A. Saif
- grid.35403.310000 0004 1936 9991Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL USA
| | - Steven L. Stice
- grid.213876.90000 0004 1936 738XRegenerative Bioscience Center, University of Georgia, Athens, GA USA ,grid.213876.90000 0004 1936 738XDivision of Neuroscience, Biomedical Health and Sciences Institute, University of Georgia, Athens, GA USA
| |
Collapse
|
48
|
Bayat FK, Polat Budak B, Yiğit EN, Öztürk G, Gülçür HÖ, Güveniş A. Adult mouse dorsal root ganglia neurons form aberrant glutamatergic connections in dissociated cultures. PLoS One 2021; 16:e0246924. [PMID: 33657119 PMCID: PMC7928449 DOI: 10.1371/journal.pone.0246924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 01/29/2021] [Indexed: 11/18/2022] Open
Abstract
Cultured sensory neurons can exhibit complex activity patterns following stimulation in terms of increased excitability and interconnected responses of multiple neurons. Although these complex activity patterns suggest a network-like configuration, research so far had little interest in synaptic network formation ability of the sensory neurons. To identify interaction profiles of Dorsal Root Ganglia (DRG) neurons and explore their putative connectivity, we developed an in vitro experimental approach. A double transgenic mouse model, expressing genetically encoded calcium indicator (GECI) in their glutamatergic neurons, was produced. Dissociated DRG cultures from adult mice were prepared with a serum-free protocol and no additional growth factors or cytokines were utilized for neuronal sensitization. DRG neurons were grown on microelectrode arrays (MEA) to induce stimulus-evoked activity with a modality-free stimulation strategy. With an almost single-cell level electrical stimulation, spontaneous and evoked activity of GCaMP6s expressing neurons were detected under confocal microscope. Typical responses were analyzed, and correlated calcium events were detected across individual DRG neurons. Next, correlated responses were successfully blocked by glutamatergic receptor antagonists, which indicated functional synaptic coupling. Immunostaining confirmed the presence of synapses mainly in the axonal terminals, axon-soma junctions and axon-axon intersection sites. Concisely, the results presented here illustrate a new type of neuron-to-neuron interaction in cultured DRG neurons conducted through synapses. The developed assay can be a valuable tool to analyze individual and collective responses of the cultured sensory neurons.
Collapse
Affiliation(s)
- F. Kemal Bayat
- Institute of Biomedical Engineering, Bogazici University, İstanbul, Turkey
- Department of Electrical and Electronics Engineering, Faculty of Engineering, Marmara University, İstanbul, Turkey
| | - Betul Polat Budak
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, İstanbul, Turkey
- Faculty of Engineering and Natural Sciences, Biruni University, İstanbul, Turkey
| | - Esra Nur Yiğit
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, İstanbul, Turkey
- Institute of Biotechnology, Gebze Technical University, İzmit, Turkey
| | - Gürkan Öztürk
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, İstanbul, Turkey
| | - Halil Özcan Gülçür
- Institute of Biomedical Engineering, Bogazici University, İstanbul, Turkey
- Faculty of Engineering and Natural Sciences, Biruni University, İstanbul, Turkey
- * E-mail:
| | - Albert Güveniş
- Institute of Biomedical Engineering, Bogazici University, İstanbul, Turkey
| |
Collapse
|
49
|
Tsunemoto K, Yamada S, Kanda Y. [Current challenges and future perspectives of iPSC-based neurotoxicity testing]. Nihon Yakurigaku Zasshi 2021; 156:107-113. [PMID: 33642528 DOI: 10.1254/fpj.20097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Predicting drug-induced side effects in central nervous system is important because they can lead to the discontinuation of new drugs/candidates or the withdrawal of marketed drugs. Although many efforts are made, evaluation system using animals have not been highly predictive in humans. In addition, animal experiments are time-consuming and costly. To address these issues, in vitro evaluation methods, such as the use of New Approach Methodologies (NAM) have been explored. Human iPS cell technology has already been applied to assess drug-induced cardiotoxicity. In addition, the use of human iPS cell technology and in silico has been promoted for neurotoxicity assessment during the developmental neurotoxicity in terms of chemical safety issues. Organization for Economic Cooperation and Development (OECD) guidance regarding developmental neurotoxicity is under preparation. In this review, we will review the current trends in safety assessment methods for the central nervous system in light of these international trends.
Collapse
Affiliation(s)
| | - Shigeru Yamada
- Division of Pharmacology, National Institute of Health Sciences (NIHS)
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences (NIHS)
| |
Collapse
|
50
|
Kasteel EEJ, Westerink RHS. Refining in vitro and in silico neurotoxicity approaches by accounting for interspecies and interindividual differences in toxicodynamics. Expert Opin Drug Metab Toxicol 2021; 17:1007-1017. [PMID: 33586568 DOI: 10.1080/17425255.2021.1885647] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION The process of chemical risk assessment traditionally relies on animal experiments and associated default uncertainty factors to account for interspecies and interindividual differences. To work toward a more precise and personalized risk assessment, these uncertainty factors should be refined and replaced by chemical-specific adjustment factors (CSAFs). AREAS COVERED This concise review discusses alternative (in vitro/in silico) approaches that can be used to assess interspecies and interindividual differences in toxicodynamics, ranging from targeted to more integrated approaches. Although data are available on interspecies differences, the increasing use of human-induced pluripotent stem cell (hiPSC)-derived neurons may provide opportunities to also assess interindividual variability in neurotoxicity. More integrated approaches, like adverse outcome pathways (AOPs) can provide a more quantitative understanding of the toxicodynamics of a chemical. EXPERT OPINION To improve chemical risk assessment, refinement of uncertainty factors is crucial. In vitro and in silico models can facilitate the development of CSAFs, but still these models cannot always capture the complexity of the in vivo situation, thereby potentially hampering regulatory acceptance. The combined use of more integrated approaches, like AOPs and physiologically based kinetic models, can aid in structuring data and increasing suitability of alternative approaches for regulatory purposes.
Collapse
Affiliation(s)
- Emma E J Kasteel
- Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Remco H S Westerink
- Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|