1
|
Lai D, Sosicka P, Williams DJ, Bowyer ME, Ressler AK, Kohrt SE, Muron SJ, Crino PB, Freeze HH, Boland MJ, Heinzen EL. SLC35A2 loss of function variants affect glycomic signatures, neuronal fate, and network dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.27.630524. [PMID: 39763953 PMCID: PMC11703275 DOI: 10.1101/2024.12.27.630524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
SLC35A2 encodes a UDP-galactose transporter essential for glycosylation of proteins and galactosylation of lipids and glycosaminoglycans. Germline genetic SLC35A2 variants have been identified in congenital disorders of glycosylation and somatic SLC35A2 variants have been linked to intractable epilepsy associated with malformations of cortical development. However, the functional consequences of these pathogenic variants on brain development and network integrity remain elusive. In this study, we use an isogenic human induced pluripotent stem cell-derived neuron model to comprehensively interrogate the functional impact of loss of function variants in SLC35A2 through the integration of cellular and molecular biology, protein glycosylation analysis, neural network dynamics, and single cell electrophysiology. We show that loss of function variants in SLC35A2 result in disrupted glycomic signatures and precocious neurodevelopment, yielding hypoactive, asynchronous neural networks. This aberrant network activity is attributed to an inhibitory/excitatory imbalance as characterization of neural composition revealed preferential differentiation of SLC35A2 loss of function variants towards the GABAergic fate. Additionally, electrophysiological recordings of synaptic activity reveal a shift in excitatory/inhibitory balance towards increased inhibitory drive, indicating changes occurring specifically at the pre-synaptic terminal. Our study is the first to provide mechanistic insight regarding the early development and functional connectivity of SLC35A2 loss of function variant harboring human neurons, providing important groundwork for future exploration of potential therapeutic interventions.
Collapse
Affiliation(s)
- Dulcie Lai
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Paulina Sosicka
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Damian J Williams
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - MaryAnn E Bowyer
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Andrew K Ressler
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Sarah E Kohrt
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Savannah J Muron
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Peter B Crino
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Hudson H Freeze
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Michael J Boland
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Center for Epilepsy and Neurodevelopmental Disorders, Perelman School of Medicine, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Erin L Heinzen
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
2
|
Genocchi B, Ahtiainen A, Niemi A, Barros MT, Tanskanen JMA, Lenk K, Hyttinen J, Puthanmadam Subramaniyam N. Astrocytes induce desynchronization and reduce predictability in neuron-astrocyte networks cultured on microelectrode arrays. ROYAL SOCIETY OPEN SCIENCE 2024; 11:240839. [PMID: 39479242 PMCID: PMC11521599 DOI: 10.1098/rsos.240839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024]
Abstract
In this article, we aim to study how astrocytes control electrophysiological activity during neuronal network formation. We used a combination of spike/burst analysis, classification of spike waveforms based on various signal properties and tools from information theory to demonstrate how astrocytes modulate the electrical activity of neurons using microelectrode array (MEA) signals. We cultured rat primary cortical neurons and astrocytes on 60-electrode MEAs with different neuron/astrocyte ratios ranging from 'pure' neuronal cultures to co-cultures containing 50% neurons and 50% astrocytes. Our results show that astrocytes desynchronize the network and reduce predictability in the signals and affect the repolarization phase of the action potentials. Our work highlights that it is crucial to go beyond standard MEA analysis to assess how astrocytes control neuronal cultures and investigate any dysfunction that could potentially result in neuronal hyperactivity.
Collapse
Affiliation(s)
- Barbara Genocchi
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Annika Ahtiainen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Annika Niemi
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Michael T. Barros
- School of Computer Science and Electronic Engineering, University of Essex, Colchester, Essex, UK
| | | | - Kerstin Lenk
- Institute of Neural Engineering, Graz University of Technology, Graz, Austria
- BioTechMed, Graz, Austria
| | - Jari Hyttinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | |
Collapse
|
3
|
Lemieux MR, Freigassner B, Hanson JL, Thathey Z, Opp MR, Hoeffer CA, Link CD. Multielectrode array characterization of human induced pluripotent stem cell derived neurons in co-culture with primary human astrocytes. PLoS One 2024; 19:e0303901. [PMID: 38917115 PMCID: PMC11198861 DOI: 10.1371/journal.pone.0303901] [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: 01/23/2024] [Accepted: 05/02/2024] [Indexed: 06/27/2024] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) derived into neurons offer a powerful in vitro model to study cellular processes. One method to characterize functional network properties of these cells is using multielectrode arrays (MEAs). MEAs can measure the electrophysiological activity of cellular cultures for extended periods of time without disruption. Here we used WTC11 hiPSCs with a doxycycline-inducible neurogenin 2 (NGN2) transgene differentiated into neurons co-cultured with primary human astrocytes. We achieved a synchrony index ∼0.9 in as little as six-weeks with a mean firing rate of ∼13 Hz. Previous reports show that derived 3D brain organoids can take several months to achieve similar strong network burst synchrony. We also used this co-culture to model aspects of blood-brain barrier breakdown by using human serum. Our fully human co-culture achieved strong network burst synchrony in a fraction of the time of previous reports, making it an excellent first pass, high-throughput method for studying network properties and neurodegenerative diseases.
Collapse
Affiliation(s)
- Maddie R. Lemieux
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Bernhard Freigassner
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Jessica L. Hanson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States of America
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Zahra Thathey
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Mark R. Opp
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Charles A. Hoeffer
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States of America
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Christopher D. Link
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States of America
| |
Collapse
|
4
|
Lemieux MR, Freigassner B, Thathey Z, Opp MR, Hoeffer CA, Link CD. Multielectrode array characterization of human induced pluripotent stem cell derived neurons in co-culture with primary human astrocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583341. [PMID: 38496655 PMCID: PMC10942372 DOI: 10.1101/2024.03.04.583341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Human induced pluripotent stem cells (hiPSCs) derived into neurons offer a powerful in vitro model to study cellular processes. One method to characterize functional network properties of these cells is using multielectrode arrays (MEAs). MEAs can measure the electrophysiological activity of cellular cultures for extended periods of time without disruption. Here we used WTC11 hiPSCs with a doxycycline-inducible neurogenin 2 (NGN2) transgene differentiated into neurons co-cultured with primary human astrocytes. We achieved a synchrony index ~0.9 in as little as six-weeks with a mean firing rate of ~13 Hz. Previous reports show that derived 3D brain organoids can take several months to achieve similar strong network burst synchrony. We also used this co-culture to model aspects of sporadic Alzheimer's disease by mimicking blood-brain barrier breakdown using a human serum. Our fully human co-culture achieved strong network burst synchrony in a fraction of the time of previous reports, making it an excellent first pass, high-throughput method for studying network properties and neurodegenerative diseases.
Collapse
Affiliation(s)
- Maddie R Lemieux
- Department of Integrative Physiology, University of Colorado Boulder
| | | | - Zahra Thathey
- Department of Integrative Physiology, University of Colorado Boulder
| | - Mark R Opp
- Department of Integrative Physiology, University of Colorado Boulder
| | - Charles A Hoeffer
- Department of Integrative Physiology, University of Colorado Boulder
- Institute for Behavioral Genetics, University of Colorado Boulder
| | | |
Collapse
|
5
|
Sakaibara M, Yamamoto H, Murota H, Monma N, Sato S, Hirano-Iwata A. Enhanced responses to inflammatory cytokine interleukin-6 in micropatterned networks of cultured cortical neurons. Biochem Biophys Res Commun 2024; 695:149379. [PMID: 38159413 DOI: 10.1016/j.bbrc.2023.149379] [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: 11/28/2023] [Accepted: 12/09/2023] [Indexed: 01/03/2024]
Abstract
Cortical neurons in dissociated cultures are an indispensable model system for pharmacological research that provides insights into chemical responses in well-defined environments. However, cortical neurons plated on homogeneous substrates develop an unstructured network that exhibits excessively synchronized activity, which occasionally masks the consequences induced by external substances. Here, we show that hyperactivity and excessive synchrony in cultured cortical networks can be effectively suppressed by growing neurons in microfluidic devices. These devices feature a hierarchically modular design that resembles the in vivo network. We focused on interleukin-6, a pro-inflammatory cytokine, and assessed its acute and chronic effects. Fluorescence calcium imaging of spontaneous neural activity for up to 20 days of culture showed detectable modulation of collective activity events and neural correlation in micropatterned neurons, which was not apparent in neurons cultured on homogeneous substrates. Our results indicate that engineered neuronal networks provide a unique platform for detecting and understanding the fundamental effects of biochemical compounds on neuronal networks.
Collapse
Affiliation(s)
- Mamoru Sakaibara
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Hideaki Yamamoto
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan.
| | - Hakuba Murota
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Nobuaki Monma
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Shigeo Sato
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Ayumi Hirano-Iwata
- Research Institute of Electrical Communication, Tohoku University, Sendai, Japan; Graduate School of Engineering, Tohoku University, Sendai, Japan; Advanced Institute for Materials Research, Tohoku University, Sendai, Japan; Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| |
Collapse
|
6
|
Prytkova I, Liu Y, Fernando M, Gameiro-Ros I, Popova D, Kamarajan C, Xuei X, Chorlian DB, Edenberg HJ, Tischfield JA, Porjesz B, Pang ZP, Hart RP, Goate A, Slesinger PA. Upregulated GIRK2 counteracts ethanol-induced changes in excitability & respiration in human neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.22.533236. [PMID: 36993693 PMCID: PMC10055374 DOI: 10.1101/2023.03.22.533236] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Genome-wide association analysis (GWAS) of electroencephalographic endophenotypes for alcohol use disorder (AUD) has identified non-coding polymorphisms within the KCNJ6 gene. KCNJ6 encodes GIRK2, a subunit of a G protein-coupled inwardly-rectifying potassium channel that regulates neuronal excitability. How changes in GIRK2 affect human neuronal excitability and the response to repeated ethanol exposure is poorly understood. Here, we studied the effect of upregulating KCNJ6 using an isogenic approach with human glutamatergic neurons derived from induced pluripotent stem cells (male and female donors). Using multi-electrode-arrays, population calcium imaging, single-cell patch-clamp electrophysiology, and mitochondrial stress tests, we find that elevated GIRK2 acts in concert with 7-21 days of ethanol exposure to inhibit neuronal activity, to counteract ethanol-induced increases in glutamate response, and to promote an increase intrinsic excitability. Furthermore, elevated GIRK2 prevented ethanol-dependent changes in basal and activity-dependent mitochondrial respiration. These data support a role for GIRK2 in mitigating the effects of ethanol and a previously unknown connection to mitochondrial function in human glutamatergic neurons. SIGNIFICANCE STATEMENT Alcohol use disorder (AUD) is a major health problem that has worsened since COVID, affecting over 100 million people worldwide. While it is known that heritability contributes to AUD, specific genes and their role in neuronal function remain poorly understood, especially in humans. In the current manuscript, we focused on the inwardly-rectifying potassium channel GIRK2, which has been identified in an AUD-endophenotype genome-wide association study. We used human excitatory neurons derived from healthy donors to study the impact of GIRK2 expression. Our results reveal that elevated GIRK2 counteracts ethanol-induced increases in glutamate response and intracellular calcium, as well as deficits in activity-dependent mitochondrial respiration. The role of GIRK2 in mitigating ethanol-induced hyper-glutamatergic and mitochondrial offers therapeutic promise for treating AUD.
Collapse
|
7
|
Ishibashi Y, Nagafuku N, Kanda Y, Suzuki I. Evaluation of neurotoxicity for pesticide-related compounds in human iPS cell-derived neurons using microelectrode array. Toxicol In Vitro 2023; 93:105668. [PMID: 37633473 DOI: 10.1016/j.tiv.2023.105668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/30/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
In vivo evaluations of chemicals in neurotoxicity have certain limitations due to the considerable time and cost required, necessity of extrapolation from rodents to humans, and limited information on toxicity mechanisms. To address this issue, the development of in vitro test methods using new approach methodologies (NAMs) is important to evaluate the chemicals in neurotoxicity. Microelectrode array (MEA) allows the assessment of changes in neural network activity caused by compound administration. However, studies on compound evaluation criteria are scarce. In this study, we evaluated the impact of pesticides on neural activity using MEA measurements of human iPSC-derived neurons. A principal component analysis was performed on the electrical physiological parameters obtained by MEA measurements, and the influence of excessive neural activity due to compound addition was defined using the standard deviation of neural activity with solvent addition as the reference. By using known seizurogenic compounds as positive controls for neurotoxicity in MEA and evaluating pesticides with insufficient verification of their neurotoxicity in humans, we demonstrated that these pesticides exhibit neurotoxicity in humans. In conclusion, our data suggest that the neurotoxicity evaluation method in human iPSC neurons using MEA measurements could be one of the in vitro neurotoxicity test methods that could replace animal experiments.
Collapse
Affiliation(s)
- Yuto Ishibashi
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi 982-8577, Japan
| | - Nami Nagafuku
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi 982-8577, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan
| | - Ikuro Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi 982-8577, Japan.
| |
Collapse
|
8
|
Lv S, He E, Luo J, Liu Y, Liang W, Xu S, Zhang K, Yang Y, Wang M, Song Y, Wu Y, Cai X. Using Human-Induced Pluripotent Stem Cell Derived Neurons on Microelectrode Arrays to Model Neurological Disease: A Review. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301828. [PMID: 37863819 PMCID: PMC10667858 DOI: 10.1002/advs.202301828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 09/04/2023] [Indexed: 10/22/2023]
Abstract
In situ physiological signals of in vitro neural disease models are essential for studying pathogenesis and drug screening. Currently, an increasing number of in vitro neural disease models are established using human-induced pluripotent stem cell (hiPSC) derived neurons (hiPSC-DNs) to overcome interspecific gene expression differences. Microelectrode arrays (MEAs) can be readily interfaced with two-dimensional (2D), and more recently, three-dimensional (3D) neural stem cell-derived in vitro models of the human brain to monitor their physiological activity in real time. Therefore, MEAs are emerging and useful tools to model neurological disorders and disease in vitro using human iPSCs. This is enabling a real-time window into neuronal signaling at the network scale from patient derived. This paper provides a comprehensive review of MEA's role in analyzing neural disease models established by hiPSC-DNs. It covers the significance of MEA fabrication, surface structure and modification schemes for hiPSC-DNs culturing and signal detection. Additionally, this review discusses advances in the development and use of MEA technology to study in vitro neural disease models, including epilepsy, autism spectrum developmental disorder (ASD), and others established using hiPSC-DNs. The paper also highlights the application of MEAs combined with hiPSC-DNs in detecting in vitro neurotoxic substances. Finally, the future development and outlook of multifunctional and integrated devices for in vitro medical diagnostics and treatment are discussed.
Collapse
Affiliation(s)
- Shiya Lv
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Enhui He
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
- The State Key Lab of Brain‐Machine IntelligenceZhejiang UniversityHangzhou321100China
| | - Jinping Luo
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yaoyao Liu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wei Liang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Shihong Xu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Kui Zhang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yan Yang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Mixia Wang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yilin Song
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yirong Wu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Xinxia Cai
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
9
|
Ishibashi Y, Nagafuku N, Kinoshita K, Okamura A, Shirakawa T, Suzuki I. Verification of the seizure liability of compounds based on their in vitro functional activity in cultured rat cortical neurons and co-cultured human iPSC-derived neurons with astrocytes and in vivo extrapolation to cerebrospinal fluid concentration. Toxicol Appl Pharmacol 2023; 476:116675. [PMID: 37661062 DOI: 10.1016/j.taap.2023.116675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/26/2023] [Accepted: 08/30/2023] [Indexed: 09/05/2023]
Abstract
Methodical screening of safe and efficient drug candidate compounds is crucial for drug development. A high-throughput and accurate compound evaluation method targeting the central nervous system can be developed using in vitro neural networks. In particular, an evaluation system based on a human-derived neural network that can act as an alternative to animal experiments is desirable to avoid interspecific differences. A microelectrode array (MEA) is one such evaluation system, and can measure in vitro neural activity; however, studies on compound evaluation criteria and in vitro to in vivo extrapolation are scarce. In this study, we identified the parameters that can eliminate the effects of solvents from neural activity data obtained using MEA allow for accurate compound evaluation. Additionally, we resolved the issue associated with compound evaluation criteria during MEA using principal component analysis by considering the neuronal activity exceeding standard deviation (SD) of the solvent as indicator of seizurogenic potential. Overall, 10 seizurogenic compounds and three negative controls were assessed using MEA-based co-cultured human-induced pluripotent stem cell-derived neurons and astrocytes, and primary rat cortical neurons. In addition, we determined rat cerebrospinal fluid (CSF) concentrations during tremor and convulsion in response to exposure to test compounds. To characterize the in vitro to in vivo extrapolation and species differences, we compared the concentrations at which neuronal activity exceeding the SD range of the solvent was detectable using the MEA system and rat CSF concentration.
Collapse
Affiliation(s)
- Y Ishibashi
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi 982-8577, Japan
| | - N Nagafuku
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi 982-8577, Japan
| | - K Kinoshita
- Drug Safety Research Labs, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - A Okamura
- Drug Safety Research Labs, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - T Shirakawa
- Drug Safety Research Labs, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - I Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi 982-8577, Japan.
| |
Collapse
|
10
|
Gameiro‐Ros I, Popova D, Prytkova I, Pang ZP, Liu Y, Dick D, Bucholz KK, Agrawal A, Porjesz B, Goate AM, Xuei X, Kamarajan C, Tischfield JA, Edenberg HJ, Slesinger PA, Hart RP. 5. Collaborative Study on the Genetics of Alcoholism: Functional genomics. GENES, BRAIN, AND BEHAVIOR 2023; 22:e12855. [PMID: 37533187 PMCID: PMC10550792 DOI: 10.1111/gbb.12855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/31/2023] [Accepted: 06/17/2023] [Indexed: 08/04/2023]
Abstract
Alcohol Use Disorder is a complex genetic disorder, involving genetic, neural, and environmental factors, and their interactions. The Collaborative Study on the Genetics of Alcoholism (COGA) has been investigating these factors and identified putative alcohol use disorder risk genes through genome-wide association studies. In this review, we describe advances made by COGA in elucidating the functional changes induced by alcohol use disorder risk genes using multimodal approaches with human cell lines and brain tissue. These studies involve investigating gene regulation in lymphoblastoid cells from COGA participants and in post-mortem brain tissues. High throughput reporter assays are being used to identify single nucleotide polymorphisms in which alternate alleles differ in driving gene expression. Specific single nucleotide polymorphisms (both coding or noncoding) have been modeled using induced pluripotent stem cells derived from COGA participants to evaluate the effects of genetic variants on transcriptomics, neuronal excitability, synaptic physiology, and the response to ethanol in human neurons from individuals with and without alcohol use disorder. We provide a perspective on future studies, such as using polygenic risk scores and populations of induced pluripotent stem cell-derived neurons to identify signaling pathways related with responses to alcohol. Starting with genes or loci associated with alcohol use disorder, COGA has demonstrated that integration of multimodal data within COGA participants and functional studies can reveal mechanisms linking genomic variants with alcohol use disorder, and potential targets for future treatments.
Collapse
Affiliation(s)
- Isabel Gameiro‐Ros
- Nash Family Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Dina Popova
- Human Genetics Institute of New JerseyRutgers UniversityPiscatawayNew JerseyUSA
| | - Iya Prytkova
- Nash Family Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Zhiping P. Pang
- Human Genetics Institute of New JerseyRutgers UniversityPiscatawayNew JerseyUSA
- Child Health Institute of New Jersey and Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical SchoolRutgers UniversityNew BrunswickNew JerseyUSA
| | - Yunlong Liu
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Danielle Dick
- Rutgers Addiction Research Center, Robert Wood Johnson Medical SchoolRutgers UniversityPiscatawayNew JerseyUSA
| | - Kathleen K. Bucholz
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
| | - Arpana Agrawal
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
| | - Bernice Porjesz
- Department of Psychiatry and Behavioral SciencesSUNY Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | - Alison M. Goate
- Nash Family Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Xiaoling Xuei
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Chella Kamarajan
- Department of Psychiatry and Behavioral SciencesSUNY Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | | | - Jay A. Tischfield
- Human Genetics Institute of New JerseyRutgers UniversityPiscatawayNew JerseyUSA
- Department of GeneticsRutgers UniversityPiscatawayNew JerseyUSA
| | - Howard J. Edenberg
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana UniversityIndianapolisIndianaUSA
| | - Paul A. Slesinger
- Nash Family Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ronald P. Hart
- Human Genetics Institute of New JerseyRutgers UniversityPiscatawayNew JerseyUSA
- Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNew JerseyUSA
| |
Collapse
|
11
|
Suzuki I, Matsuda N, Han X, Noji S, Shibata M, Nagafuku N, Ishibashi Y. Large-Area Field Potential Imaging Having Single Neuron Resolution Using 236 880 Electrodes CMOS-MEA Technology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023:e2207732. [PMID: 37088859 PMCID: PMC10369302 DOI: 10.1002/advs.202207732] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/21/2023] [Indexed: 05/03/2023]
Abstract
The electrophysiological technology having a high spatiotemporal resolution at the single-cell level and noninvasive measurements of large areas provide insights on underlying neuronal function. Here, a complementary metal-oxide semiconductor (CMOS)-microelectrode array (MEA) is used that uses 236 880 electrodes each with an electrode size of 11.22 × 11.22 µm and 236 880 covering a wide area of 5.5 × 5.9 mm in presenting a detailed and single-cell-level neural activity analysis platform for brain slices, human iPS cell-derived cortical networks, peripheral neurons, and human brain organoids. Propagation pattern characteristics between brain regions changes the synaptic propagation into compounds based on single-cell time-series patterns, classification based on single DRG neuron firing patterns and compound responses, axonal conduction characteristics and changes to anticancer drugs, and network activities and transition to compounds in brain organoids are extracted. This detailed analysis of neural activity at the single-cell level using the CMOS-MEA provides a new understanding of the basic mechanisms of brain circuits in vitro and ex vivo, on human neurological diseases for drug discovery, and compound toxicity assessment.
Collapse
Affiliation(s)
- Ikuro Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Naoki Matsuda
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Xiaobo Han
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Shuhei Noji
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Mikako Shibata
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Nami Nagafuku
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Yuto Ishibashi
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| |
Collapse
|
12
|
Lu HR, Seo M, Kreir M, Tanaka T, Yamoto R, Altrocchi C, van Ammel K, Tekle F, Pham L, Yao X, Teisman A, Gallacher DJ. High-Throughput Screening Assay for Detecting Drug-Induced Changes in Synchronized Neuronal Oscillations and Potential Seizure Risk Based on Ca 2+ Fluorescence Measurements in Human Induced Pluripotent Stem Cell (hiPSC)-Derived Neuronal 2D and 3D Cultures. Cells 2023; 12:cells12060958. [PMID: 36980298 PMCID: PMC10046961 DOI: 10.3390/cells12060958] [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: 02/03/2023] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Drug-induced seizure liability is a significant safety issue and the basis for attrition in drug development. Occurrence in late development results in increased costs, human risk, and delayed market availability of novel therapeutics. Therefore, there is an urgent need for biologically relevant, in vitro high-throughput screening assays (HTS) to predict potential risks for drug-induced seizure early in drug discovery. We investigated drug-induced changes in neural Ca2+ oscillations, using fluorescent dyes as a potential indicator of seizure risk, in hiPSC-derived neurons co-cultured with human primary astrocytes in both 2D and 3D forms. The dynamics of synchronized neuronal calcium oscillations were measured with an FDSS kinetics reader. Drug responses in synchronized Ca2+ oscillations were recorded in both 2D and 3D hiPSC-derived neuron/primary astrocyte co-cultures using positive controls (4-aminopyridine and kainic acid) and negative control (acetaminophen). Subsequently, blinded tests were carried out for 25 drugs with known clinical seizure incidence. Positive predictive value (accuracy) based on significant changes in the peak number of Ca2+ oscillations among 25 reference drugs was 91% in 2D vs. 45% in 3D hiPSC-neuron/primary astrocyte co-cultures. These data suggest that drugs that alter neuronal activity and may have potential risk for seizures can be identified with high accuracy using an HTS approach using the measurements of Ca2+ oscillations in hiPSC-derived neurons co-cultured with primary astrocytes in 2D.
Collapse
Affiliation(s)
- Hua-Rong Lu
- Global Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen R&D, A Division of Janssen Pharmaceutica NV, B-2340 Beerse, Belgium
| | - Manabu Seo
- Elixirgen Scientific, Incorporated, Baltimore, MD 21205, USA
| | - Mohamed Kreir
- Global Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen R&D, A Division of Janssen Pharmaceutica NV, B-2340 Beerse, Belgium
| | - Tetsuya Tanaka
- Elixirgen Scientific, Incorporated, Baltimore, MD 21205, USA
| | - Rie Yamoto
- Healthcare Business Group, Drug Discovery Business Department, Ricoh Company Ltd., Tokyo 143-8555, Japan
| | - Cristina Altrocchi
- Global Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen R&D, A Division of Janssen Pharmaceutica NV, B-2340 Beerse, Belgium
| | - Karel van Ammel
- Global Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen R&D, A Division of Janssen Pharmaceutica NV, B-2340 Beerse, Belgium
| | - Fetene Tekle
- Statistics and Decision Sciences, Global Development, Janssen R&D, A Division of Janssen Pharmaceutica NV, B-2340 Beerse, Belgium
| | - Ly Pham
- Computational Biology & Toxicology, Preclinical Sciences and Translational Safety, A Division of Janssen Pharmaceutica NV, San Diego, CA 921921, USA
| | - Xiang Yao
- Computational Biology & Toxicology, Preclinical Sciences and Translational Safety, A Division of Janssen Pharmaceutica NV, San Diego, CA 921921, USA
| | - Ard Teisman
- Global Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen R&D, A Division of Janssen Pharmaceutica NV, B-2340 Beerse, Belgium
| | - David J Gallacher
- Global Safety Pharmacology, Preclinical Sciences and Translational Safety, Janssen R&D, A Division of Janssen Pharmaceutica NV, B-2340 Beerse, Belgium
| |
Collapse
|
13
|
Lawson J, LaVancher E, DeAlmeida M, Black BJ. Electrically-evoked oscillating calcium transients in mono- and co-cultures of iPSC glia and sensory neurons. Front Cell Neurosci 2023; 17:1094070. [PMID: 37006467 PMCID: PMC10060658 DOI: 10.3389/fncel.2023.1094070] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/17/2023] [Indexed: 03/18/2023] Open
Abstract
Activated glia are known to exhibit either neuroprotective or neurodegenerative effects, depending on their phenotype, while participating in chronic pain regulation. Until recently, it has been believed that satellite glial cells and astrocytes are electrically slight and process stimuli only through intracellular calcium flux that triggers downstream signaling mechanisms. Though glia do not exhibit action potentials, they do express both voltage- and ligand-gated ion channels that facilitate measurable calcium transients, a measure of their own phenotypic excitability, and support and modulate sensory neuron excitability through ion buffering and secretion of excitatory or inhibitory neuropeptides (i.e., paracrine signaling). We recently developed a model of acute and chronic nociception using co-cultures of iPSC sensory neurons (SN) and spinal astrocytes on microelectrode arrays (MEAs). Until recently, only neuronal extracellular activity has been recorded using MEAs with a high signal-to-noise ratio and in a non-invasive manner. Unfortunately, this method has limited compatibility with simultaneous calcium transient imaging techniques, which is the most common method for monitoring the phenotypic activity of astrocytes. Moreover, both dye-based and genetically encoded calcium indicator imaging rely on calcium chelation, affecting the culture's long-term physiology. Therefore, it would be ideal to allow continuous and simultaneous direct phenotypic monitoring of both SNs and astrocytes in a high-to-moderate throughput non-invasive manner and would significantly advance the field of electrophysiology. Here, we characterize astrocytic oscillating calcium transients (OCa2+Ts) in mono- and co-cultures of iPSC astrocytes as well as iPSC SN-astrocyte co-cultures on 48 well plate MEAs. We demonstrate that astrocytes exhibit OCa2+Ts in an electrical stimulus amplitude- and duration-dependent manner. We show that OCa2+Ts can be pharmacologically inhibited with the gap junction antagonist, carbenoxolone (100 μM). Most importantly, we demonstrate that both neurons and glia can be phenotypically characterized in real time, repeatedly, over the duration of the culture. In total, our findings suggest that calcium transients in glial populations may serve as a stand-alone or supplemental screening technique for identifying potential analgesics or compounds targeting other glia-mediated pathologies.
Collapse
Affiliation(s)
| | | | | | - Bryan James Black
- Department of Biomedical Engineering, Francis College of Engineering, University of Massachusetts Lowell, Lowell, MA, United States
| |
Collapse
|
14
|
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: 7] [Impact Index Per Article: 3.5] [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
|
15
|
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
|
16
|
Whye D, Wood D, Saber WA, Norabuena EM, Makhortova NR, Sahin M, Buttermore ED. A Robust Pipeline for the Multi-Stage Accelerated Differentiation of Functional 3D Cortical Organoids from Human Pluripotent Stem Cells. Curr Protoc 2023; 3:e641. [PMID: 36633423 PMCID: PMC9839317 DOI: 10.1002/cpz1.641] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Disordered cellular development, abnormal neuroanatomical formations, and dysfunction of neuronal circuitry are among the pathological manifestations of cortical regions in the brain that are often implicated in complex neurodevelopmental disorders. With the advancement of stem cell methodologies such as cerebral organoid generation, it is possible to study these processes in vitro using 3D cellular platforms that mirror key developmental stages occurring throughout embryonic neurogenesis. Patterning-based stem cell models of directed neuronal development offer one approach to accomplish this, but these protocols often require protracted periods of cell culture to generate diverse cell types and current methods are plagued by a lack of specificity, reproducibility, and temporal control over cell derivation. Although ectopic expression of transcription factors offers another avenue to rapidly generate neurons, this process of direct lineage conversion bypasses critical junctures of neurodevelopment during which disease-relevant manifestations may occur. Here, we present a directed differentiation approach for generating human pluripotent stem cell (hPSC)-derived cortical organoids with accelerated lineage specification to generate functionally mature cortical neurons in a shorter timeline than previously established protocols. This novel protocol provides precise guidance for the specification of neuronal cell type identity as well as temporal control over the pace at which cortical lineage trajectories are established. Furthermore, we present assays that can be used as tools to interrogate stage-specific developmental signaling mechanisms. By recapitulating major components of embryonic neurogenesis, this protocol allows for improved in vitro modeling of cortical development while providing a platform that can be utilized to uncover disease-specific mechanisms of disordered development at various stages across the differentiation timeline. © 2023 Wiley Periodicals LLC. Basic Protocol 1: 3D hPSC neural induction Support Protocol 1: Neural rosette formation assay Support Protocol 2: Neurosphere generation Support Protocol 3: Enzymatic dissociation, NSC expansion, and cryopreservation Basic Protocol 2: 3D neural progenitor expansion Basic Protocol 3: 3D accelerated cortical lineage patterning and terminal differentiation.
Collapse
Affiliation(s)
- Dosh Whye
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| | - Delaney Wood
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| | - Wardiya Afshar Saber
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
- Department of Neurology, Harvard Medical School, Boston, MA
| | - Erika M. Norabuena
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| | - Nina R. Makhortova
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
- Department of Neurology, Harvard Medical School, Boston, MA
| | - Mustafa Sahin
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
- Department of Neurology, Harvard Medical School, Boston, MA
| | - Elizabeth D. Buttermore
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| |
Collapse
|
17
|
Pré D, Wooten AT, Zhou H, Neil A, Bang AG. Assaying Chemical Long-Term Potentiation in Human iPSC-Derived Neuronal Networks. Methods Mol Biol 2023; 2683:275-289. [PMID: 37300783 DOI: 10.1007/978-1-0716-3287-1_22] [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: 06/12/2023]
Abstract
Impairment of long-term potentiation (LTP) is a common feature of many preclinical models of neurological disorders. Modeling LTP on human induced pluripotent stem cells (hiPSC) enables the investigation of this critical plasticity process in disease-specific genetic backgrounds. Here, we describe a method to chemically induce LTP across entire networks of hiPSC-derived neurons on multi-electrode arrays (MEAs) and investigate effects on neuronal network activity and associated molecular changes.
Collapse
Affiliation(s)
- Deborah Pré
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Alexander T Wooten
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Haowen Zhou
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Ashley Neil
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Anne G Bang
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
18
|
Han X, Matsuda N, Ishibashi Y, Odawara A, Takahashi S, Tooi N, Kinoshita K, Suzuki I. A functional neuron maturation device provides convenient application on microelectrode array for neural network measurement. Biomater Res 2022; 26:84. [PMID: 36539898 PMCID: PMC9768978 DOI: 10.1186/s40824-022-00324-z] [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: 08/12/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Microelectrode array (MEA) systems are valuable for in vitro assessment of neurotoxicity and drug efficiency. However, several difficulties such as protracted functional maturation and high experimental costs hinder the use of MEA analysis requiring human induced pluripotent stem cells (hiPSCs). Neural network functional parameters are also needed for in vitro to in vivo extrapolation. METHODS In the present study, we produced a cost effective nanofiber culture platform, the SCAD device, for long-term culture of hiPSC-derived neurons and primary peripheral neurons. The notable advantage of SCAD device is convenient application on multiple MEA systems for neuron functional analysis. RESULTS We showed that the SCAD device could promote functional maturation of cultured hiPSC-derived neurons, and neurons responded appropriately to convulsant agents. Furthermore, we successfully analyzed parameters for in vitro to in vivo extrapolation, i.e., low-frequency components and synaptic propagation velocity of the signal, potentially reflecting neural network functions from neurons cultured on SCAD device. Finally, we measured the axonal conduction velocity of peripheral neurons. CONCLUSIONS Neurons cultured on SCAD devices might constitute a reliable in vitro platform to investigate neuron functions, drug efficacy and toxicity, and neuropathological mechanisms by MEA.
Collapse
Affiliation(s)
- Xiaobo Han
- grid.444756.00000 0001 2165 0596Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-Ku, Sendai, Miyagi 982-8577 Japan
| | - Naoki Matsuda
- grid.444756.00000 0001 2165 0596Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-Ku, Sendai, Miyagi 982-8577 Japan
| | - Yuto Ishibashi
- grid.444756.00000 0001 2165 0596Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-Ku, Sendai, Miyagi 982-8577 Japan
| | - Aoi Odawara
- grid.444756.00000 0001 2165 0596Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-Ku, Sendai, Miyagi 982-8577 Japan
| | - Sayuri Takahashi
- grid.444756.00000 0001 2165 0596Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-Ku, Sendai, Miyagi 982-8577 Japan
| | - Norie Tooi
- Stem Cell & Device Laboratory, Inc. (SCAD), OFFICE-ONE Shijo Karasuma 11F, 480, Niwatoriboko-Cho, Shimogyo-Ku, Kyoto, 600-8491 Japan
| | - Koshi Kinoshita
- Stem Cell & Device Laboratory, Inc. (SCAD), OFFICE-ONE Shijo Karasuma 11F, 480, Niwatoriboko-Cho, Shimogyo-Ku, Kyoto, 600-8491 Japan
| | - Ikuro Suzuki
- grid.444756.00000 0001 2165 0596Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-Ku, Sendai, Miyagi 982-8577 Japan
| |
Collapse
|
19
|
Ishibashi Y, Kimura S, Suzuki I. Responses to antibiotics in human iPSC-derived neurons based on the clinical antibiotic-associated encephalopathy classification. J Toxicol Sci 2022; 47:429-437. [PMID: 36184562 DOI: 10.2131/jts.47.429] [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: 11/02/2022]
Abstract
Antibiotic-associated encephalopathy (AAE) is a central nervous system disorder caused by antibiotics administration and classified into three types based on clinical symptoms. Type 1 AAE causes seizures and myoclonus, type 2 causes psychiatric symptoms, and type 3 is characterized by cerebellar ataxia. In this study, we investigated whether the electrical activity of in vitro human iPSC-derived neurons to antibiotics could be classified based on the 3 types of AAEs classified by clinical symptoms. Glutamatergic, GABAergic neurons and astrocytes differentiated from human iPS cells were seeded on micro-electrode array (MEA). The cumulative administration of 13 different antimicrobials detected changes in neural activity that differed according to AAE type. Next, we classified the antimicrobials by principal component analysis (PCA) and confirmed the AAE type of each agent. We found that Types 1-3 AAE agents were distributed separately. The classification of antibiotics depending on electrophysiological response characteristics was consistent with the clinical practice classification of AAEs. In conclusion, the combination of electrophysiological responses of human iPS cell-derived neural networks measured by MEA plus multivariate analysis methods will effectively detect and classify antibiotics developmental risks.
Collapse
Affiliation(s)
- Yuto Ishibashi
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology
| | - Shingo Kimura
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology
| | - Ikuro Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology
| |
Collapse
|
20
|
Habibey R, Striebel J, Schmieder F, Czarske J, Busskamp V. Long-term morphological and functional dynamics of human stem cell-derived neuronal networks on high-density micro-electrode arrays. Front Neurosci 2022; 16:951964. [PMID: 36267241 PMCID: PMC9578684 DOI: 10.3389/fnins.2022.951964] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Comprehensive electrophysiological characterizations of human induced pluripotent stem cell (hiPSC)-derived neuronal networks are essential to determine to what extent these in vitro models recapitulate the functional features of in vivo neuronal circuits. High-density micro-electrode arrays (HD-MEAs) offer non-invasive recording with the best spatial and temporal resolution possible to date. For 3 months, we tracked the morphology and activity features of developing networks derived from a transgenic hiPSC line in which neurogenesis is inducible by neurogenic transcription factor overexpression. Our morphological data revealed large-scale structural changes from homogeneously distributed neurons in the first month to the formation of neuronal clusters over time. This led to a constant shift in position of neuronal cells and clusters on HD-MEAs and corresponding changes in spatial distribution of the network activity maps. Network activity appeared as scarce action potentials (APs), evolved as local bursts with longer duration and changed to network-wide synchronized bursts with higher frequencies but shorter duration over time, resembling the emerging burst features found in the developing human brain. Instantaneous firing rate data indicated that the fraction of fast spiking neurons (150–600 Hz) increases sharply after 63 days post induction (dpi). Inhibition of glutamatergic synapses erased burst features from network activity profiles and confirmed the presence of mature excitatory neurotransmission. The application of GABAergic receptor antagonists profoundly changed the bursting profile of the network at 120 dpi. This indicated a GABAergic switch from excitatory to inhibitory neurotransmission during circuit development and maturation. Our results suggested that an emerging GABAergic system at older culture ages is involved in regulating spontaneous network bursts. In conclusion, our data showed that long-term and continuous microscopy and electrophysiology readouts are crucial for a meaningful characterization of morphological and functional maturation in stem cell-derived human networks. Most importantly, assessing the level and duration of functional maturation is key to subject these human neuronal circuits on HD-MEAs for basic and biomedical applications.
Collapse
Affiliation(s)
- Rouhollah Habibey
- Department of Ophthalmology, Universitäts-Augenklinik Bonn, University of Bonn, Bonn, Germany
| | - Johannes Striebel
- Department of Ophthalmology, Universitäts-Augenklinik Bonn, University of Bonn, Bonn, Germany
| | - Felix Schmieder
- Laboratory of Measurement and Sensor System Technique, Faculty of Electrical and Computer Engineering, TU Dresden, Dresden, Germany
| | - Jürgen Czarske
- Laboratory of Measurement and Sensor System Technique, Faculty of Electrical and Computer Engineering, TU Dresden, Dresden, Germany
- Competence Center for Biomedical Computational Laser Systems (BIOLAS), TU Dresden, Dresden, Germany
- Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany
- School of Science, Institute of Applied Physics, TU Dresden, Dresden, Germany
| | - Volker Busskamp
- Department of Ophthalmology, Universitäts-Augenklinik Bonn, University of Bonn, Bonn, Germany
- *Correspondence: Volker Busskamp,
| |
Collapse
|
21
|
Habibey R, Rojo Arias JE, Striebel J, Busskamp V. Microfluidics for Neuronal Cell and Circuit Engineering. Chem Rev 2022; 122:14842-14880. [PMID: 36070858 PMCID: PMC9523714 DOI: 10.1021/acs.chemrev.2c00212] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Indexed: 02/07/2023]
Abstract
The widespread adoption of microfluidic devices among the neuroscience and neurobiology communities has enabled addressing a broad range of questions at the molecular, cellular, circuit, and system levels. Here, we review biomedical engineering approaches that harness the power of microfluidics for bottom-up generation of neuronal cell types and for the assembly and analysis of neural circuits. Microfluidics-based approaches are instrumental to generate the knowledge necessary for the derivation of diverse neuronal cell types from human pluripotent stem cells, as they enable the isolation and subsequent examination of individual neurons of interest. Moreover, microfluidic devices allow to engineer neural circuits with specific orientations and directionality by providing control over neuronal cell polarity and permitting the isolation of axons in individual microchannels. Similarly, the use of microfluidic chips enables the construction not only of 2D but also of 3D brain, retinal, and peripheral nervous system model circuits. Such brain-on-a-chip and organoid-on-a-chip technologies are promising platforms for studying these organs as they closely recapitulate some aspects of in vivo biological processes. Microfluidic 3D neuronal models, together with 2D in vitro systems, are widely used in many applications ranging from drug development and toxicology studies to neurological disease modeling and personalized medicine. Altogether, microfluidics provide researchers with powerful systems that complement and partially replace animal models.
Collapse
Affiliation(s)
- Rouhollah Habibey
- Department
of Ophthalmology, Universitäts-Augenklinik
Bonn, University of Bonn, Ernst-Abbe-Straße 2, D-53127 Bonn, Germany
| | - Jesús Eduardo Rojo Arias
- Wellcome—MRC
Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge
Biomedical Campus, University of Cambridge, Cambridge CB2 0AW, United Kingdom
| | - Johannes Striebel
- Department
of Ophthalmology, Universitäts-Augenklinik
Bonn, University of Bonn, Ernst-Abbe-Straße 2, D-53127 Bonn, Germany
| | - Volker Busskamp
- Department
of Ophthalmology, Universitäts-Augenklinik
Bonn, University of Bonn, Ernst-Abbe-Straße 2, D-53127 Bonn, Germany
| |
Collapse
|
22
|
Elder N, Fattahi F, McDevitt TC, Zholudeva LV. Diseased, differentiated and difficult: Strategies for improved engineering of in vitro neurological systems. Front Cell Neurosci 2022; 16:962103. [PMID: 36238834 PMCID: PMC9550918 DOI: 10.3389/fncel.2022.962103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/22/2022] [Indexed: 12/01/2022] Open
Abstract
The rapidly growing field of cellular engineering is enabling scientists to more effectively create in vitro models of disease and develop specific cell types that can be used to repair damaged tissue. In particular, the engineering of neurons and other components of the nervous system is at the forefront of this field. The methods used to engineer neural cells can be largely divided into systems that undergo directed differentiation through exogenous stimulation (i.e., via small molecules, arguably following developmental pathways) and those that undergo induced differentiation via protein overexpression (i.e., genetically induced and activated; arguably bypassing developmental pathways). Here, we highlight the differences between directed differentiation and induced differentiation strategies, how they can complement one another to generate specific cell phenotypes, and impacts of each strategy on downstream applications. Continued research in this nascent field will lead to the development of improved models of neurological circuits and novel treatments for those living with neurological injury and disease.
Collapse
Affiliation(s)
- Nicholas Elder
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, United States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- Gladstone Institutes, San Francisco, CA, United States
| | - Faranak Fattahi
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, United States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
| | - Todd C. McDevitt
- Gladstone Institutes, San Francisco, CA, United States
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, United States
- Sana Biotechnology, Inc., South San Francisco, CA, United States
| | - Lyandysha V. Zholudeva
- Gladstone Institutes, San Francisco, CA, United States
- *Correspondence: Lyandysha V. Zholudeva,
| |
Collapse
|
23
|
Pré D, Wooten AT, Biesmans S, Hinckley S, Zhou H, Sherman SP, Kakad P, Gearhart J, Bang AG. Development of a platform to investigate long-term potentiation in human iPSC-derived neuronal networks. Stem Cell Reports 2022; 17:2141-2155. [PMID: 35985330 PMCID: PMC9481914 DOI: 10.1016/j.stemcr.2022.07.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 01/25/2023] Open
Abstract
Impairment of long-term potentiation (LTP) is a common feature of many pre-clinical models of neurological disorders; however, studies in humans are limited by the inaccessibility of the brain. Human induced pluripotent stem cells (hiPSCs) provide a unique opportunity to study LTP in disease-specific genetic backgrounds. Here we describe a multi-electrode array (MEA)-based assay to investigate chemically induced LTP (cLTP) across entire networks of hiPSC-derived midbrain dopaminergic (DA) and cortical neuronal populations that lasts for days, allowing studies of the late phases of LTP and enabling detection of associated molecular changes. We show that cLTP on midbrain DA neuronal networks is largely independent of the N-methyl-D-aspartate receptor (NMDAR) and partially dependent on brain-derived neurotrophic factor (BDNF). Finally, we describe activity-regulated gene expression induced by cLTP. This cLTP-MEA assay platform will enable phenotype discovery and higher-throughput analyses of synaptic plasticity on hiPSC-derived neurons.
Collapse
Affiliation(s)
- Deborah Pré
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Alexander T Wooten
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Steven Biesmans
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Sandy Hinckley
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Haowen Zhou
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Sean P Sherman
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Priyanka Kakad
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jeffrey Gearhart
- Henry M. Jackson Foundation for the Advancement of Military Medicine on Contract to USAF School of Aerospace Medicine, Wright-Patterson AFB, Dayton, OH 45433, USA
| | - Anne G Bang
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
24
|
Kreir M, Floren W, Policarpo R, De Bondt A, Van den Wyngaert I, Teisman A, Gallacher DJ, Lu HR. Is the forming of neuronal network activity in human-induced pluripotent stem cells important for the detection of drug-induced seizure risks? Eur J Pharmacol 2022; 931:175189. [PMID: 35987255 DOI: 10.1016/j.ejphar.2022.175189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/03/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Functional network activity is a characteristic for neuronal cells, and the complexity of the network activity represents the necessary substrate to support complex brain functions. Drugs that drastically increase the neuronal network activity may have a potential higher risk for seizures in human. Although there has been some recent considerable progress made using cultures from different types of human-induced pluripotent stem cell (hiPSC) derived neurons, one of the primary limitations is the lack of - or very low - network activity. METHOD In the present study, we investigated whether the limited neuronal network activity in commercial hiPSC-neurons (CNS.4U®) is capable of detecting drug-induced potential seizure risks. Therefore, we compared the hiPSC-results to those in rat primary neurons with known high neuronal network activity in vitro. RESULTS Gene expression and electrical activity from in vitro developing neuronal networks were assessed at multiple time-points. Transcriptomes of 7, 28, and 50 days in vitro were analyzed and compared to those from human brain tissues. Data from measurements of electrical activity using multielectrode arrays (MEAs) indicate that neuronal networks matured gradually over time, albeit in hiPSC this developed slower than rat primary cultures. The response of neuronal networks to neuronal active reference drugs modulating glutamatergic, acetylcholinergic and GABAergic pathways could be detected in both hiPSC-neurons and rat primary neurons. However, in comparison, GABAergic responses were limited in hiPSC-neurons. CONCLUSION Overall, despite a slower network development and lower network activity, CNS.4U® hiPSC-neurons can be used to detect drug induced changes in neuronal network activity, as shown by well-known seizurogenic drugs (affecting e.g., the Glycine receptor and Na+ channel). However, lower sensitivity to GABA antagonists has been observed.
Collapse
Affiliation(s)
- Mohamed Kreir
- Global Safety Pharmacology, Predictive & Investigative Translational Toxicology, Nonclinical Safety, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium.
| | - Wim Floren
- Global Safety Pharmacology, Predictive & Investigative Translational Toxicology, Nonclinical Safety, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Rafaela Policarpo
- Neuroscience Therapeutic Area, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Belgium
| | - An De Bondt
- High Dimensional & Computational Biology, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Ilse Van den Wyngaert
- High Dimensional & Computational Biology, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Ard Teisman
- Global Safety Pharmacology, Predictive & Investigative Translational Toxicology, Nonclinical Safety, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - David J Gallacher
- Global Safety Pharmacology, Predictive & Investigative Translational Toxicology, Nonclinical Safety, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Hua Rong Lu
- Global Safety Pharmacology, Predictive & Investigative Translational Toxicology, Nonclinical Safety, Janssen Research and Development, A Division of Janssen Pharmaceutica NV, Beerse, Belgium
| |
Collapse
|
25
|
Kang KR, Kim CY, Kim J, Ryu B, Lee SG, Baek J, Kim YJ, Lee JM, Lee Y, Choi SO, Woo DH, Park IH, Chung HM. Establishment of Neurotoxicity Assessment Using Microelectrode Array (MEA) with hiPSC-Derived Neurons and Evaluation of New Psychoactive Substances (NPS). Int J Stem Cells 2022; 15:258-269. [PMID: 35769054 PMCID: PMC9396014 DOI: 10.15283/ijsc21217] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 04/01/2022] [Accepted: 04/01/2022] [Indexed: 11/09/2022] Open
Abstract
Background and Objectives Currently, safety pharmacological tests for the central nervous system depend on animal behavioral analysis. However, due to the subjectivity of behavioral analysis and differences between species, there is a limit to appropriate nervous system toxicity assessment, therefore a new neurotoxicity assessment that can simulate the human central nervous system is required. Methods and Results In our study, we developed an in vitro neurotoxicity assessment focusing on neuronal function. To minimize the differences between species and fast screening, hiPSC-derived neurons and a microelectrode array (MEA) that could simultaneously measure the action potentials of the neuronal networks were used. After analyzing the molecular and electrophysiological characters of our neuronal network, we conducted a neurotoxicity assessment on neurotransmitters, neurotoxicants, illicit drugs, and new psychoactive substances (NPS). We found that most substances used in our experiments responded more sensitively to our MEA-based neurotoxicity assessment than to the conventional neurotoxicity assessment. Also, this is the first paper that evaluates various illicit drugs and NPS using MEA-based neurotoxicity assessment using hiPSC-derived neurons. Conclusions Our study expanded the scope of application of neurotoxicity assessment using hiPSC-derived neurons to NPS, and accumulated evaluation data of various toxic substances for hiPSC-derived neurons.
Collapse
Affiliation(s)
- Kyu-Ree Kang
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - C-Yoon Kim
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea.,Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Korea
| | - Jin Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Bokyeong Ryu
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Seul-Gi Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Jieun Baek
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Ye-Ji Kim
- Drug Abuse Research Group, Research Center of Convergence Toxicology, Korea Institute of Toxicology, Daejeon, Korea
| | - Jin-Moo Lee
- Pharmacological Research Division, Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Korea
| | - Yootmo Lee
- Pharmacological Research Division, Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Korea
| | - Sun-Ok Choi
- Pharmacological Research Division, Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Korea
| | - Dong Ho Woo
- Drug Abuse Research Group, Research Center of Convergence Toxicology, Korea Institute of Toxicology, Daejeon, Korea
| | - Il Hwan Park
- Departments of Thoracis and Cardiovascular Surgery, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| |
Collapse
|
26
|
Van Breedam E, Ponsaerts P. Promising Strategies for the Development of Advanced In Vitro Models with High Predictive Power in Ischaemic Stroke Research. Int J Mol Sci 2022; 23:ijms23137140. [PMID: 35806146 PMCID: PMC9266337 DOI: 10.3390/ijms23137140] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022] Open
Abstract
Although stroke is one of the world’s leading causes of death and disability, and more than a thousand candidate neuroprotective drugs have been proposed based on extensive in vitro and animal-based research, an effective neuroprotective/restorative therapy for ischaemic stroke patients is still missing. In particular, the high attrition rate of neuroprotective compounds in clinical studies should make us question the ability of in vitro models currently used for ischaemic stroke research to recapitulate human ischaemic responses with sufficient fidelity. The ischaemic stroke field would greatly benefit from the implementation of more complex in vitro models with improved physiological relevance, next to traditional in vitro and in vivo models in preclinical studies, to more accurately predict clinical outcomes. In this review, we discuss current in vitro models used in ischaemic stroke research and describe the main factors determining the predictive value of in vitro models for modelling human ischaemic stroke. In light of this, human-based 3D models consisting of multiple cell types, either with or without the use of microfluidics technology, may better recapitulate human ischaemic responses and possess the potential to bridge the translational gap between animal-based in vitro and in vivo models, and human patients in clinical trials.
Collapse
|
27
|
Zhang W, Ross PJ, Ellis J, Salter MW. Targeting NMDA receptors in neuropsychiatric disorders by drug screening on human neurons derived from pluripotent stem cells. Transl Psychiatry 2022; 12:243. [PMID: 35680847 PMCID: PMC9184461 DOI: 10.1038/s41398-022-02010-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 01/04/2023] Open
Abstract
NMDA receptors (NMDARs), a prominent subtype of glutamatergic receptors, are implicated in the pathogenesis and development of neuropsychiatric disorders such as epilepsy, intellectual disability, autism spectrum disorder, and schizophrenia, and are therefore a potential therapeutic target in treating these disorders. Neurons derived from induced pluripotent stem cells (iPSCs) have provided the opportunity to investigate human NMDARs in their native environment. In this review, we describe the expression, function, and regulation of NMDARs in human iPSC-derived neurons and discuss approaches for utilizing human neurons for identifying potential drugs that target NMDARs in the treatment of neuropsychiatric disorders. A challenge in studying NMDARs in human iPSC-derived neurons is a predominance of those receptors containing the GluN2B subunit and low synaptic expression, suggesting a relatively immature phenotype of these neurons and delayed development of functional NMDARs. We outline potential approaches for improving neuronal maturation of human iPSC-derived neurons and accelerating the functional expression of NMDARs. Acceleration of functional expression of NMDARs in human iPSC-derived neurons will improve the modeling of neuropsychiatric disorders and facilitate the discovery and development of novel therapeutics targeting NMDARs for the treatment of these disorders.
Collapse
Affiliation(s)
- Wenbo Zhang
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - P Joel Ross
- Biology Department, University of Prince Edward Island, Charlottetown, PE, C1A 4P3, Canada
| | - James Ellis
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, M5S 1A8, Canada
| | - Michael W Salter
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
28
|
Schmieder F, Habibey R, Striebel J, Büttner L, Czarske J, Busskamp V. Tracking connectivity maps in human stem cell-derived neuronal networks by holographic optogenetics. Life Sci Alliance 2022; 5:5/7/e202101268. [PMID: 35418473 PMCID: PMC9008225 DOI: 10.26508/lsa.202101268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 11/24/2022] Open
Abstract
Holographic optogenetic stimulation of human iPSC–derived neuronal networks was exploited to map precise functional connectivity motifs and their long-term dynamics during network development. Neuronal networks derived from human induced pluripotent stem cells have been exploited widely for modeling neuronal circuits, neurological diseases, and drug screening. As these networks require extended culturing periods to functionally mature in vitro, most studies are based on immature networks. To obtain insights on long-term functional features, we improved a glia–neuron co-culture protocol within multi-electrode arrays, facilitating continuous assessment of electrical features in weekly intervals. By full-field optogenetic stimulation, we detected an earlier onset of neuronal firing and burst activity compared with spontaneous activity. Full-field stimulation enhanced the number of active neurons and their firing rates. Compared with full-field stimulation, which evoked synchronized activity across all neurons, holographic stimulation of individual neurons resulted in local activity. Single-cell holographic stimulation facilitated to trace propagating evoked activities of 400 individually stimulated neurons per multi-electrode array. Thereby, we revealed precise functional neuronal connectivity motifs. Holographic stimulation data over time showed increasing connection numbers and strength with culture age. This holographic stimulation setup has the potential to establish a profound functional testbed for in-depth analysis of human-induced pluripotent stem cell-derived neuronal networks.
Collapse
Affiliation(s)
- Felix Schmieder
- Laboratory of Measurement and Sensor System Technique, Faculty of Electrical and Computer Engineering, TU Dresden, Dresden, Germany
| | - Rouhollah Habibey
- Department of Ophthalmology, Universitäts-Augenklinik Bonn, University of Bonn, Bonn, Germany
| | - Johannes Striebel
- Department of Ophthalmology, Universitäts-Augenklinik Bonn, University of Bonn, Bonn, Germany
| | - Lars Büttner
- Laboratory of Measurement and Sensor System Technique, Faculty of Electrical and Computer Engineering, TU Dresden, Dresden, Germany
| | - Jürgen Czarske
- Laboratory of Measurement and Sensor System Technique, Faculty of Electrical and Computer Engineering, TU Dresden, Dresden, Germany .,Competence Center for Biomedical Computational Laser Systems (BIOLAS), TU Dresden, Dresden, Germany.,Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany.,Institute of Applied Physics, School of Science, TU Dresden, Dresden, Germany
| | - Volker Busskamp
- Department of Ophthalmology, Universitäts-Augenklinik Bonn, University of Bonn, Bonn, Germany
| |
Collapse
|
29
|
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
|
30
|
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
|
31
|
Yokoi R, Shigemoto-Kuroda T, Matsuda N, Odawara A, Suzuki I. Electrophysiological responses to seizurogenic compounds dependent on E/I balance in human iPSC-derived cortical neural networks. J Pharmacol Sci 2022; 148:267-278. [DOI: 10.1016/j.jphs.2021.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/18/2021] [Accepted: 12/27/2021] [Indexed: 10/19/2022] Open
|
32
|
Hong J, Dragas R, Khazaei M, Ahuja CS, Fehlings MG. Hepatocyte Growth Factor-Preconditioned Neural Progenitor Cells Attenuate Astrocyte Reactivity and Promote Neurite Outgrowth. Front Cell Neurosci 2021; 15:741681. [PMID: 34955750 PMCID: PMC8695970 DOI: 10.3389/fncel.2021.741681] [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: 07/15/2021] [Accepted: 11/09/2021] [Indexed: 11/13/2022] Open
Abstract
The astroglial scar is a defining hallmark of secondary pathology following central nervous system (CNS) injury that, despite its role in limiting tissue damage, presents a significant barrier to neuroregeneration. Neural progenitor cell (NPC) therapies for tissue repair and regeneration have demonstrated favorable outcomes, the effects of which are ascribed not only to direct cell replacement but trophic support. Cytokines and growth factors secreted by NPCs aid in modifying the inhibitory and cytotoxic post-injury microenvironment. In an effort to harness and enhance the reparative potential of NPC secretome, we utilized the multifunctional and pro-regenerative cytokine, hepatocyte growth factor (HGF), as a cellular preconditioning agent. We first demonstrated the capacity of HGF to promote NPC survival in the presence of oxidative stress. We then assessed the capacity of this modified conditioned media (CM) to attenuate astrocyte reactivity and promote neurite outgrowth in vitro. HGF pre-conditioned NPCs demonstrated significantly increased levels of tissue inhibitor of metalloproteinases-1 and reduced vascular endothelial growth factor compared to untreated NPCs. In reactive astrocytes, HGF-enhanced NPC-CM effectively reduced glial fibrillary acidic protein (GFAP) expression and chondroitin sulfate proteoglycan deposition to a greater extent than either treatment alone, and enhanced neurite outgrowth of co-cultured neurons. in vivo, this combinatorial treatment strategy might enable tactical modification of the post-injury inhibitory astroglial environment to one that is more conducive to regeneration and functional recovery. These findings have important translational implications for the optimization of current cell-based therapies for CNS injury.
Collapse
Affiliation(s)
- James Hong
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Rachel Dragas
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Mohammad Khazaei
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Christopher S Ahuja
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Michael G Fehlings
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Spinal Program, University Health Network, Toronto Western Hospital, Toronto, ON, Canada
| |
Collapse
|
33
|
Knock E, Julian LM. Building on a Solid Foundation: Adding Relevance and Reproducibility to Neurological Modeling Using Human Pluripotent Stem Cells. Front Cell Neurosci 2021; 15:767457. [PMID: 34867204 PMCID: PMC8637745 DOI: 10.3389/fncel.2021.767457] [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: 08/30/2021] [Accepted: 10/20/2021] [Indexed: 11/25/2022] Open
Abstract
The brain is our most complex and least understood organ. Animal models have long been the most versatile tools available to dissect brain form and function; however, the human brain is highly distinct from that of standard model organisms. In addition to existing models, access to human brain cells and tissues is essential to reach new frontiers in our understanding of the human brain and how to intervene therapeutically in the face of disease or injury. In this review, we discuss current and developing culture models of human neural tissue, outlining advantages over animal models and key challenges that remain to be overcome. Our principal focus is on advances in engineering neural cells and tissue constructs from human pluripotent stem cells (PSCs), though primary human cell and slice culture are also discussed. By highlighting studies that combine animal models and human neural cell culture techniques, we endeavor to demonstrate that clever use of these orthogonal model systems produces more reproducible, physiological, and clinically relevant data than either approach alone. We provide examples across a range of topics in neuroscience research including brain development, injury, and cancer, neurodegenerative diseases, and psychiatric conditions. Finally, as testing of PSC-derived neurons for cell replacement therapy progresses, we touch on the advancements that are needed to make this a clinical mainstay.
Collapse
Affiliation(s)
- Erin Knock
- Research and Development, STEMCELL Technologies Inc., Vancouver, BC, Canada.,Department of Biological Sciences, Faculty of Science, Simon Fraser University, Burnaby, BC, Canada
| | - Lisa M Julian
- Department of Biological Sciences, Faculty of Science, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
34
|
Electrophysiological- and Neuropharmacological-Based Benchmarking of Human Induced Pluripotent Stem Cell-Derived and Primary Rodent Neurons. Stem Cell Rev Rep 2021; 18:259-277. [PMID: 34687385 DOI: 10.1007/s12015-021-10263-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2021] [Indexed: 12/15/2022]
Abstract
Human induced pluripotent stem cell (iPSC)-derived neurons are of interest for studying neurological disease mechanisms, developing potential therapies and deepening our understanding of the human nervous system. However, compared to an extensive history of practice with primary rodent neuron cultures, human iPSC-neurons still require more robust characterization of expression of neuronal receptors and ion channels and functional and predictive pharmacological responses. In this study, we differentiated human amniotic fluid-derived iPSCs into a mixed population of neurons (AF-iNs). Functional assessments were performed by evaluating electrophysiological (patch-clamp) properties and the effect of a panel of neuropharmacological agents on spontaneous activity (multi-electrode arrays; MEAs). These electrophysiological data were benchmarked relative to commercially sourced human iPSC-derived neurons (CNS.4U from Ncardia), primary human neurons (ScienCell™) and primary rodent cortical/hippocampal neurons. Patch-clamp whole-cell recordings showed that mature AF-iNs generated repetitive firing of action potentials in response to depolarizations, similar to that of primary rodent cortical/hippocampal neurons, with nearly half of the neurons displaying spontaneous post-synaptic currents. Immunochemical and MEA-based analyses indicated that AF-iNs were composed of functional glutamatergic excitatory and inhibitory GABAergic neurons. Principal component analysis of MEA data indicated that human AF-iN and rat neurons exhibited distinct pharmacological and electrophysiological properties. Collectively, this study establishes a necessary prerequisite for AF-iNs as a human neuron culture model suitable for pharmacological studies.
Collapse
|
35
|
Ishibashi Y, Odawara A, Kinoshita K, Okamura A, Shirakawa T, Suzuki I. Principal Component Analysis to Distinguish Seizure Liability of Drugs in Human iPS Cell-Derived Neurons. Toxicol Sci 2021; 184:265-275. [PMID: 34570236 DOI: 10.1093/toxsci/kfab116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Screening for drug discovery targeting the central nervous system requires the establishment of efficient and highly accurate toxicity test methods that can reduce costs and time while maintaining high throughput using the function of an in vitro neural network. In particular, an evaluation system using a human-derived neural network is desirable in terms of species difference. Despite the attention the microelectrode array (MEA) is attracting among the evaluation systems that can measure in vitro neural activity, an effective analysis method for evaluation of toxicity and mechanism of action has not yet been established. Here we established analytical parameters and multivariate analysis method capable of detecting seizure liability of drugs using MEA measurement of human iPS cell-derived neurons. Using the spike time series data of all drugs, we established periodicity as a new analytical parameter. Periodicity has facilitated the detection of responses to seizurogenic drugs, previously difficult to detect with conventional analytical parameters. By constructing a multivariate analytical method that identifies a parameter set that achieves an arbitrary condition, we found that the parameter set comprising total spikes, maximum frequency, inter maximum frequency interval, coefficient of variance of inter maximum frequency interval, and periodicity can uniformly detect the seizure liability of seizurogenic drugs with different mechanisms of action. Seizurogenic drugs were suggested to increase the regularity of the network burst in MEA measurements in human iPS cell-derived neurons.
Collapse
Affiliation(s)
- Y Ishibashi
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - 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, 305-0841, Japan
| | - A Okamura
- Drug Safety Research Labs, Astellas Pharma Inc, 21 Miyukigaoka, Tsukuba, Ibaraki, 305-0841, Japan
| | - T Shirakawa
- Drug Safety Research Labs, Astellas Pharma Inc, 21 Miyukigaoka, Tsukuba, Ibaraki, 305-0841, 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
|
36
|
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
|
37
|
Halliwell RF, Salmanzadeh H, Coyne L, Cao WS. An Electrophysiological and Pharmacological Study of the Properties of Human iPSC-Derived Neurons for Drug Discovery. Cells 2021; 10:cells10081953. [PMID: 34440722 PMCID: PMC8395001 DOI: 10.3390/cells10081953] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 01/01/2023] Open
Abstract
Human stem cell-derived neurons are increasingly considered powerful models in drug discovery and disease modeling, despite limited characterization of their molecular properties. Here, we have conducted a detailed study of the properties of a commercial human induced Pluripotent Stem Cell (iPSC)-derived neuron line, iCell [GABA] neurons, maintained for up to 3 months in vitro. We confirmed that iCell neurons display neurite outgrowth within 24 h of plating and label for the pan-neuronal marker, βIII tubulin within the first week. Our multi-electrode array (MEA) recordings clearly showed neurons generated spontaneous, spike-like activity within 2 days of plating, which peaked at one week, and rapidly decreased over the second week to remain at low levels up to one month. Extracellularly recorded spikes were reversibly inhibited by tetrodotoxin. Patch-clamp experiments showed that iCell neurons generated spontaneous action potentials and expressed voltage-gated Na and K channels with membrane capacitances, resistances and membrane potentials that are consistent with native neurons. Our single neuron recordings revealed that reduced spiking observed in the MEA after the first week results from development of a dominant inhibitory tone from GABAergic neuron circuit maturation. GABA evoked concentration-dependent currents that were inhibited by the convulsants, bicuculline and picrotoxin, and potentiated by the positive allosteric modulators, diazepam, chlordiazepoxide, phenobarbital, allopregnanolone and mefenamic acid, consistent with native neuronal GABAA receptors. We also show that glycine evoked robust concentration-dependent currents that were inhibited by the neurotoxin, strychnine. Glutamate, AMPA, Kainate and NMDA each evoked concentration-dependent currents in iCell neurons that were blocked by their selective antagonists, consistent with the expression of ionotropic glutamate receptors. The NMDA currents required the presence of the co-agonist glycine and were blocked in a highly voltage-dependent manner by Mg2+ consistent with the properties of native neuronal NMDA receptors. Together, our data suggest that such human iPSC-derived neurons may have significant value in drug discovery and development and may eventually largely replace the need for animal tissues in human biomedical research.
Collapse
|
38
|
Puppo F, Pré D, Bang AG, Silva GA. Super-Selective Reconstruction of Causal and Direct Connectivity With Application to in vitro iPSC Neuronal Networks. Front Neurosci 2021; 15:647877. [PMID: 34335152 PMCID: PMC8323822 DOI: 10.3389/fnins.2021.647877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 05/31/2021] [Indexed: 12/22/2022] Open
Abstract
Despite advancements in the development of cell-based in-vitro neuronal network models, the lack of appropriate computational tools limits their analyses. Methods aimed at deciphering the effective connections between neurons from extracellular spike recordings would increase utility of in vitro local neural circuits, especially for studies of human neural development and disease based on induced pluripotent stem cells (hiPSC). Current techniques allow statistical inference of functional couplings in the network but are fundamentally unable to correctly identify indirect and apparent connections between neurons, generating redundant maps with limited ability to model the causal dynamics of the network. In this paper, we describe a novel mathematically rigorous, model-free method to map effective-direct and causal-connectivity of neuronal networks from multi-electrode array data. The inference algorithm uses a combination of statistical and deterministic indicators which, first, enables identification of all existing functional links in the network and then reconstructs the directed and causal connection diagram via a super-selective rule enabling highly accurate classification of direct, indirect, and apparent links. Our method can be generally applied to the functional characterization of any in vitro neuronal networks. Here, we show that, given its accuracy, it can offer important insights into the functional development of in vitro hiPSC-derived neuronal cultures.
Collapse
Affiliation(s)
- Francesca Puppo
- BioCircuits Institute and Center for Engineered Natural Intelligence, University of California, San Diego, La Jolla, CA, United States
| | - Deborah Pré
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Anne G. Bang
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Gabriel A. Silva
- BioCircuits Institute, Center for Engineered Natural Intelligence, Department of Bioengineering, Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
39
|
Mossink B, Verboven AHA, van Hugte EJH, Klein Gunnewiek TM, Parodi G, Linda K, Schoenmaker C, Kleefstra T, Kozicz T, van Bokhoven H, Schubert D, Nadif Kasri N, Frega M. Human neuronal networks on micro-electrode arrays are a highly robust tool to study disease-specific genotype-phenotype correlations in vitro. Stem Cell Reports 2021; 16:2182-2196. [PMID: 34329594 PMCID: PMC8452490 DOI: 10.1016/j.stemcr.2021.07.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 01/16/2023] Open
Abstract
Micro-electrode arrays (MEAs) are increasingly used to characterize neuronal network activity of human induced pluripotent stem cell (hiPSC)-derived neurons. Despite their gain in popularity, MEA recordings from hiPSC-derived neuronal networks are not always used to their full potential in respect to experimental design, execution, and data analysis. Therefore, we benchmarked the robustness of MEA-derived neuronal activity patterns from ten healthy individual control lines, and uncover comparable network phenotypes. To achieve standardization, we provide recommendations on experimental design and analysis. With such standardization, MEAs can be used as a reliable platform to distinguish (disease-specific) network phenotypes. In conclusion, we show that MEAs are a powerful and robust tool to uncover functional neuronal network phenotypes from hiPSC-derived neuronal networks, and provide an important resource to advance the hiPSC field toward the use of MEAs for disease phenotyping and drug discovery. MEAs are a robust tool to model neuronal network functioning Neuronal networks from different healthy donors show comparable network activity MEAs are able to distinguish disease-specific neuronal network phenotypes We provide recommendations to standardize neuronal network recordings on MEA
Collapse
Affiliation(s)
- Britt Mossink
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, 6500 HB Nijmegen, the Netherlands; Department of Clinical Neurophysiology, University of Twente, 7522 NB Enschede, the Netherlands
| | - Anouk H A Verboven
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, 6500 HB Nijmegen, the Netherlands; Centre for Molecular and Biomolecular Informatics, Radboudumc, Radboud Institute for Molecular Life Sciences, 6500 HB Nijmegen, the Netherlands
| | - Eline J H van Hugte
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, 6500 HB Nijmegen, the Netherlands; ACE Kempenhaeghe, Department of Epileptology, 5591 VE Heeze, the Netherlands
| | - Teun M Klein Gunnewiek
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, 6500 HB Nijmegen, the Netherlands; Department of Medical Imaging, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Giulia Parodi
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, 6500 HB Nijmegen, the Netherlands
| | - Katrin Linda
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, 6500 HB Nijmegen, the Netherlands
| | - Chantal Schoenmaker
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, 6500 HB Nijmegen, the Netherlands
| | - Tjitske Kleefstra
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, 6500 HB Nijmegen, the Netherlands
| | - Tamas Kozicz
- Department of Medical Imaging, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Hans van Bokhoven
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, 6500 HB Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behavior, 6500 HB Nijmegen, the Netherlands
| | - Dirk Schubert
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, 6500 HB Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behavior, 6500 HB Nijmegen, the Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, 6500 HB Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behavior, 6500 HB Nijmegen, the Netherlands
| | - Monica Frega
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behavior, 6500 HB Nijmegen, the Netherlands; Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
40
|
Live Viral Vaccine Neurovirulence Screening: Current and Future Models. Vaccines (Basel) 2021; 9:vaccines9070710. [PMID: 34209433 PMCID: PMC8310194 DOI: 10.3390/vaccines9070710] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/12/2022] Open
Abstract
Live viral vaccines are one of the most successful methods for controlling viral infections but require strong evidence to indicate that they are properly attenuated. Screening for residual neurovirulence is an important aspect for live viral vaccines against potentially neurovirulent diseases. Approximately half of all emerging viral diseases have neurological effects, so testing of future vaccines will need to be rapid and accurate. The current method, the monkey neurovirulence test (MNVT), shows limited translatability for human diseases and does not account for different viral pathogenic mechanisms. This review discusses the MNVT and potential alternative models, including in vivo and in vitro methods. The advantages and disadvantages of these methods are discussed, and there are promising data indicating high levels of translatability. There is a need to investigate these models more thoroughly and to devise more accurate and rapid alternatives to the MNVT.
Collapse
|
41
|
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
|
42
|
Roberts R, Authier S, Mellon RD, Morton M, Suzuki I, Tjalkens RB, Valentin JP, Pierson JB. Can We Panelize Seizure? Toxicol Sci 2021; 179:3-13. [PMID: 33165543 DOI: 10.1093/toxsci/kfaa167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Seizure liability remains a significant cause of attrition in drug discovery and development, leading to loss of competitiveness, delays, and increased costs. Current detection methods rely on observations made in in vivo studies intended to support clinical trials, such as tremors or other abnormal movements. These signs could be missed or misinterpreted; thus, definitive confirmation of drug-induced seizure requires a follow-up electroencephalogram study. There has been progress in in vivo detection of seizure using automated video systems that record and analyze animal movements. Nonetheless, it would be preferable to have earlier prediction of seizurogenic risk that could be used to eliminate liabilities early in discovery while there are options for medicinal chemists making potential new drugs. Attrition due to cardiac adverse events has benefited from routine early screening; could we reduce attrition due to seizure using a similar approach? Specifically, microelectrode arrays could be used to detect potential seizurogenic signals in stem-cell-derived neurons. In addition, there is clear evidence implicating neuronal voltage-gated and ligand-gated ion channels, GPCRs and transporters in seizure. Interactions with surrounding glial cells during states of stress or inflammation can also modulate ion channel function in neurons, adding to the challenge of seizure prediction. It is timely to evaluate the opportunity to develop an in vitro assessment of seizure linked to a panel of ion channel assays that predict seizure, with the aim of influencing structure-activity relationship at the design stage and eliminating compounds predicted to be associated with pro-seizurogenic state.
Collapse
Affiliation(s)
- Ruth Roberts
- ApconiX, Alderley Park, SK10 4TG, UK.,University of Birmingham, B15 2SD, UK
| | | | - R Daniel Mellon
- US Food and Drug Administration, Silver Spring, Maryland 20993
| | | | - Ikuro Suzuki
- Tohoku Institute of Technology, Sendai, 980-8577, Japan
| | - Ronald B Tjalkens
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado 80523
| | - Jean-Pierre Valentin
- UCB Biopharma SRL, Early Solutions, Development Science, Investigative Toxicology, Chemin du Foriest, B-1420, Braine-l'Alleud, Belgium
| | - Jennifer B Pierson
- Health and Environmental Sciences Institute, Washington, District of Columbia 20005
| |
Collapse
|
43
|
Xie J, Wettschurack K, Yuan C. Review: In vitro Cell Platform for Understanding Developmental Toxicity. Front Genet 2020; 11:623117. [PMID: 33424939 PMCID: PMC7785584 DOI: 10.3389/fgene.2020.623117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/03/2020] [Indexed: 12/30/2022] Open
Abstract
Developmental toxicity and its affiliation to long-term health, particularly neurodegenerative disease (ND) has attracted significant attentions in recent years. There is, however, a significant gap in current models to track longitudinal changes arising from developmental toxicity. The advent of induced pluripotent stem cell (iPSC) derived neuronal culture has allowed for more complex and functionally active in vitro neuronal models. Coupled with recent progress in the detection of ND biomarkers, we are equipped with promising new tools to understand neurotoxicity arising from developmental exposure. This review provides a brief overview of current progress in neuronal culture derived from iPSC and in ND markers.
Collapse
Affiliation(s)
- Junkai Xie
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, United States
| | - Kyle Wettschurack
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, United States
| | - Chongli Yuan
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, United States
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
44
|
He J, Zhang N, Zhu Y, Jin R, Wu F. MSC spheroids-loaded collagen hydrogels simultaneously promote neuronal differentiation and suppress inflammatory reaction through PI3K-Akt signaling pathway. Biomaterials 2020; 265:120448. [PMID: 33068892 DOI: 10.1016/j.biomaterials.2020.120448] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/10/2020] [Indexed: 02/06/2023]
Abstract
It is critical for the clinical success to take the anti-inflammatory function into consideration when integrating the neurogenesis into the nerve repair materials. To this aim, we prepared mesenchymal stem cell (MSC) spheroids-loaded collagen (Col) hydrogels with combined superior anti-inflammatory efficacy and neurogenic activity. The size of the MSC spheroids showed a strong modulation effect on both functions, and the MSC spheroids-100 sample exhibited the best neuronal and anti-inflammatory potentials. The observed dual functions were likely based on the elevated intrinsic cell-cell contacts and cell-extracellular matrix interactions from the MSC spheroids. MSC self-assembly as spheroids expedited the secretions of endogenous trophic factors and extracellular matrix (ECM), which was beneficial to drive neural stem cell differentiation into the neuronal lineage. In addition, the formation of the MSC spheroids secreted more amounts and types of cytokines as well as immunomodulatory paracrine factors to suppress LPS-induced inflammatory reaction. LC-MS/MS analysis further demonstrated that MSC spheroids contributed to the activation of neuroactive ligand-receptor interaction, thereby triggering downstream PI3K-Akt signal pathway, which was likely due to the acceleration of ECM-receptor interaction, gap junction and tight junction. Importantly, inhibiting Akt pathway significantly suppressed the neuronal differentiation, indicating that PI3K-Akt signal pathway was critically involved in the Col-MSC spheroid hydrogel mediated neuroprotection and neurogenesis. Such findings not only provided a simple approach for improving MSC-based therapies for neuron-related diseases, but also shed insight on understanding the underlying mechanisms of MSC-mediated neuronal differentiation.
Collapse
Affiliation(s)
- Jing He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Nihui Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Yue Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China.
| | - Fang Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China.
| |
Collapse
|
45
|
Hirose S, Tanaka Y, Shibata M, Kimura Y, Ishikawa M, Higurashi N, Yamamoto T, Ichise E, Chiyonobu T, Ishii A. Application of induced pluripotent stem cells in epilepsy. Mol Cell Neurosci 2020; 108:103535. [DOI: 10.1016/j.mcn.2020.103535] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/10/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023] Open
|
46
|
Shirakawa T, Suzuki I. Approach to Neurotoxicity using Human iPSC Neurons: Consortium for Safety Assessment using Human iPS Cells. Curr Pharm Biotechnol 2020; 21:780-786. [DOI: 10.2174/1389201020666191129103730] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/27/2019] [Accepted: 11/03/2019] [Indexed: 01/05/2023]
Abstract
Neurotoxicity, as well as cardiotoxicity and hepatotoxicity, resulting from administration of
a test article is considered a major adverse effect both pre-clinically and clinically. Among the different
types of neurotoxicity occurring during the drug development process, seizure is one of the most serious
one. Seizure occurrence is usually assessed using in vivo animal models, the Functional Observational
Battery, the Irwin test or electroencephalograms. In in vitro studies, a number of assessments can
be performed using animal organs/cells. Interestingly, recent developments in stem cell biology, especially
the development of Human-Induced Pluripotent Stem (iPS) cells, are enabling the assessment of
neurotoxicity in human iPS cell-derived neurons. Further, a Multi-Electrode Array (MEA) using rodent
neurons is a useful tool for identifying seizure-inducing compounds. The Consortium for Safety Assessment
using Human iPS Cells (CSAHi; http://csahi.org/en/) was established in 2013 by the Japan
Pharmaceutical Manufacturers Association (JPMA) to verify the application of human iPS cell-derived
neuronal cells to drug safety evaluation. The Neuro Team of CSAHi has been attempting to evaluate the
seizure risk of compounds using the MEA platform. Here, we review the current status of neurotoxicity
and recent work, including problems related to the use of the MEA assay with human iPS neuronal
cell-derived neurons, and future developments.
Collapse
Affiliation(s)
- Takafumi Shirakawa
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Neuro Team, Japan
| | - Ikuro Suzuki
- Consortium for Safety Assessment using Human iPS Cells (CSAHi), Neuro Team, Japan
| |
Collapse
|
47
|
Application of co-culture technology of epithelial type cells and mesenchymal type cells using nanopatterned structures. PLoS One 2020; 15:e0232899. [PMID: 32392240 PMCID: PMC7213697 DOI: 10.1371/journal.pone.0232899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/23/2020] [Indexed: 01/23/2023] Open
Abstract
Various nanopatterning techniques have been developed to improve cell proliferation and differentiation efficiency. As we previously reported, nanopillars and pores are able to sustain human pluripotent stem cells and differentiate pancreatic cells. From this, the nanoscale patterns would be effective environment for the co-culturing of epithelial and mesenchymal cell types. Interestingly, the nanopatterning selectively reduced the proliferative rate of mesenchymal cells while increasing the expression of adhesion protein in epithelial type cells. Additionally, co-cultured cells on the nanopatterning were not negatively affected in terms of cell function metabolic ability or cell survival. This is in contrast to conventional co-culturing methods such as ultraviolet or chemical treatments. The nanopatterning appears to be an effective environment for mesenchymal co-cultures with typically low proliferative rates cells such as astrocytes, neurons, melanocytes, and fibroblasts without using potentially damaging treatments.
Collapse
|
48
|
Liu L, Koo Y, Russell T, Gay E, Li Y, Yun Y. Three-dimensional brain-on-chip model using human iPSC-derived GABAergic neurons and astrocytes: Butyrylcholinesterase post-treatment for acute malathion exposure. PLoS One 2020; 15:e0230335. [PMID: 32163499 PMCID: PMC7067464 DOI: 10.1371/journal.pone.0230335] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 02/26/2020] [Indexed: 12/24/2022] Open
Abstract
Organophosphates (OPs) induce acute and chronic neurotoxicity, primarily by inhibiting acetylcholinesterase (AChE) activity as well as by necrosis, and apoptosis. Butyrylcholinesterase (BuChE), an exogenous bioscavenger of OPs, can be used as a treatment for OP exposure. It is prerequisite to develop in vitro brain models that can study BuChE post-treatment for acute OP exposure. In this study, we developed a three-dimensional (3D) brain-on-chip platform with human induced pluripotent stem cell (iPSC)-derived neurons and astrocytes to simulate human brain behavior. The platform consists of two compartments: 1) a hydrogel embedded with human iPSC-derived GABAergic neurons and astrocytes and 2) a perfusion channel with dynamic medium flow. The brain tissue constructs were exposed to Malathion (MT) at various concentrations and then treated with BuChE after 20 minutes of MT exposure. Results show that the iPSC-derived neurons and astrocytes directly interacted and formed synapses in the 3D matrix, and that treatment with BuChE improved viability after MT exposure up to a concentration of 10−3 M. We conclude that the 3D brain-on-chip platform with human iPSC-derived brain cells is a suitable model to study the neurotoxicity of OP exposure and evaluate therapeutic compounds for treatment.
Collapse
Affiliation(s)
- Lumei Liu
- FIT BEST Laboratory, Department of Chemical, Biological, and Bio Engineering, North Carolina Agricultural and Technical State University, Greensboro, North Carolina, United States of America
| | - Youngmi Koo
- FIT BEST Laboratory, Department of Chemical, Biological, and Bio Engineering, North Carolina Agricultural and Technical State University, Greensboro, North Carolina, United States of America
| | - Teal Russell
- FIT BEST Laboratory, Department of Chemical, Biological, and Bio Engineering, North Carolina Agricultural and Technical State University, Greensboro, North Carolina, United States of America
| | - Elaine Gay
- Center for Drug Discovery, RTI International, Research Triangle Park, Durham, North Carolina, United States of America
| | - Yan Li
- Chemical Engineering, Florida A&M University-Florida State University, Tallahassee, Florida, United States of America
| | - Yeoheung Yun
- FIT BEST Laboratory, Department of Chemical, Biological, and Bio Engineering, North Carolina Agricultural and Technical State University, Greensboro, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
49
|
Characterization and application of electrically active neuronal networks established from human induced pluripotent stem cell-derived neural progenitor cells for neurotoxicity evaluation. Stem Cell Res 2020; 45:101761. [PMID: 32244191 DOI: 10.1016/j.scr.2020.101761] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 02/20/2020] [Accepted: 03/05/2020] [Indexed: 12/13/2022] Open
Abstract
Neurotoxicity is mediated by a variety of modes-of-actions leading to disturbance of neuronal function. In order to screen larger numbers of compounds for their neurotoxic potential, in vitro functional neuronal networks (NN) might be helpful tools. We established and characterized human NN (hNN) from hiPSC-derived neural progenitor cells by comparing hNN formation with two different differentiation media: in presence (CINDA) and absence (neural differentiation medium (NDM)) of maturation-supporting factors. As a NN control we included differentiating rat NN (rNN) in the study. Gene/protein expression and electrical activity from in vitro developing NN were assessed at multiple time points. Transcriptomes of 5, 14 and 28 days in vitro CINDA-grown hNN were compared to gene expression profiles of in vivo human developing brains. Molecular expression analyses as well as measures of electrical activity indicate that NN mature into neurons of different subtypes and astrocytes over time. In contrast to rNN, hNN are less electrically active within the same period of differentiation time, yet hNN grown in CINDA medium develop higher firing rates than hNN without supplements. Challenge of NN with neuronal receptor stimulators and inhibitors demonstrate presence of inhibitory, GABAergic neurons, whereas glutamatergic responses are limited. hiPSC-derived GABAergic hNN grown in CINDA medium might be a useful tool as part of an in vitro battery for assessing neurotoxicity.
Collapse
|
50
|
Zou Y, Zhao Y, Xiao Z, Chen B, Ma D, Shen H, Gu R, Dai J. Comparison of Regenerative Effects of Transplanting Three-Dimensional Longitudinal Scaffold Loaded-Human Mesenchymal Stem Cells and Human Neural Stem Cells on Spinal Cord Completely Transected Rats. ACS Biomater Sci Eng 2020; 6:1671-1680. [DOI: 10.1021/acsbiomaterials.9b01790] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Yunlong Zou
- China−Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing 100101, China
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing 100101, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing 100101, China
| | - Dezun Ma
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing 100101, China
| | - He Shen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing 100101, China
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Rui Gu
- China−Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing 100101, China
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| |
Collapse
|