1
|
Zhao Y, Liu K, Wang Y, Ma Y, Guo W, Shi C. Human-mouse chimeric brain models constructed from iPSC-derived brain cells: Applications and challenges. Exp Neurol 2024; 379:114848. [PMID: 38857749 DOI: 10.1016/j.expneurol.2024.114848] [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: 03/08/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/12/2024]
Abstract
The establishment of reliable human brain models is pivotal for elucidating specific disease mechanisms and facilitating the discovery of novel therapeutic strategies for human brain disorders. Human induced pluripotent stem cell (iPSC) exhibit remarkable self-renewal capabilities and can differentiate into specialized cell types. This makes them a valuable cell source for xenogeneic or allogeneic transplantation. Human-mouse chimeric brain models constructed from iPSC-derived brain cells have emerged as valuable tools for modeling human brain diseases and exploring potential therapeutic strategies for brain disorders. Moreover, the integration and functionality of grafted stem cells has been effectively assessed using these models. Therefore, this review provides a comprehensive overview of recent progress in differentiating human iPSC into various highly specialized types of brain cells. This review evaluates the characteristics and functions of the human-mouse chimeric brain model. We highlight its potential roles in brain function and its ability to reconstruct neural circuitry in vivo. Additionally, we elucidate factors that influence the integration and differentiation of human iPSC-derived brain cells in vivo. This review further sought to provide suitable research models for cell transplantation therapy. These research models provide new insights into neuropsychiatric disorders, infectious diseases, and brain injuries, thereby advancing related clinical and academic research.
Collapse
Affiliation(s)
- Ya Zhao
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Ke Liu
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China; Gansu University of traditional Chinese medicine, Lanzhou 730030, PR China
| | - Yinghua Wang
- Medical College of Yan'an University, Yan'an 716000, PR China
| | - Yifan Ma
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China; Gansu University of traditional Chinese medicine, Lanzhou 730030, PR China
| | - Wenwen Guo
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Changhong Shi
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| |
Collapse
|
2
|
Nonaka H, Kondo T, Suga M, Yamanaka R, Sagara Y, Tsukita K, Mitsutomi N, Homma K, Saito R, Miyoshi F, Ohzeki H, Okuyama M, Inoue H. Induced pluripotent stem cell-based assays recapture multiple properties of human astrocytes. J Cell Mol Med 2024; 28:e18214. [PMID: 38509731 PMCID: PMC10955154 DOI: 10.1111/jcmm.18214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/01/2024] [Accepted: 02/20/2024] [Indexed: 03/22/2024] Open
Abstract
The majority of the population of glial cells in the central nervous system consists of astrocytes, and impairment of astrocytes causes various disorders. It is useful to assess the multiple astrocytic properties in order to understand their complex roles in the pathophysiology. Although we can differentiate human astrocytes from induced pluripotent stem cells (iPSCs), it remains unknown how we can analyse and reveal the multiple properties of astrocytes in complexed human disease conditions. For this purpose, we tested astrocytic differentiation protocols from feeder-free iPSCs based on the previous method with some modifications. Then, we set up extra- and intracellular assessments of iPSC-derived astrocytes by testing cytokine release, calcium influx, autophagy induction and migration. The results led us to analytic methods with conditions in which iPSC-derived astrocytes behave as in vivo. Finally, we applied these methods for modelling an astrocyte-related disease, Alexander disease. An analytic system using iPSC-derived astrocytes could be used to recapture complexities in human astrocyte diseases.
Collapse
Affiliation(s)
- Hideki Nonaka
- iPSC‐based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC)KyotoJapan
- Mitsubishi Tanabe Pharma CorporationYokohamaJapan
| | - Takayuki Kondo
- iPSC‐based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC)KyotoJapan
- Center for iPS Cell Research and Application (CiRA)Kyoto UniversityKyotoJapan
- Medical‐risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP)KyotoJapan
| | - Mika Suga
- iPSC‐based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC)KyotoJapan
- Center for iPS Cell Research and Application (CiRA)Kyoto UniversityKyotoJapan
| | - Ryu Yamanaka
- Mitsubishi Tanabe Pharma CorporationYokohamaJapan
| | - Yukako Sagara
- iPSC‐based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC)KyotoJapan
- Center for iPS Cell Research and Application (CiRA)Kyoto UniversityKyotoJapan
| | - Kayoko Tsukita
- iPSC‐based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC)KyotoJapan
- Center for iPS Cell Research and Application (CiRA)Kyoto UniversityKyotoJapan
| | | | - Kengo Homma
- Mitsubishi Tanabe Pharma CorporationYokohamaJapan
| | - Ryuta Saito
- Mitsubishi Tanabe Pharma CorporationYokohamaJapan
| | | | | | | | - Haruhisa Inoue
- iPSC‐based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC)KyotoJapan
- Center for iPS Cell Research and Application (CiRA)Kyoto UniversityKyotoJapan
- Medical‐risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP)KyotoJapan
| |
Collapse
|
3
|
Provenzano F, Torazza C, Bonifacino T, Bonanno G, Milanese M. The Key Role of Astrocytes in Amyotrophic Lateral Sclerosis and Their Commitment to Glutamate Excitotoxicity. Int J Mol Sci 2023; 24:15430. [PMID: 37895110 PMCID: PMC10607805 DOI: 10.3390/ijms242015430] [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: 08/25/2023] [Revised: 10/12/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
In the last two decades, there has been increasing evidence supporting non-neuronal cells as active contributors to neurodegenerative disorders. Among glial cells, astrocytes play a pivotal role in driving amyotrophic lateral sclerosis (ALS) progression, leading the scientific community to focus on the "astrocytic signature" in ALS. Here, we summarized the main pathological mechanisms characterizing astrocyte contribution to MN damage and ALS progression, such as neuroinflammation, mitochondrial dysfunction, oxidative stress, energy metabolism impairment, miRNAs and extracellular vesicles contribution, autophagy dysfunction, protein misfolding, and altered neurotrophic factor release. Since glutamate excitotoxicity is one of the most relevant ALS features, we focused on the specific contribution of ALS astrocytes in this aspect, highlighting the known or potential molecular mechanisms by which astrocytes participate in increasing the extracellular glutamate level in ALS and, conversely, undergo the toxic effect of the excessive glutamate. In this scenario, astrocytes can behave as "producers" and "targets" of the high extracellular glutamate levels, going through changes that can affect themselves and, in turn, the neuronal and non-neuronal surrounding cells, thus actively impacting the ALS course. Moreover, this review aims to point out knowledge gaps that deserve further investigation.
Collapse
Affiliation(s)
- Francesca Provenzano
- Department of Pharmacy (DIFAR), University of Genoa, 16148 Genova, Italy; (F.P.); (C.T.); (G.B.); (M.M.)
| | - Carola Torazza
- Department of Pharmacy (DIFAR), University of Genoa, 16148 Genova, Italy; (F.P.); (C.T.); (G.B.); (M.M.)
| | - Tiziana Bonifacino
- Department of Pharmacy (DIFAR), University of Genoa, 16148 Genova, Italy; (F.P.); (C.T.); (G.B.); (M.M.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| | - Giambattista Bonanno
- Department of Pharmacy (DIFAR), University of Genoa, 16148 Genova, Italy; (F.P.); (C.T.); (G.B.); (M.M.)
| | - Marco Milanese
- Department of Pharmacy (DIFAR), University of Genoa, 16148 Genova, Italy; (F.P.); (C.T.); (G.B.); (M.M.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
4
|
Arioka Y, Okumura H, Sakaguchi H, Ozaki N. Shedding light on latent pathogenesis and pathophysiology of mental disorders: The potential of iPS cell technology. Psychiatry Clin Neurosci 2023; 77:308-314. [PMID: 36929185 PMCID: PMC11488641 DOI: 10.1111/pcn.13545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 03/04/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
Mental disorders are considered as one of the major healthcare issues worldwide owing to their significant impact on the quality of life of patients, causing serious social burdens. However, it is hard to examine the living brain-a source of psychiatric symptoms-at the cellular, subcellular, and molecular levels, which poses difficulty in determining the pathogenesis and pathophysiology of mental disorders. Recently, induced pluripotent stem cell (iPSC) technology has been used as a novel tool for research on mental disorders. We believe that the iPSC-based studies will address the limitations of other research approaches, such as human genome, postmortem brain study, brain imaging, and animal model analysis. Notably, studies using integrated iPSC technology with genetic information have provided significant novel findings to date. This review aimed to discuss the history, current trends, potential, and future of iPSC technology in the field of mental disorders. Although iPSC technology has several limitations, this technology can be used in combination with the other approaches to facilitate studies on mental disorders.
Collapse
Affiliation(s)
- Yuko Arioka
- Pathophysiology of Mental DisordersNagoya University Graduate School of MedicineNagoyaJapan
- Center for Advanced Medicine and Clinical ResearchNagoya University HospitalNagoyaJapan
| | - Hiroki Okumura
- Pathophysiology of Mental DisordersNagoya University Graduate School of MedicineNagoyaJapan
- Hospital PharmacyNagoya University HospitalNagoyaJapan
| | - Hideya Sakaguchi
- BDR‐Otsuka Pharmaceutical Collaboration Center, RIKEN Center for Biosystems Dynamics ResearchKobeJapan
| | - Norio Ozaki
- Pathophysiology of Mental DisordersNagoya University Graduate School of MedicineNagoyaJapan
- Institute for Glyco‐core Research (iGCORE)Nagoya UniversityNagoyaJapan
| |
Collapse
|
5
|
Muzzi L, Di Lisa D, Falappa M, Pepe S, Maccione A, Pastorino L, Martinoia S, Frega M. Human-Derived Cortical Neurospheroids Coupled to Passive, High-Density and 3D MEAs: A Valid Platform for Functional Tests. Bioengineering (Basel) 2023; 10:bioengineering10040449. [PMID: 37106636 PMCID: PMC10136157 DOI: 10.3390/bioengineering10040449] [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: 03/03/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023] Open
Abstract
With the advent of human-induced pluripotent stem cells (hiPSCs) and differentiation protocols, methods to create in-vitro human-derived neuronal networks have been proposed. Although monolayer cultures represent a valid model, adding three-dimensionality (3D) would make them more representative of an in-vivo environment. Thus, human-derived 3D structures are becoming increasingly used for in-vitro disease modeling. Achieving control over the final cell composition and investigating the exhibited electrophysiological activity is still a challenge. Thence, methodologies to create 3D structures with controlled cellular density and composition and platforms capable of measuring and characterizing the functional aspects of these samples are needed. Here, we propose a method to rapidly generate neurospheroids of human origin with control over cell composition that can be used for functional investigations. We show a characterization of the electrophysiological activity exhibited by the neurospheroids by using micro-electrode arrays (MEAs) with different types (i.e., passive, C-MOS, and 3D) and number of electrodes. Neurospheroids grown in free culture and transferred on MEAs exhibited functional activity that can be chemically and electrically modulated. Our results indicate that this model holds great potential for an in-depth study of signal transmission to drug screening and disease modeling and offers a platform for in-vitro functional testing.
Collapse
Affiliation(s)
- Lorenzo Muzzi
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genoa, 16145 Genoa, Italy
| | - Donatella Di Lisa
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genoa, 16145 Genoa, Italy
| | - Matteo Falappa
- 3Brain AG, 8808 Pfäffikon, Switzerland
- Corticale Srl., 16145 Genoa, Italy
| | - Sara Pepe
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
| | | | - Laura Pastorino
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genoa, 16145 Genoa, Italy
| | - Sergio Martinoia
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genoa, 16145 Genoa, Italy
| | - Monica Frega
- Department of Clinical Neurophysiology, University of Twente, 7522 NB Enschede, The Netherlands
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
6
|
Konstantinidis E, Portal B, Mothes T, Beretta C, Lindskog M, Erlandsson A. Intracellular deposits of amyloid-beta influence the ability of human iPSC-derived astrocytes to support neuronal function. J Neuroinflammation 2023; 20:3. [PMID: 36593462 PMCID: PMC9809017 DOI: 10.1186/s12974-022-02687-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Astrocytes are crucial for maintaining brain homeostasis and synaptic function, but are also tightly connected to the pathogenesis of Alzheimer's disease (AD). Our previous data demonstrate that astrocytes ingest large amounts of aggregated amyloid-beta (Aβ), but then store, rather than degrade the ingested material, which leads to severe cellular stress. However, the involvement of pathological astrocytes in AD-related synaptic dysfunction remains to be elucidated. METHODS In this study, we aimed to investigate how intracellular deposits of Aβ in astrocytes affect their interplay with neurons, focusing on neuronal function and viability. For this purpose, human induced pluripotent stem cell (hiPSC)-derived astrocytes were exposed to sonicated Αβ42 fibrils. The direct and indirect effects of the Αβ-exposed astrocytes on hiPSC-derived neurons were analyzed by performing astrocyte-neuron co-cultures as well as additions of conditioned media or extracellular vesicles to pure neuronal cultures. RESULTS Electrophysiological recordings revealed significantly decreased frequency of excitatory post-synaptic currents in neurons co-cultured with Aβ-exposed astrocytes, while conditioned media from Aβ-exposed astrocytes had the opposite effect and resulted in hyperactivation of the synapses. Clearly, factors secreted from control, but not from Aβ-exposed astrocytes, benefited the wellbeing of neuronal cultures. Moreover, reactive astrocytes with Aβ deposits led to an elevated clearance of dead cells in the co-cultures. CONCLUSIONS Taken together, our results demonstrate that inclusions of aggregated Aβ affect the reactive state of the astrocytes, as well as their ability to support neuronal function.
Collapse
Affiliation(s)
- Evangelos Konstantinidis
- grid.8993.b0000 0004 1936 9457Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, 751 85 Uppsala, Sweden
| | - Benjamin Portal
- grid.8993.b0000 0004 1936 9457Department of Medical Cell Biology, Uppsala University, 751 23 Uppsala, Sweden
| | - Tobias Mothes
- grid.8993.b0000 0004 1936 9457Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, 751 85 Uppsala, Sweden
| | - Chiara Beretta
- grid.8993.b0000 0004 1936 9457Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, 751 85 Uppsala, Sweden
| | - Maria Lindskog
- grid.8993.b0000 0004 1936 9457Department of Medical Cell Biology, Uppsala University, 751 23 Uppsala, Sweden
| | - Anna Erlandsson
- grid.8993.b0000 0004 1936 9457Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, 751 85 Uppsala, Sweden
| |
Collapse
|
7
|
Yang Z, Gong M, Yang C, Chen C, Zhang K. Applications of Induced Pluripotent Stem Cell-Derived Glia in Brain Disease Research and Treatment. Handb Exp Pharmacol 2023; 281:103-140. [PMID: 37735301 DOI: 10.1007/164_2023_697] [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] [Indexed: 09/23/2023]
Abstract
Glia are integral components of neural networks and are crucial in both physiological functions and pathological processes of the brain. Many brain diseases involve glial abnormalities, including inflammatory changes, mitochondrial damage, calcium signaling disturbance, hemichannel opening, and loss of glutamate transporters. Induced pluripotent stem cell (iPSC)-derived glia provide opportunities to study the contributions of glia in human brain diseases. These cells have been used for human disease modeling as well as generating new therapies. This chapter introduces glial involvement in brain diseases, then summarizes different methods of generating iPSC-derived glia disease models of these cells. Finally, strategies for treating disease using iPSC-derived glia are discussed. The goal of this chapter is to provide an overview and shed light on the applications of iPSC-derived glia in brain disease research and treatment.
Collapse
Affiliation(s)
- Zhiqi Yang
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China
| | - Mingyue Gong
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China
| | - Chuanyan Yang
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China
| | - Chunhai Chen
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Kuan Zhang
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China.
| |
Collapse
|
8
|
Uchino K, Tanaka Y, Kawaguchi S, Kubota K, Watanabe T, Katsurabayashi S, Hirose S, Iwasaki K. Establishment of autaptic culture with human-induced pluripotent stem cell-derived astrocytes. iScience 2022; 25:104762. [PMID: 35942096 PMCID: PMC9356095 DOI: 10.1016/j.isci.2022.104762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/25/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022] Open
Abstract
Although astrocytes are involved in the pathogenesis of CNS diseases, how they induce synaptic abnormalities is unclear. Currently, in vitro pathological astrocyte cultures or animal models do not reproduce human disease phenotypes accurately. Induced pluripotent stem cells (iPSCs) are replacing animal models in pathological studies. We developed an autaptic culture (AC) system containing single neuron cultures grown on microislands of astrocytes. AC with human iPSC-derived astrocytes (HiA) was established. We evaluated the effect of astrocytes on the synaptic functions of human-derived neurons. We found a significantly higher Na+ current amplitude, membrane capacitance, and number of synapses, as well as longer dendrites, in HiAACs compared with neuron monocultures. Furthermore, HiAs were involved in the formation and maturation of functional synapses that exhibited excitatory postsynaptic currents. This system can facilitate the study of CNS diseases and advance the development of drugs targeting glial cells. We developed an autaptic culture with human iPSCs-derived astrocytes Neurons in HiAACs developed after culture and formed functional synapses EPSC and mEPSC were recorded showing HiAs promoted synapse formation/maturation Autaptic cultures can be used to analyze synaptic activity and human CNS disease
Collapse
Affiliation(s)
- Kouya Uchino
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Yasuyoshi Tanaka
- Department of Advanced Pharmacology, Daiichi University of Pharmacy, 22-1 Tamagawa-machi, Minami-ku, Fukuoka 815-8511, Japan
- iONtarget, Co., Inc., 1-3-70-5805 Momochihama, Sawara-ku, Fukuoka 814-0006, Japan
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Sayaka Kawaguchi
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Kaori Kubota
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Takuya Watanabe
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Shutaro Katsurabayashi
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
- Corresponding author
| | - Shinichi Hirose
- iONtarget, Co., Inc., 1-3-70-5805 Momochihama, Sawara-ku, Fukuoka 814-0006, Japan
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
- General Medical Research Center, School of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Katsunori Iwasaki
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| |
Collapse
|
9
|
Insights into Human-Induced Pluripotent Stem Cell-Derived Astrocytes in Neurodegenerative Disorders. Biomolecules 2022; 12:biom12030344. [PMID: 35327542 PMCID: PMC8945600 DOI: 10.3390/biom12030344] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
Most neurodegenerative disorders have complex and still unresolved pathology characterized by progressive neuronal damage and death. Astrocytes, the most-abundant non-neuronal cell population in the central nervous system, play a vital role in these processes. They are involved in various functions in the brain, such as the regulation of synapse formation, neuroinflammation, and lactate and glutamate levels. The development of human-induced pluripotent stem cells (iPSCs) reformed the research in neurodegenerative disorders allowing for the generation of disease-relevant neuronal and non-neuronal cell types that can help in disease modeling, drug screening, and, possibly, cell transplantation strategies. In the last 14 years, the differentiation of human iPSCs into astrocytes allowed for the opportunity to explore the contribution of astrocytes to neurodegenerative diseases. This review discusses the development protocols and applications of human iPSC-derived astrocytes in the most common neurodegenerative conditions.
Collapse
|
10
|
Functional Characterization of Human Pluripotent Stem Cell-Derived Models of the Brain with Microelectrode Arrays. Cells 2021; 11:cells11010106. [PMID: 35011667 PMCID: PMC8750870 DOI: 10.3390/cells11010106] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 12/26/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-derived neuron cultures have emerged as models of electrical activity in the human brain. Microelectrode arrays (MEAs) measure changes in the extracellular electric potential of cell cultures or tissues and enable the recording of neuronal network activity. MEAs have been applied to both human subjects and hPSC-derived brain models. Here, we review the literature on the functional characterization of hPSC-derived two- and three-dimensional brain models with MEAs and examine their network function in physiological and pathological contexts. We also summarize MEA results from the human brain and compare them to the literature on MEA recordings of hPSC-derived brain models. MEA recordings have shown network activity in two-dimensional hPSC-derived brain models that is comparable to the human brain and revealed pathology-associated changes in disease models. Three-dimensional hPSC-derived models such as brain organoids possess a more relevant microenvironment, tissue architecture and potential for modeling the network activity with more complexity than two-dimensional models. hPSC-derived brain models recapitulate many aspects of network function in the human brain and provide valid disease models, but certain advancements in differentiation methods, bioengineering and available MEA technology are needed for these approaches to reach their full potential.
Collapse
|
11
|
Human iPSC-Derived Glia as a Tool for Neuropsychiatric Research and Drug Development. Int J Mol Sci 2021; 22:ijms221910254. [PMID: 34638595 PMCID: PMC8508580 DOI: 10.3390/ijms221910254] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 12/19/2022] Open
Abstract
Neuropsychiatric disorders such as schizophrenia or autism spectrum disorder represent a leading and growing burden on worldwide mental health. Fundamental lack in understanding the underlying pathobiology compromises efficient drug development despite the immense medical need. So far, antipsychotic drugs reduce symptom severity and enhance quality of life, but there is no cure available. On the molecular level, schizophrenia and autism spectrum disorders correlate with compromised neuronal phenotypes. There is increasing evidence that aberrant neuroinflammatory responses of glial cells account for synaptic pathologies through deregulated communication and reciprocal modulation. Consequently, microglia and astrocytes emerge as central targets for anti-inflammatory treatment to preserve organization and homeostasis of the central nervous system. Studying the impact of neuroinflammation in the context of neuropsychiatric disorders is, however, limited by the lack of relevant human cellular test systems that are able to represent the dynamic cellular processes and molecular changes observed in human tissue. Today, patient-derived induced pluripotent stem cells offer the opportunity to study neuroinflammatory mechanisms in vitro that comprise the genetic background of affected patients. In this review, we summarize the major findings of iPSC-based microglia and astrocyte research in the context of neuropsychiatric diseases and highlight the benefit of 2D and 3D co-culture models for the generation of efficient in vitro models for target screening and drug development.
Collapse
|
12
|
Gradišnik L, Bošnjak R, Maver T, Velnar T. Advanced Bio-Based Polymers for Astrocyte Cell Models. MATERIALS (BASEL, SWITZERLAND) 2021; 14:3664. [PMID: 34209194 PMCID: PMC8269866 DOI: 10.3390/ma14133664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 12/27/2022]
Abstract
The development of in vitro neural tissue analogs is of great interest for many biomedical engineering applications, including the tissue engineering of neural interfaces, treatment of neurodegenerative diseases, and in vitro evaluation of cell-material interactions. Since astrocytes play a crucial role in the regenerative processes of the central nervous system, the development of biomaterials that interact favorably with astrocytes is of great research interest. The sources of human astrocytes, suitable natural biomaterials, guidance scaffolds, and ligand patterned surfaces are discussed in the article. New findings in this field are essential for the future treatment of spinal cord and brain injuries.
Collapse
Affiliation(s)
- Lidija Gradišnik
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia;
- AMEU-ECM, Slovenska 17, 2000 Maribor, Slovenia
| | - Roman Bošnjak
- Department of Neurosurgery, University Medical Centre Ljubljana, Zaloska 7, 1000 Ljubljana, Slovenia;
| | - Tina Maver
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia;
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
| | - Tomaž Velnar
- AMEU-ECM, Slovenska 17, 2000 Maribor, Slovenia
- Department of Neurosurgery, University Medical Centre Ljubljana, Zaloska 7, 1000 Ljubljana, Slovenia;
| |
Collapse
|
13
|
Chen YT, Miao K, Zhou L, Xiong WN. Stem cell therapy for chronic obstructive pulmonary disease. Chin Med J (Engl) 2021; 134:1535-1545. [PMID: 34250959 PMCID: PMC8280064 DOI: 10.1097/cm9.0000000000001596] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Indexed: 12/25/2022] Open
Abstract
ABSTRACT Chronic obstructive pulmonary disease (COPD), characterized by persistent and not fully reversible airflow restrictions, is currently one of the most widespread chronic lung diseases in the world. The most common symptoms of COPD are cough, expectoration, and exertional dyspnea. Although various strategies have been developed during the last few decades, current medical treatment for COPD only focuses on the relief of symptoms, and the reversal of lung function deterioration and improvement in patient's quality of life are very limited. Consequently, development of novel effective therapeutic strategies for COPD is urgently needed. Stem cells were known to differentiate into a variety of cell types and used to regenerate lung parenchyma and airway structures. Stem cell therapy is a promising therapeutic strategy that has the potential to restore the lung function and improve the quality of life in patients with COPD. This review summarizes the current state of knowledge regarding the clinical research on the treatment of COPD with mesenchymal stem cells (MSCs) and aims to update the understanding of the role of MSCs in COPD treatment, which may be helpful for developing effective therapeutic strategies in clinical settings.
Collapse
Affiliation(s)
- Yun-Tian Chen
- Department of Pulmonary and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| | - Kang Miao
- Department of Pulmonary and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Linfu Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Wei-Ning Xiong
- Department of Pulmonary and Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
14
|
Cameron T, Bennet T, Rowe EM, Anwer M, Wellington CL, Cheung KC. Review of Design Considerations for Brain-on-a-Chip Models. MICROMACHINES 2021; 12:441. [PMID: 33921018 PMCID: PMC8071412 DOI: 10.3390/mi12040441] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023]
Abstract
In recent years, the need for sophisticated human in vitro models for integrative biology has motivated the development of organ-on-a-chip platforms. Organ-on-a-chip devices are engineered to mimic the mechanical, biochemical and physiological properties of human organs; however, there are many important considerations when selecting or designing an appropriate device for investigating a specific scientific question. Building microfluidic Brain-on-a-Chip (BoC) models from the ground-up will allow for research questions to be answered more thoroughly in the brain research field, but the design of these devices requires several choices to be made throughout the design development phase. These considerations include the cell types, extracellular matrix (ECM) material(s), and perfusion/flow considerations. Choices made early in the design cycle will dictate the limitations of the device and influence the end-point results such as the permeability of the endothelial cell monolayer, and the expression of cell type-specific markers. To better understand why the engineering aspects of a microfluidic BoC need to be influenced by the desired biological environment, recent progress in microfluidic BoC technology is compared. This review focuses on perfusable blood-brain barrier (BBB) and neurovascular unit (NVU) models with discussions about the chip architecture, the ECM used, and how they relate to the in vivo human brain. With increased knowledge on how to make informed choices when selecting or designing BoC models, the scientific community will benefit from shorter development phases and platforms curated for their application.
Collapse
Affiliation(s)
- Tiffany Cameron
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.C.); (T.B.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Tanya Bennet
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.C.); (T.B.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Elyn M. Rowe
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (E.M.R.); (M.A.); (C.L.W.)
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Mehwish Anwer
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (E.M.R.); (M.A.); (C.L.W.)
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Cheryl L. Wellington
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (E.M.R.); (M.A.); (C.L.W.)
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Karen C. Cheung
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.C.); (T.B.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Electrical & Computer Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
15
|
Current and future applications of induced pluripotent stem cell-based models to study pathological proteins in neurodegenerative disorders. Mol Psychiatry 2021; 26:2685-2706. [PMID: 33495544 PMCID: PMC8505258 DOI: 10.1038/s41380-020-00999-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/02/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022]
Abstract
Neurodegenerative disorders emerge from the failure of intricate cellular mechanisms, which ultimately lead to the loss of vulnerable neuronal populations. Research conducted across several laboratories has now provided compelling evidence that pathogenic proteins can also contribute to non-cell autonomous toxicity in several neurodegenerative contexts, including Alzheimer's, Parkinson's, and Huntington's diseases as well as Amyotrophic Lateral Sclerosis. Given the nearly ubiquitous nature of abnormal protein accumulation in such disorders, elucidating the mechanisms and routes underlying these processes is essential to the development of effective treatments. To this end, physiologically relevant human in vitro models are critical to understand the processes surrounding uptake, release and nucleation under physiological or pathological conditions. This review explores the use of human-induced pluripotent stem cells (iPSCs) to study prion-like protein propagation in neurodegenerative diseases, discusses advantages and limitations of this model, and presents emerging technologies that, combined with the use of iPSC-based models, will provide powerful model systems to propel fundamental research forward.
Collapse
|
16
|
D’Souza GX, Rose SE, Knupp A, Nicholson DA, Keene CD, Young JE. The application of in vitro-derived human neurons in neurodegenerative disease modeling. J Neurosci Res 2021; 99:124-140. [PMID: 32170790 PMCID: PMC7487003 DOI: 10.1002/jnr.24615] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/19/2020] [Accepted: 02/27/2020] [Indexed: 02/02/2023]
Abstract
The development of safe and effective treatments for age-associated neurodegenerative disorders is an on-going challenge faced by the scientific field. Key to the development of such therapies is the appropriate selection of modeling systems in which to investigate disease mechanisms and to test candidate interventions. There are unique challenges in the development of representative laboratory models of neurodegenerative diseases, including the complexity of the human brain, the cumulative and variable contributions of genetic and environmental factors over the course of a lifetime, inability to culture human primary neurons, and critical central nervous system differences between small animal models and humans. While traditional rodent models have advanced our understanding of neurodegenerative disease mechanisms, key divergences such as the species-specific genetic background can limit the application of animal models in many cases. Here we review in vitro human neuronal systems that employ stem cell and reprogramming technology and their application to a range of neurodegenerative diseases. Specifically, we compare human-induced pluripotent stem cell-derived neurons to directly converted, or transdifferentiated, induced neurons, as both model systems can take advantage of patient-derived human tissue to produce neurons in culture. We present recent technical developments using these two modeling systems, as well as current limitations to these systems, with the aim of advancing investigation of neuropathogenic mechanisms using these models.
Collapse
Affiliation(s)
- Gary X. D’Souza
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Shannon E. Rose
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Allison Knupp
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Daniel A. Nicholson
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - C. Dirk Keene
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Jessica E. Young
- Department of Pathology, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine (ISCRM), University of Washington, Seattle, WA, USA
| |
Collapse
|
17
|
Lipus A, Janosz E, Ackermann M, Hetzel M, Dahlke J, Buchegger T, Wunderlich S, Martin U, Cathomen T, Schambach A, Moritz T, Lachmann N. Targeted Integration of Inducible Caspase-9 in Human iPSCs Allows Efficient in vitro Clearance of iPSCs and iPSC-Macrophages. Int J Mol Sci 2020; 21:E2481. [PMID: 32260086 PMCID: PMC7177583 DOI: 10.3390/ijms21072481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 03/30/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) offer great promise for the field of regenerative medicine, and iPSC-derived cells have already been applied in clinical practice. However, potential contamination of effector cells with residual pluripotent cells (e.g., teratoma-initiating cells) or effector cell-associated side effects may limit this approach. This also holds true for iPSC-derived hematopoietic cells. Given the therapeutic benefit of macrophages in different disease entities and the feasibility to derive macrophages from human iPSCs, we established human iPSCs harboring the inducible Caspase-9 (iCasp9) suicide safety switch utilizing transcription activator-like effector nuclease (TALEN)-based designer nuclease technology. Mono- or bi-allelic integration of the iCasp9 gene cassette into the AAVS1 locus showed no effect on the pluripotency of human iPSCs and did not interfere with their differentiation towards macrophages. In both, iCasp9-mono and iCasp9-bi-allelic clones, concentrations of 0.1 nM AP20187 were sufficient to induce apoptosis in more than 98% of iPSCs and their progeny-macrophages. Thus, here we provide evidence that the introduction of the iCasp9 suicide gene into the AAVS1 locus enables the effective clearance of human iPSCs and thereof derived macrophages.
Collapse
Affiliation(s)
- Alexandra Lipus
- RG Reprogramming and Gene Therapy, Hannover Medical School, Hannover 30625, Germany; (A.L.); (E.J.); (M.H.); (T.M.)
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Ewa Janosz
- RG Reprogramming and Gene Therapy, Hannover Medical School, Hannover 30625, Germany; (A.L.); (E.J.); (M.H.); (T.M.)
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Mania Ackermann
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
- RG Translational Hematology of Congenital Diseases, Hannover Medical School, Hannover 30625, Germany
| | - Miriam Hetzel
- RG Reprogramming and Gene Therapy, Hannover Medical School, Hannover 30625, Germany; (A.L.); (E.J.); (M.H.); (T.M.)
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Julia Dahlke
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Theresa Buchegger
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
- RG Translational Hematology of Congenital Diseases, Hannover Medical School, Hannover 30625, Germany
| | - Stephanie Wunderlich
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, REBIRTH, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover 30625, Germany; (S.W.); (U.M.)
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, REBIRTH, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover 30625, Germany; (S.W.); (U.M.)
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg 79106, Germany;
- Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, Freiburg 79095, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Thomas Moritz
- RG Reprogramming and Gene Therapy, Hannover Medical School, Hannover 30625, Germany; (A.L.); (E.J.); (M.H.); (T.M.)
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Nico Lachmann
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
- RG Translational Hematology of Congenital Diseases, Hannover Medical School, Hannover 30625, Germany
| |
Collapse
|
18
|
Zarei-Kheirabadi M, Vaccaro AR, Rahimi-Movaghar V, Kiani S, Baharvand H. An Overview of Extrinsic and Intrinsic Mechanisms Involved in Astrocyte Development in the Central Nervous System. Stem Cells Dev 2020; 29:266-280. [PMID: 31847709 DOI: 10.1089/scd.2019.0189] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Over the past few decades, our knowledge about the function of the central nervous system (CNS) and astrocytes has improved, and research has confirmed the key roles that astrocytes play in the physiology and pathology of the CNS. Here, we reviewed the intrinsic and extrinsic mechanisms that regulate the development of astrocytes, which are generated from radial glial cells. These regulatory systems modulate various signaling pathways and transcription factors. In this review, four stages of astrocyte development-specification (patterning and switch), migration, proliferation, and maturation, are discussed. In astrocyte patterning, VA1-VA3 domains create the astrocyte subtypes by differential expression of Slit1 and Reelin in the spinal cord. In the brain, patterning creates several astrocyte subtypes by different organizing centers. At the switch step, the janus kinase-signal transducer and activator of transcription pathway governs the transition of neurogenesis to gliogenesis. Bone marrow protein and Notch pathways are also important players of the progliogenic switch. Intrinsic regulation is mediated by DNA methylation transferases, and polycomb group complexes can intrinsically affect the development of astrocytes. In the next stage, these cells proliferate and migrate to their final location. Astrocyte maturation is accomplished through the development of cellular processes, molecular markers, and functions.
Collapse
Affiliation(s)
- Masoumeh Zarei-Kheirabadi
- Department of Brain, Cognitive Sciences and Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Alexander R Vaccaro
- Department of Orthopedics, Rothman Orthopedic Institute, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Vafa Rahimi-Movaghar
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sahar Kiani
- Department of Brain, Cognitive Sciences and Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
19
|
Neural In Vitro Models for Studying Substances Acting on the Central Nervous System. Handb Exp Pharmacol 2020; 265:111-141. [PMID: 32594299 DOI: 10.1007/164_2020_367] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Animal models have been greatly contributing to our understanding of physiology, mechanisms of diseases, and toxicity. Yet, their limitations due to, e.g., interspecies variation are reflected in the high number of drug attrition rates, especially in central nervous system (CNS) diseases. Therefore, human-based neural in vitro models for studying safety and efficacy of substances acting on the CNS are needed. Human iPSC-derived cells offer such a platform with the unique advantage of reproducing the "human context" in vitro by preserving the genetic and molecular phenotype of their donors. Guiding the differentiation of hiPSC into cells of the nervous system and combining them in a 2D or 3D format allows to obtain complex models suitable for investigating neurotoxicity or brain-related diseases with patient-derived cells. This chapter will give an overview over stem cell-based human 2D neuronal and mixed neuronal/astrocyte models, in vitro cultures of microglia, as well as CNS disease models and considers new developments in the field, more specifically the use of brain organoids and 3D bioprinted in vitro models for safety and efficacy evaluation.
Collapse
|