1
|
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
|
2
|
Sun C, Rosenstock TR, Cohen MA, Sarkar S. Autophagy Dysfunction as a Phenotypic Readout in hiPSC-Derived Neuronal Cell Models of Neurodegenerative Diseases. Methods Mol Biol 2022; 2549:103-136. [PMID: 34490597 DOI: 10.1007/7651_2021_420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Autophagy is an evolutionarily conserved catabolic pathway for the degradation of cytoplasmic constituents in eukaryotic cells. It is the primary disposal route for selective removal of undesirable cellular materials like aggregation-prone proteins and damaged organelles for maintaining cellular homeostasis, and for bulk degradation of intracellular macromolecules and recycling the breakdown products for providing energy homeostasis during starvation. These functions of autophagy are attributed to cellular survival and thus pertinent for human health; however, malfunction of this process is detrimental to the cells, particularly for post-mitotic neurons. Thus, basal autophagy is vital for maintaining neuronal homeostasis, whereas autophagy dysfunction contributes to neurodegeneration. Defective autophagy has been demonstrated in several neurodegenerative diseases wherein pharmacological induction of autophagy is beneficial in many of these disease models. Elucidating the mechanisms underlying defective autophagy is imperative for the development of therapies targeting this process. Disease-affected human neuronal cells can be established from patient-derived human induced pluripotent stem cells (hiPSCs) that provide a clinically relevant platform for studying disease mechanisms and drug discovery. Thus, modeling autophagy dysfunction as a phenotypic readout in patient-derived neurons provides a more direct platform for investigating the mechanisms underlying defective autophagy and evaluating the therapeutic efficacy of autophagy inducers. Toward this, several hiPSC-derived neuronal cell models of neurodegenerative diseases have been employed. In this review, we highlight the key methodologies pertaining to hiPSC maintenance and neuronal differentiation, and studying autophagy at an endogenous level in hiPSC-derived neuronal cells.
Collapse
Affiliation(s)
- Congxin Sun
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Tatiana R Rosenstock
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Pharmacology, University of São Paulo, São Paulo, Brazil
| | - Malkiel A Cohen
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Sovan Sarkar
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
3
|
Rosenstock TR, Sun C, Hughes GW, Winter K, Sarkar S. Analysis of Mitochondrial Dysfunction by Microplate Reader in hiPSC-Derived Neuronal Cell Models of Neurodegenerative Disorders. Methods Mol Biol 2022; 2549:1-21. [PMID: 35347693 DOI: 10.1007/7651_2021_451] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Mitochondria are responsible for many vital pathways governing cellular homeostasis, including cellular energy management, heme biosynthesis, lipid metabolism, cellular proliferation and differentiation, cell cycle regulation, and cellular viability. Electron transport and ADP phosphorylation coupled with proton pumping through the mitochondrial complexes contribute to the preservation of mitochondrial membrane potential (ΔΨm). Importantly, mitochondrial polarization is essential for reactive oxygen species (ROS) production and cytosolic calcium (Ca2+) handling. Thus, changes in mitochondrial oxidative phosphorylation (OXPHOS), ΔΨm, and ATP/ADP may occur in parallel or stimulate each other. Brain cells like neurons are heavily reliant on mitochondrial OXPHOS for its high-energy demands, and hence improper mitochondrial function is detrimental for neuronal survival. Indeed, several neurodegenerative disorders are associated with mitochondrial dysfunction. Modeling this disease-relevant phenotype in neuronal cells differentiated from patient-derived human induced pluripotent stem cells (hiPSCs) provide an appropriate cellular platform for studying the disease pathology and drug discovery. In this review, we describe high-throughput analysis of crucial parameters related to mitochondrial function in hiPSC-derived neurons. These methodologies include measurement of ΔΨm, intracellular Ca2+, oxidative stress, and ATP/ADP levels using fluorescence probes via a microplate reader. Benefits of such an approach include analysis of mitochondrial parameters on a large population of cells, simultaneous analysis of different cell lines and experimental conditions, and for drug screening to identify compounds restoring mitochondrial function.
Collapse
Affiliation(s)
- Tatiana R Rosenstock
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
- Department of Pharmacology, University of São Paulo, São Paulo, Brazil
- Department of Bioscience, Sygnature Discovery, BioCity, Nottingham, United Kingdom
| | - Congxin Sun
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Georgina Wynne Hughes
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Katherine Winter
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Sovan Sarkar
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK.
| |
Collapse
|
4
|
A chalcone derivative suppresses TSLP induction in mice and human keratinocytes through binding to BET family proteins. Biochem Pharmacol 2021; 194:114819. [PMID: 34757034 DOI: 10.1016/j.bcp.2021.114819] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 11/23/2022]
Abstract
Although treatments for allergic diseases have improved, side effects and treatment resistance remain as challenges. New therapeutic drugs for allergic diseases are urgently required. Thymic stromal lymphopoietin (TSLP) is a cytokine target for prevention and treatment of allergic diseases. Since TSLP is produced from epithelial cells in allergic diseases, TSLP inhibitors may be new anti-allergic drugs. We previously identified a new inhibitor of TSLP production, named 16D10. However, its target of action remained unclarified. In this study, we found proteins binding to 16D10 from 24,000 human protein arrays by AlphaScreen-based high-throughput screening and identified bromodomain and extra-terminal (BET) family proteins as targets. We also clarified the detailed mode of interaction between 16D10 and a BET family protein using X-ray crystallography. Furthermore, we confirmed that inhibitors of BET family proteins suppressed TSLP induction and IL-33 and IL-36γ expression in both mouse and human keratinocyte cell lines. Taken together, our findings suggest that BET family proteins are involved in the suppression of TSLP production by 16D10. These proteins can contribute to the pathology of atopic dermatitis via TSLP regulation in keratinocytes and have potential as therapeutic targets in allergic diseases.
Collapse
|
5
|
Hosaka N, Kanda S, Shimono T, Nishiyama T. Induction of γδT cells from HSC-enriched BMCs co-cultured with iPSC-derived thymic epithelial cells. J Cell Mol Med 2021; 25:10604-10613. [PMID: 34687276 PMCID: PMC8581322 DOI: 10.1111/jcmm.16993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/25/2021] [Accepted: 09/30/2021] [Indexed: 12/29/2022] Open
Abstract
T cells bearing γδ antigen receptors have been investigated as potential treatments for several diseases, including malignant tumours. However, the clinical application of γδT cells has been hampered by their relatively low abundance in vivo and the technical difficulty of inducing their differentiation from hematopoietic stem cells (HSCs) in vitro. Here, we describe a novel method for generating mouse γδT cells by co-culturing HSC-enriched bone marrow cells (HSC-eBMCs) with induced thymic epithelial cells (iTECs) derived from induced pluripotent stem cells (iPSCs). We used BMCs from CD45.1 congenic C57BL/6 mice to distinguish them from iPSCs, which expressed CD45.2. We showed that HSC-eBMCs and iTECs cultured with IL-2 + IL-7 for up to 21 days induced CD45.1+ γδT cells that expressed a broad repertoire of Vγ and Vδ T-cell receptors. Notably, the induced lymphocytes contained few or no αβT cells, NK1.1+ natural killer cells, or B220+ B cells. Adoptive transfer of the induced γδT cells to leukemia-bearing mice significantly reduced tumour growth and prolonged mouse survival with no obvious side effects, such as tumorigenesis and autoimmune diseases. This new method suggests that it could also be used to produce human γδT cells for clinical applications.
Collapse
Affiliation(s)
- Naoki Hosaka
- Department of Pathology, Fuchu Hospital, Izumi, Osaka, Japan.,Department of Hygiene and Public Health, Kansai Medical University, Hitakata, Osaka, Japan
| | - Seiji Kanda
- Department of Hygiene and Public Health, Kansai Medical University, Hitakata, Osaka, Japan.,Regenerative Research Center for Intractable Diseases, Kansai Medical University, Hitakata, Osaka, Japan
| | - Takaki Shimono
- Department of Hygiene and Public Health, Kansai Medical University, Hitakata, Osaka, Japan.,Regenerative Research Center for Intractable Diseases, Kansai Medical University, Hitakata, Osaka, Japan
| | - Toshimasa Nishiyama
- Department of Hygiene and Public Health, Kansai Medical University, Hitakata, Osaka, Japan
| |
Collapse
|
6
|
Izsak J, Seth H, Iljin M, Theiss S, Ågren H, Funa K, Aigner L, Hanse E, Illes S. Differential acute impact of therapeutically effective and overdose concentrations of lithium on human neuronal single cell and network function. Transl Psychiatry 2021; 11:281. [PMID: 33980815 PMCID: PMC8115174 DOI: 10.1038/s41398-021-01399-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 04/10/2021] [Accepted: 04/19/2021] [Indexed: 01/18/2023] Open
Abstract
Lithium salts are used as mood-balancing medication prescribed to patients suffering from neuropsychiatric disorders, such as bipolar disorder and major depressive disorder. Lithium salts cross the blood-brain barrier and reach the brain parenchyma within few hours after oral application, however, how lithium influences directly human neuronal function is unknown. We applied patch-clamp and microelectrode array technology on human induced pluripotent stem cell (iPSC)-derived cortical neurons acutely exposed to therapeutic (<1 mM) and overdose concentrations (>1 mM) of lithium chloride (LiCl) to assess how therapeutically effective and overdose concentrations of LiCl directly influence human neuronal electrophysiological function at the synapse, single-cell, and neuronal network level. We describe that human iPSC-cortical neurons exposed to lithium showed an increased neuronal activity under all tested concentrations. Furthermore, we reveal a lithium-induced, concentration-dependent, transition of regular synchronous neuronal network activity using therapeutically effective concentration (<1 mM LiCl) to epileptiform-like neuronal discharges using overdose concentration (>1 mM LiCl). The overdose concentration lithium-induced epileptiform-like activity was similar to the epileptiform-like activity caused by the GABAA-receptor antagonist. Patch-clamp recordings reveal that lithium reduces action potential threshold at all concentrations, however, only overdose concentration causes increased frequency of spontaneous AMPA-receptor mediated transmission. By applying the AMPA-receptor antagonist and anti-epileptic drug Perampanel, we demonstrate that Perampanel suppresses lithium-induced epileptiform-like activity in human cortical neurons. We provide insights in how therapeutically effective and overdose concentration of lithium directly influences human neuronal function at synapse, a single neuron, and neuronal network levels. Furthermore, we provide evidence that Perampanel suppresses pathological neuronal discharges caused by overdose concentrations of lithium in human neurons.
Collapse
Affiliation(s)
- Julia Izsak
- grid.8761.80000 0000 9919 9582Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Henrik Seth
- grid.8761.80000 0000 9919 9582Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Margarita Iljin
- grid.8761.80000 0000 9919 9582Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Stephan Theiss
- grid.411327.20000 0001 2176 9917Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany ,Result Medical GmbH, Düsseldorf, Germany
| | - Hans Ågren
- grid.8761.80000 0000 9919 9582Institute of Neuroscience and Physiology, Section of Psychiatry and Neurochemistry, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Keiko Funa
- grid.8761.80000 0000 9919 9582Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden ,grid.1649.a000000009445082XOncology Laboratory, Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ludwig Aigner
- grid.21604.310000 0004 0523 5263Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Eric Hanse
- grid.8761.80000 0000 9919 9582Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Sebastian Illes
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
7
|
Pansri P, Phanthong P, Suthprasertporn N, Kitiyanant Y, Tubsuwan A, Dinnyes A, Kobolak J, Kitiyanant N. Brain-derived neurotrophic factor increases cell number of neural progenitor cells derived from human induced pluripotent stem cells. PeerJ 2021; 9:e11388. [PMID: 34026357 PMCID: PMC8123227 DOI: 10.7717/peerj.11388] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 04/10/2021] [Indexed: 12/23/2022] Open
Abstract
Background Several pieces of evidence from in vitro studies showed that brain-derived neurotrophic factor (BDNF) promotes proliferation and differentiation of neural stem/progenitor cells (NSCs) into neurons. Moreover, the JAK2 pathway was proposed to be associated with mouse NSC proliferation. BDNF could activate the STAT-3 pathway and induce proliferation in mouse NSCs. However, its effects on proliferation are not fully understood and JAK/STAT pathway was proposed to play a role in this activity. Methods In the present study, the effects of BDNF on cell proliferation and neurite outgrowth of Alzheimer’s disease (AD) induced pluripotent stem cells (iPSCs)-derived human neural progenitor cells (hNPCs) were examined. Moreover, a specific signal transduction pathway important in cell proliferation was investigated using a JAK2 inhibitor (AG490) to clarify the role of that pathway. Results The proliferative effect of BDNF was remarkably observed as an increase in Ki-67 positive cells. The cell number of hNPCs was significantly increased after BDNF treatment represented by cellular metabolic activity of the cells measured by MTT assay. This noticeable effect was statistically shown at 20 ng/ml of BDNF treatment. BDNF, however, did not promote neurite outgrowth but increased neuronal cell number. It was found that AG490 suppressed hNPCs proliferation. However, this inhibitor partially decreased BDNF-induced hNPCs proliferation. These results demonstrated the potential role of BDNF for the amelioration of AD through the increase of AD-derived hNPCs number.
Collapse
Affiliation(s)
- Panetha Pansri
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand.,BioTalentum Ltd., Gödöllö, Hungary
| | - Phetcharat Phanthong
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Nopparat Suthprasertporn
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Yindee Kitiyanant
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Alisa Tubsuwan
- Molecular Medical Biosciences Cluster, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Andras Dinnyes
- BioTalentum Ltd., Gödöllö, Hungary.,HCEMM-USZ StemCell Research Group, University of Szeged, Szeged, Hungary.,Department of Physiology and Animal Health, Hungarian University of Agriculture and Life Sciences, Gödöllö, Hungary
| | | | - Narisorn Kitiyanant
- Molecular Medical Biosciences Cluster, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| |
Collapse
|
8
|
MacMullen C, Davis RL. High-Throughput Phenotypic Assay for Compounds That Influence Mitochondrial Health Using iPSC-Derived Human Neurons. SLAS DISCOVERY 2021; 26:811-822. [PMID: 33724077 DOI: 10.1177/24725552211000671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
There is a critical need to develop high-throughput assays to identify compounds that offer therapy for individuals suffering from neurodegenerative diseases. Most brain disorders, including neurodegenerative diseases, share the common neuropathology of mitochondria dysfunction, which can lead to apoptosis of neurons, overproduction of reactive oxygen species (ROS), and other cellular neuropathologies characteristic of these diseases. Human induced pluripotent stem cells (iPSCs) with a stable genomic insertion of the neurogenin-2 transcription factor under the control of the TetOn promoter can be differentiated into excitatory human neurons (i3Neurons) within 3 days of exposure to doxycycline. These neurons have been used to develop and validate a live-cell assay for parameters of mitochondrial dynamics and function using two compounds known to promote mitochondrial elongation in mouse neurons, 4-hydroxychalcone and 2,4-dihyrdroxychalcone. The assay involves plating the neurons in 384-well microtiter plates, treating them with known or unknown substances, and then capturing morphological information for the neuronal mitochondria using a lentivirus vector to express a mitochondrial-targeted fluorescence reporter. The i3Neuron cultures exposed to these two compounds for 24 h exhibit significantly decreased circularity and significantly increased length compared to controls, two morphological parameters correlated with increased mitochondrial health. The assay is rapid, with results obtained after a one-week-long i3Neuron culture or one month if neurons are co-cultured with astrocytes. This live-cell, mitochondrial phenotypic assay can be used for high-throughput screening or as an orthogonal assay for compounds obtained via other high-throughput screening campaigns.
Collapse
Affiliation(s)
- Courtney MacMullen
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Ronald L Davis
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| |
Collapse
|
9
|
Zhang Y, Xie X, Hu J, Afreen KS, Zhang CL, Zhuge Q, Yang J. Prospects of Directly Reprogrammed Adult Human Neurons for Neurodegenerative Disease Modeling and Drug Discovery: iN vs. iPSCs Models. Front Neurosci 2020; 14:546484. [PMID: 33328842 PMCID: PMC7710799 DOI: 10.3389/fnins.2020.546484] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
A reliable disease model is critical to the study of specific disease mechanisms as well as for the discovery and development of new drugs. Despite providing crucial insights into the mechanisms of neurodegenerative diseases, translation of this information to develop therapeutics in clinical trials have been unsuccessful. Reprogramming technology to convert adult somatic cells to induced Pluripotent Stem Cells (iPSCs) or directly reprogramming adult somatic cells to induced Neurons (iN), has allowed for the creation of better models to understand the molecular mechanisms and design of new drugs. In recent times, iPSC technology has been commonly used for modeling neurodegenerative diseases and drug discovery. However, several technological challenges have limited the application of iN. As evidence suggests, iN for the modeling of neurodegenerative disorders is advantageous compared to those derived from iPSCs. In this review, we will compare iPSCs and iN models for neurodegenerative diseases and their potential applications in the future.
Collapse
Affiliation(s)
- Ying Zhang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinyang Xie
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,International Department of The Affiliated High School of South China Normal University (HFI), Guangzhou, China
| | - Jiangnan Hu
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Kazi Sabrina Afreen
- Department of Microbiology & Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Chun-Li Zhang
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianjing Yang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
Barbuti P, Antony P, Santos B, Massart F, Cruciani G, Dording C, Arias J, Schwamborn J, Krüger R. Using High-Content Screening to Generate Single-Cell Gene-Corrected Patient-Derived iPS Clones Reveals Excess Alpha-Synuclein with Familial Parkinson's Disease Point Mutation A30P. Cells 2020; 9:cells9092065. [PMID: 32927687 PMCID: PMC7564375 DOI: 10.3390/cells9092065] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
The generation of isogenic induced pluripotent stem cell (iPSC) lines using CRISPR-Cas9 technology is a technically challenging, time-consuming process with variable efficiency. Here we use fluorescence-activated cell sorting (FACS) to sort biallelic CRISPR-Cas9 edited single-cell iPSC clones into high-throughput 96-well microtiter plates. We used high-content screening (HCS) technology and generated an in-house developed algorithm to select the correctly edited isogenic clones for continued expansion and validation. In our model we have gene-corrected the iPSCs of a Parkinson’s disease (PD) patient carrying the autosomal dominantly inherited heterozygous c.88G>C mutation in the SNCA gene, which leads to the pathogenic p.A30P form of the alpha-synuclein protein. Undertaking a PCR restriction-digest mediated clonal selection strategy prior to sequencing, we were able to post-sort validate each isogenic clone using a quadruple screening strategy prior to generating footprint-free isogenic iPSC lines, retaining a normal molecular karyotype, pluripotency and three germ-layer differentiation potential. Directed differentiation into midbrain dopaminergic neurons revealed that SNCA expression is reduced in the gene-corrected clones, which was validated by a reduction at the alpha-synuclein protein level. The generation of single-cell isogenic clones facilitates new insights in the role of alpha-synuclein in PD and furthermore is applicable across patient-derived disease models.
Collapse
Affiliation(s)
- Peter Barbuti
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Luxembourg, Luxembourg; (P.A.); (B.S.); (F.M.); (G.C.); (C.D.)
- Transversal Translational Medicine, Luxembourg Institute of Health, L-1445 Luxembourg, Luxembourg
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Correspondence: (P.B.); (R.K.); Tel.: +352-26970-967 (R.K.)
| | - Paul Antony
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Luxembourg, Luxembourg; (P.A.); (B.S.); (F.M.); (G.C.); (C.D.)
| | - Bruno Santos
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Luxembourg, Luxembourg; (P.A.); (B.S.); (F.M.); (G.C.); (C.D.)
- Transversal Translational Medicine, Luxembourg Institute of Health, L-1445 Luxembourg, Luxembourg
| | - François Massart
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Luxembourg, Luxembourg; (P.A.); (B.S.); (F.M.); (G.C.); (C.D.)
| | - Gérald Cruciani
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Luxembourg, Luxembourg; (P.A.); (B.S.); (F.M.); (G.C.); (C.D.)
| | - Claire Dording
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Luxembourg, Luxembourg; (P.A.); (B.S.); (F.M.); (G.C.); (C.D.)
- Transversal Translational Medicine, Luxembourg Institute of Health, L-1445 Luxembourg, Luxembourg
| | - Jonathan Arias
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Luxembourg, Luxembourg; (J.A.); (J.S.)
- Department of Biosciences and Nutrition, Neo, Karolinska Institutet, SE-141 83 Huddinge, Sweden
| | - Jens Schwamborn
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Luxembourg, Luxembourg; (J.A.); (J.S.)
| | - Rejko Krüger
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Luxembourg, Luxembourg; (P.A.); (B.S.); (F.M.); (G.C.); (C.D.)
- Transversal Translational Medicine, Luxembourg Institute of Health, L-1445 Luxembourg, Luxembourg
- Department of Neurodegenerative diseases, Hertie Institute for Clinical Brain Research, University Clinics Tübingen, 72076 Tübingen, Germany
- Parkinson Research Clinic, Centre Hospitalier de Luxembourg (CHL), L-1210 Luxembourg, Luxembourg
- Correspondence: (P.B.); (R.K.); Tel.: +352-26970-967 (R.K.)
| |
Collapse
|
11
|
Hamamura K, Hotta H, Murakumo Y, Shibuya H, Kondo Y, Furukawa K. SSEA-3 and 4 are not essential for the induction or properties of mouse iPS cells. J Oral Sci 2020; 62:393-396. [PMID: 32684574 DOI: 10.2334/josnusd.19-0513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Stage-specific embryonic antigens (SSEA-1, 3, and 4) are carbohydrate antigens that have been used as markers of embryonic stem (ES) cells. However, the roles of these antigens in the establishment and maintenance of stemness of ES and induced pluripotent stem (iPS) cells are still poorly understood. This study investigated the biological and functional significance of globo-series glycolipids such as SSEA-3 and 4 in mouse iPS cells induced from tail-tip fibroblasts (TTFs) of α1,4Gal-T-knockout mice (lacking SSEA-3 and 4). These iPS cells were induced by retroviral transduction of four factors (Oct3/4, Sox2, Klf4, and c-Myc) into TTFs, and colonies were picked up. Morphologically, the colonies resembled ES cells and were positive for alkaline phosphatase and ES cell markers. Furthermore, in vitro-differentiated induction experiments after embryoid body formation revealed that some colonies derived from α1, 4Gal-T knockout mice were able to differentiate into three germ layers. Three germ layers were also observed in teratomas from iPS cells derived from α1,4Gal-T-knockout mice. These results suggest that SSEA-3 and 4 are not essential, at least for the establishment and maintenance of stemness of mouse iPS cells.
Collapse
Affiliation(s)
- Kazunori Hamamura
- Department of Pharmacology, School of Dentistry, Aichi Gakuin University
| | - Hiroshi Hotta
- Department of Biochemistry II, Nagoya University Graduate School of Medicine
| | | | - Hidenobu Shibuya
- Department of Biochemistry II, Nagoya University Graduate School of Medicine
| | - Yuji Kondo
- Department of Biochemistry II, Nagoya University Graduate School of Medicine
| | - Koichi Furukawa
- Department of Biochemistry II, Nagoya University Graduate School of Medicine.,Department of Biomedical Sciences, Chubu University College of Life and Health Sciences
| |
Collapse
|
12
|
Silva MC, Haggarty SJ. Human pluripotent stem cell-derived models and drug screening in CNS precision medicine. Ann N Y Acad Sci 2020; 1471:18-56. [PMID: 30875083 PMCID: PMC8193821 DOI: 10.1111/nyas.14012] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/02/2019] [Accepted: 01/07/2019] [Indexed: 12/12/2022]
Abstract
Development of effective therapeutics for neurological disorders has historically been challenging partly because of lack of accurate model systems in which to investigate disease etiology and test new therapeutics at the preclinical stage. Human stem cells, particularly patient-derived induced pluripotent stem cells (iPSCs) upon differentiation, have the ability to recapitulate aspects of disease pathophysiology and are increasingly recognized as robust scalable systems for drug discovery. We review advances in deriving cellular models of human central nervous system (CNS) disorders using iPSCs along with strategies for investigating disease-relevant phenotypes, translatable biomarkers, and therapeutic targets. Given their potential to identify novel therapeutic targets and leads, we focus on phenotype-based, small-molecule screens employing human stem cell-derived models. Integrated efforts to assemble patient iPSC-derived cell models with deeply annotated clinicopathological data, along with molecular and drug-response signatures, may aid in the stratification of patients, diagnostics, and clinical trial success, shifting translational science and precision medicine approaches. A number of remaining challenges, including the optimization of cost-effective, large-scale culture of iPSC-derived cell types, incorporation of aging into neuronal models, as well as robustness and automation of phenotypic assays to support quantitative drug efficacy, toxicity, and metabolism testing workflows, are covered. Continued advancement of the field is expected to help fully humanize the process of CNS drug discovery.
Collapse
Affiliation(s)
- M. Catarina Silva
- Chemical Neurobiology Laboratory, Departments of Neurology and Psychiatry, Massachusetts General Hospital, Center for Genomic Medicine, Harvard Medical School, Boston MA, USA
| | - Stephen J. Haggarty
- Chemical Neurobiology Laboratory, Departments of Neurology and Psychiatry, Massachusetts General Hospital, Center for Genomic Medicine, Harvard Medical School, Boston MA, USA
| |
Collapse
|
13
|
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
|
14
|
Auvin S, Avbersek A, Bast T, Chiron C, Guerrini R, Kaminski RM, Lagae L, Muglia P, Cross JH. Drug Development for Rare Paediatric Epilepsies: Current State and Future Directions. Drugs 2020; 79:1917-1935. [PMID: 31734883 DOI: 10.1007/s40265-019-01223-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Rare diseases provide a challenge in the evaluation of new therapies. However, orphan drug development is of increasing interest because of the legislation enabling facilitated support by regulatory agencies through scientific advice, and the protection of the molecules with orphan designation. In the landscape of the rare epilepsies, very few syndromes, namely Dravet syndrome, Lennox-Gastaut syndrome and West syndrome, have been subject to orphan drug development. Despite orphan designations for rare epilepsies having dramatically increased in the past 10 years, the number of approved drugs remains limited and restricted to a handful of epilepsy syndromes. In this paper, we describe the current state of orphan drug development for rare epilepsies. We identified a large number of compounds currently under investigation, but mostly in the same rare epilepsy syndromes as in the past. A rationale for further development in rare epilepsies could be based on the match between the drug mechanisms of action and the knowledge of the causative gene mutation or by evidence from animal models. In case of the absence of strong pathophysiological hypotheses, exploratory/basket clinical studies could be helpful to identify a subpopulation that may benefit from the new drug. We provide some suggestions for future improvements in orphan drug development such as promoting paediatric drug investigations, better evaluation of the incidence and the prevalence, together with the natural history data, and the development of new primary outcomes.
Collapse
Affiliation(s)
- Stéphane Auvin
- PROTECT, INSERM U1141, Université de Paris, Paris, France. .,Service de Neurologie Pédiatrique, AP-HP, Hôpital Universitaire Robert-Debré, 48, Boulevard Sérurier, 75935, Paris Cedex 19, France.
| | | | - Thomas Bast
- The Kork Epilepsy Center, Kehl-Kork, Germany.,Medical Faculty of the University of Freiburg, Freiburg, Germany
| | - Catherine Chiron
- PROTECT, INSERM U1141, Université de Paris, Paris, France.,Service de Neurologie Pédiatrique, AP-HP, Hôpital Necker-Enfanst Malades, Paris, France
| | - Renzo Guerrini
- Neuroscience Department, Children's Hospital Anna Meyer-University of Florence, Florence, Italy
| | - Rafal M Kaminski
- UCB Pharma, Braine-l'Alleud, Belgium.,Roche Pharma Research and Early Development (pRED), Roche Innovation Center, Basel, Switzerland
| | - Lieven Lagae
- Department Development and Regeneration, Section Paediatric Neurology, University Hospitals, University of Leuven, Leuven, Belgium
| | | | - J Helen Cross
- UCL NIHR BRC Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
15
|
Kinnear C, Agrawal R, Loo C, Pahnke A, Rodrigues DC, Thompson T, Akinrinade O, Ahadian S, Keeley F, Radisic M, Mital S, Ellis J. Everolimus Rescues the Phenotype of Elastin Insufficiency in Patient Induced Pluripotent Stem Cell-Derived Vascular Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2020; 40:1325-1339. [PMID: 32212852 PMCID: PMC7176340 DOI: 10.1161/atvbaha.119.313936] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Supplemental Digital Content is available in the text. Objective: Elastin gene deletion or mutation leads to arterial stenoses due to vascular smooth muscle cell (SMC) proliferation. Human induced pluripotent stem cells–derived SMCs can model the elastin insufficiency phenotype in vitro but show only partial rescue with rapamycin. Our objective was to identify drug candidates with superior efficacy in rescuing the SMC phenotype in elastin insufficiency patients. Approach and Results: SMCs generated from induced pluripotent stem cells from 5 elastin insufficiency patients with severe recurrent vascular stenoses (3 Williams syndrome and 2 elastin mutations) were phenotypically immature, hyperproliferative, poorly responsive to endothelin, and exerted reduced tension in 3-dimensional smooth muscle biowires. Elastin mRNA and protein were reduced in SMCs from patients compared to healthy control SMCs. Fourteen drug candidates were tested on patient SMCs. Of the mammalian target of rapamycin inhibitors studied, everolimus restored differentiation, rescued proliferation, and improved endothelin-induced calcium flux in all patient SMCs except one Williams syndrome. Of the calcium channel blockers, verapamil increased SMC differentiation and reduced proliferation in Williams syndrome patient cells but not in elastin mutation patients and had no effect on endothelin response. Combination treatment with everolimus and verapamil was not superior to everolimus alone. Other drug candidates had limited efficacy. Conclusions: Everolimus caused the most consistent improvement in SMC differentiation, proliferation and in SMC function in patients with both syndromic and nonsyndromic elastin insufficiency, and offers the best candidate for drug repurposing for treatment of elastin insufficiency associated vasculopathy.
Collapse
Affiliation(s)
- Caroline Kinnear
- From the Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada (C.K., R.A., O.A., S.M.)
| | - Rahul Agrawal
- From the Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada (C.K., R.A., O.A., S.M.)
| | - Caitlin Loo
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada (C.L., D.C.R., T.T., J.E.).,Department of Molecular Genetics (C.L., J.E.), University of Toronto, Ontario, Canada
| | - Aric Pahnke
- Institute of Biomaterials and Biomedical Engineering (A.P., S.A., M.R.), University of Toronto, Ontario, Canada.,Department of Chemical Engineering and Applied Chemistry (A.P., S.A., M.R.), University of Toronto, Ontario, Canada
| | - Deivid Carvalho Rodrigues
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada (C.L., D.C.R., T.T., J.E.)
| | - Tadeo Thompson
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada (C.L., D.C.R., T.T., J.E.)
| | - Oyediran Akinrinade
- From the Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada (C.K., R.A., O.A., S.M.)
| | - Samad Ahadian
- Institute of Biomaterials and Biomedical Engineering (A.P., S.A., M.R.), University of Toronto, Ontario, Canada.,Department of Chemical Engineering and Applied Chemistry (A.P., S.A., M.R.), University of Toronto, Ontario, Canada
| | - Fred Keeley
- Department of Biochemistry (F.K.), University of Toronto, Ontario, Canada.,Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada (F.K.)
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering (A.P., S.A., M.R.), University of Toronto, Ontario, Canada.,Department of Chemical Engineering and Applied Chemistry (A.P., S.A., M.R.), University of Toronto, Ontario, Canada
| | - Seema Mital
- From the Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada (C.K., R.A., O.A., S.M.).,Department of Pediatrics, The Hospital for Sick Children (S.M.), University of Toronto, Ontario, Canada
| | - James Ellis
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada (C.L., D.C.R., T.T., J.E.).,Department of Molecular Genetics (C.L., J.E.), University of Toronto, Ontario, Canada
| |
Collapse
|
16
|
Generating Human iPSC-Derived Astrocytes with Chemically Defined Medium for In Vitro Disease Modeling. Methods Mol Biol 2020; 1994:31-39. [PMID: 31124102 DOI: 10.1007/978-1-4939-9477-9_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
To better understand and model neurological, in particular neurodegenerative diseases, human induced pluripotent stem cells (hiPSCs) offer a great source for generation of neural cells. We provide a protocol for the differentiation of hiPSc-derived astrocytes in vitro. This protocol not only is chemically defined, that is, it does not use serum, but also allows for the expansion of astrocyte progenitor cells and mature astrocytes. Large batches of hiPSc-derived astrocytes can be stored and used for defined in vitro disease models.
Collapse
|
17
|
Takada H, Kaieda A, Tawada M, Nagino T, Sasa K, Oikawa T, Oki A, Sameshima T, Miyamoto K, Miyamoto M, Kokubu Y, Tozawa R, Sakurai H, Saito B. Identification of 2,6-Disubstituted 3 H-Imidazo[4,5- b]pyridines as Therapeutic Agents for Dysferlinopathies through Phenotypic Screening on Patient-Derived Induced Pluripotent Stem Cells. J Med Chem 2019; 62:9175-9187. [PMID: 31550153 DOI: 10.1021/acs.jmedchem.9b01100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dysferlinopathies, which are muscular diseases caused by mutations in the dysferlin gene, remain serious medical problems due to the lack of therapeutic agents. Herein, we report the design, synthesis, and structure-activity relationships of a 2,6-disubstituted 3H-imidazo[4,5-b]pyridine series, which was identified from the phenotypic screening of chemicals that increase the level of dysferlin in myocytes differentiated from patient-derived induced pluripotent stem cells (iPSCs). Optimization studies with cell-based phenotypic assay led to the identification of a highly potent compound, 19, with dysferlin elevation effects at double-digit nanomolar concentrations. In addition, the molecular target of our chemical series was identified as tubulin, through a tubulin polymerization assay and a competitive binding assay using a photoaffinity labeling probe.
Collapse
Affiliation(s)
- Hiroyuki Takada
- Research , Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Akira Kaieda
- Research , Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Michiko Tawada
- Research , Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Tomoko Nagino
- T-CiRA Discovery , Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Katsunori Sasa
- T-CiRA Discovery , Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Tatsuo Oikawa
- T-CiRA Discovery , Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Akiko Oki
- Research , Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Tomoya Sameshima
- Research , Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Kazumasa Miyamoto
- Research , Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Makoto Miyamoto
- Research , Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Yuko Kokubu
- Center for iPS Cell Research and Application , Kyoto University , 53 Kawahara-cho , Shogoin, Sakyo-ku, Kyoto 606-8507 , Japan
| | - Ryuichi Tozawa
- T-CiRA Discovery , Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application , Kyoto University , 53 Kawahara-cho , Shogoin, Sakyo-ku, Kyoto 606-8507 , Japan
| | - Bunnai Saito
- Research , Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome , Fujisawa , Kanagawa 251-8555 , Japan
| |
Collapse
|
18
|
Friese A, Ursu A, Hochheimer A, Schöler HR, Waldmann H, Bruder JM. The Convergence of Stem Cell Technologies and Phenotypic Drug Discovery. Cell Chem Biol 2019; 26:1050-1066. [PMID: 31231030 DOI: 10.1016/j.chembiol.2019.05.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 04/04/2019] [Accepted: 05/20/2019] [Indexed: 02/06/2023]
Abstract
Recent advances in induced pluripotent stem cell technologies and phenotypic screening shape the future of bioactive small-molecule discovery. In this review we analyze the impact of small-molecule phenotypic screens on drug discovery as well as on the investigation of human development and disease biology. We further examine the role of 3D spheroid/organoid structures, microfluidic systems, and miniaturized on-a-chip systems for future discovery strategies. In highlighting representative examples, we analyze how recent achievements can translate into future therapies. Finally, we discuss remaining challenges that need to be overcome for the adaptation of the next generation of screening approaches.
Collapse
Affiliation(s)
- Alexandra Friese
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| | - Andrei Ursu
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany; Department of Chemistry, The Scripps Research Institute, Jupiter, FL 33458, USA; Faculty of Chemistry and Chemical Biology, TU Dortmund, Otto-Hahn-Str. 4a, 44227 Dortmund, Germany
| | - Andreas Hochheimer
- ISAR Bioscience GmbH, Institute for Stem Cell & Applied Regenerative Medicine Research, 82152 Planegg, Germany
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany; Medical Faculty, University of Münster, Domagkstrasse 3, 48149 Münster, Germany.
| | - Herbert Waldmann
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany; Faculty of Chemistry and Chemical Biology, TU Dortmund, Otto-Hahn-Str. 4a, 44227 Dortmund, Germany.
| | - Jan M Bruder
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany.
| |
Collapse
|
19
|
Izsak J, Seth H, Andersson M, Vizlin-Hodzic D, Theiss S, Hanse E, Ågren H, Funa K, Illes S. Robust Generation of Person-Specific, Synchronously Active Neuronal Networks Using Purely Isogenic Human iPSC-3D Neural Aggregate Cultures. Front Neurosci 2019; 13:351. [PMID: 31068774 PMCID: PMC6491690 DOI: 10.3389/fnins.2019.00351] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/27/2019] [Indexed: 12/13/2022] Open
Abstract
Reproducibly generating human induced pluripotent stem cell-based functional neuronal circuits, solely obtained from single individuals, poses particular challenges to achieve personalized and patient specific functional neuronal in vitro models. A hallmark of functional neuronal assemblies, synchronous neuronal activity, can be non-invasively studied by microelectrode array (MEA) technology, reliably capturing physiological and pathophysiological aspects of human brain function. In our here presented manuscript, we demonstrate a procedure to generate 3D neural aggregates comprising astrocytes, oligodendroglial cells, and neurons obtained from the same human tissue sample. Moreover, we demonstrate the robust ability of those neurons to create a highly synchronously active neuronal network within 3 weeks in vitro, without additionally applied astrocytes. The fusion of MEA-technology with functional neuronal circuits solely obtained from one individual's cells represent isogenic person-specific human neuronal sensor chips that pave the way for specific personalized in vitro neuronal networks as well as neurological and neuropsychiatric disease modeling.
Collapse
Affiliation(s)
- Julia Izsak
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Henrik Seth
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Mats Andersson
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Dzeneta Vizlin-Hodzic
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden.,Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Stephan Theiss
- Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.,Result Medical GmbH, Düsseldorf, Germany
| | - Eric Hanse
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Hans Ågren
- Institute of Neuroscience and Physiology, Section of Psychiatry and Neurochemistry, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Keiko Funa
- Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden.,Oncology Laboratory, Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Sebastian Illes
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| |
Collapse
|
20
|
The power of combining phenotypic and target-focused drug discovery. Drug Discov Today 2019; 24:526-532. [DOI: 10.1016/j.drudis.2018.10.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/10/2018] [Accepted: 10/16/2018] [Indexed: 01/09/2023]
|
21
|
Bluhmki T, Traub S, Schruf E, Garnett J, Gantner F, Bischoff D, Heilker R. Differentiation of hiPS Cells into Definitive Endoderm for High-Throughput Screening. Methods Mol Biol 2019; 1994:101-115. [PMID: 31124108 DOI: 10.1007/978-1-4939-9477-9_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
In drug discovery, there is an increasing demand for more physiological in vitro models that recapitulate the disease situation in patients. Human induced pluripotent stem (hiPS) cell-derived model cells could serve this purpose. To date, several directed differentiation approaches have been described to generate definitive endoderm (DE) from hiPS cells, but protocols suitable for drug development and high-throughput screening (HTS) have not been reported yet. In this work, a large-scale expansion of hiPS cells for high-throughput adaption is presented and an optimized stepwise differentiation of hiPS cells into DE cells is described. The produced DE cells were demonstrated to express classical DE markers on the gene expression and protein level. The here described DE cells are multipotent progenitors and act as starting points for a broad spectrum of endodermal model cells in HTS and other areas of drug discovery.
Collapse
Affiliation(s)
- Teresa Bluhmki
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Eva Schruf
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - James Garnett
- Immunology & Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Florian Gantner
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Daniel Bischoff
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Ralf Heilker
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany.
| |
Collapse
|
22
|
Abstract
Human induced pluripotent stem (hiPS) cell-derived neurons promise to provide better model cells for drug discovery in the context of neurodegenerative and neuropsychiatric diseases. The neuronal differentiation protocol described encompasses a cellular amplification phase for hiPS-derived neural progenitor (NP) cells. Thus, the combination of growth factor-driven expansion and inhibition of notch (GRINCH) enabled the scalable production of neurons in sufficient numbers to meet the immense material needs of a high-throughput screening (HTS) campaign. These GRINCH cells matured in 384-well microplates display neuronal markers and electrophysiological activity. The differentiation protocol was applicable to various human hiPS cell clones. In a finding and profiling campaign for modulators of the tropomyosin receptor kinase B (TrkB), the GRINCH neurons were shown to be suitable for measuring the phosphorylation and downstream signaling of the endogenously expressed TrkB. The employed techniques in the amplified luminescent proximity homogeneous assay (Alpha) and the high-throughput reverse transcription polymerase chain reaction (RT-PCR) format are transferable to other pharmaceutical drug targets. Together with the GRINCH neurons, these detection technologies open new experimental routes with tremendous potential for early drug discovery.
Collapse
Affiliation(s)
| | - Ralf Heilker
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany.
| |
Collapse
|
23
|
Guo Y, Yu M, Jing N, Zhang S. Production of soluble bioactive mouse leukemia inhibitory factor from Escherichia coli using MBP tag. Protein Expr Purif 2018; 150:86-91. [PMID: 29758321 DOI: 10.1016/j.pep.2018.05.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 05/04/2018] [Accepted: 05/10/2018] [Indexed: 01/19/2023]
Abstract
Embryonic stem cells and induced pluripotent stem cells depend on one of cytokines called leukemia inhibitory factor (LIF) to retain their undifferentiated state and pluripotency. Nevertheless, further progresses of stem cell scientific investigation and its possible application are limited owing to the expense of commercial LIF. Here we introduced a simple, practical and high level expression of MBP-mouse LIF through Escherichia coli system which was bioactive. The mLIF cDNA was inserted into vector of pMAL-C2X in order to generate N-terminal MBP-mLIF recombinant proteins in the cytoplasm of Escherichia coli. MBP-mLIF as a soluble form was expressed. One-step purification through gravitational affinity chromatography was accomplished to acquire high purity (>92%) MBP-mLIF. The MBP-mLIF products specifically suppressed the growth of M1cells in a dose-dependent pattern. MBP-mLIF also was proved the ability to maintain the pluripotency of iPSCs. These outcomes revealed that the N-end MBP tags of the MBP-mLIF did not obstruct mLIF bioactivity. This method to generate recombinant MBP-mLIF is a simple, practical, economical and user-friendly protocol.
Collapse
Affiliation(s)
- Yanan Guo
- School of Life Sciences, Zhengzhou University, No.100 Science Avenue, Zhengzhou, Henan, 450001, China.
| | - Miao Yu
- School of Life Sciences, Zhengzhou University, No.100 Science Avenue, Zhengzhou, Henan, 450001, China
| | - Na Jing
- School of Life Sciences, Zhengzhou University, No.100 Science Avenue, Zhengzhou, Henan, 450001, China
| | - Shoutao Zhang
- School of Life Sciences, Zhengzhou University, No.100 Science Avenue, Zhengzhou, Henan, 450001, China
| |
Collapse
|
24
|
Matsushiro Y, Kato-Negishi M, Onoe H. Differentiation of 3D-shape-controlled mouse neural stem cell to neural tissues in closed agarose microchambers. Biotechnol Bioeng 2018; 115:1614-1623. [DOI: 10.1002/bit.26559] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/14/2018] [Accepted: 02/05/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Yuki Matsushiro
- Department of Mechanical Engineering; Faculty of Science and Technology; Keio University; Kohoku-ku Yokohama Japan
| | - Midori Kato-Negishi
- Institute of Industrial Science; The University of Tokyo; Meguro-ku Tokyo Japan
- Laboratory of Bio-Analytical Chemistry; Research Institute of Pharmaceutical Sciences; Faculty of Pharmacy; Musashino University; Nishitokyo-shi Japan
| | - Hiroaki Onoe
- Department of Mechanical Engineering; Faculty of Science and Technology; Keio University; Kohoku-ku Yokohama Japan
| |
Collapse
|
25
|
Sherman SP, Bang AG. High-throughput screen for compounds that modulate neurite growth of human induced pluripotent stem cell-derived neurons. Dis Model Mech 2018; 11:dmm.031906. [PMID: 29361516 PMCID: PMC5894944 DOI: 10.1242/dmm.031906] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/29/2017] [Indexed: 01/01/2023] Open
Abstract
Development of technology platforms to perform compound screens of human induced pluripotent stem cell (hiPSC)-derived neurons with relatively high throughput is essential to realize their potential for drug discovery. Here, we demonstrate the feasibility of high-throughput screening of hiPSC-derived neurons using a high-content, image-based approach focused on neurite growth, a process that is fundamental to formation of neural networks and nerve regeneration. From a collection of 4421 bioactive small molecules, we identified 108 hit compounds, including 37 approved drugs, that target molecules or pathways known to regulate neurite growth, as well as those not previously associated with this process. These data provide evidence that many pathways and targets known to play roles in neurite growth have similar activities in hiPSC-derived neurons that can be identified in an unbiased phenotypic screen. The data also suggest that hiPSC-derived neurons provide a useful system to study the mechanisms of action and off-target activities of the approved drugs identified as hits, leading to a better understanding of their clinical efficacy and toxicity, especially in the context of specific human genetic backgrounds. Finally, the hit set we report constitutes a sublibrary of approved drugs and tool compounds that modulate neurites. This sublibrary will be invaluable for phenotypic analyses and interrogation of hiPSC-based disease models as probes for defining phenotypic differences and cellular vulnerabilities in patient versus control cells, as well as for investigations of the molecular mechanisms underlying human neurite growth in development and maintenance of neuronal networks, and nerve regeneration. Summary: High-throughput, small molecule screening of hiPSC-derived neurons using a high-content, image-based approach focused on neurite growth identified hit compounds, including approved drugs, which target molecules or pathways known to regulate neurite growth.
Collapse
Affiliation(s)
- Sean P Sherman
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute La Jolla, CA 92037, USA
| | - Anne G Bang
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute La Jolla, CA 92037, USA
| |
Collapse
|
26
|
Liang N, Trujillo CA, Negraes PD, Muotri AR, Lameu C, Ulrich H. Stem cell contributions to neurological disease modeling and personalized medicine. Prog Neuropsychopharmacol Biol Psychiatry 2018; 80:54-62. [PMID: 28576415 DOI: 10.1016/j.pnpbp.2017.05.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/27/2017] [Accepted: 05/30/2017] [Indexed: 01/16/2023]
Abstract
Human induced pluripotent stem cells (iPSCs) represent a revolutionary tool for disease modeling and drug discovery. The generation of tissue-relevant cell types exhibiting a patient's genetic and molecular background offers the ability to develop individual and effective therapies. In this review, we present some major achievements in the neuroscience field using iPSCs and discuss promising perspectives in personalized medicine. In addition to disease modeling, the understanding of the cellular and molecular basis of neurological disorders is explored, including the discovery of new targets and potential drugs. Ultimately, we highlight how iPSC technology, together with genome editing approaches, may bring a deep impact on pre-clinical trials by reducing costs and increasing the success of treatments in a personalized fashion.
Collapse
Affiliation(s)
- Nicholas Liang
- University of California San Diego, School of Medicine, Department of Pediatrics/Rady Children's Hospital San Diego, Stem Cell Program, La Jolla, CA 92093, USA
| | - Cleber A Trujillo
- University of California San Diego, School of Medicine, Department of Pediatrics/Rady Children's Hospital San Diego, Stem Cell Program, La Jolla, CA 92093, USA
| | - Priscilla D Negraes
- University of California San Diego, School of Medicine, Department of Pediatrics/Rady Children's Hospital San Diego, Stem Cell Program, La Jolla, CA 92093, USA
| | - Alysson R Muotri
- University of California San Diego, School of Medicine, Department of Pediatrics/Rady Children's Hospital San Diego, Stem Cell Program, La Jolla, CA 92093, USA
| | - Claudiana Lameu
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil.
| |
Collapse
|
27
|
Moffat JG, Vincent F, Lee JA, Eder J, Prunotto M. Opportunities and challenges in phenotypic drug discovery: an industry perspective. Nat Rev Drug Discov 2017; 16:531-543. [PMID: 28685762 DOI: 10.1038/nrd.2017.111] [Citation(s) in RCA: 570] [Impact Index Per Article: 71.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Phenotypic drug discovery (PDD) approaches do not rely on knowledge of the identity of a specific drug target or a hypothesis about its role in disease, in contrast to the target-based strategies that have been widely used in the pharmaceutical industry in the past three decades. However, in recent years, there has been a resurgence in interest in PDD approaches based on their potential to address the incompletely understood complexity of diseases and their promise of delivering first-in-class drugs, as well as major advances in the tools for cell-based phenotypic screening. Nevertheless, PDD approaches also have considerable challenges, such as hit validation and target deconvolution. This article focuses on the lessons learned by researchers engaged in PDD in the pharmaceutical industry and considers the impact of 'omics' knowledge in defining a cellular disease phenotype in the era of precision medicine, introducing the concept of a chain of translatability. We particularly aim to identify features and areas in which PDD can best deliver value to drug discovery portfolios and can contribute to the identification and the development of novel medicines, and to illustrate the challenges and uncertainties that are associated with PDD in order to help set realistic expectations with regard to its benefits and costs.
Collapse
Affiliation(s)
- John G Moffat
- Biochemical &Cellular Pharmacology, Genentech, South San Francisco, California 94080, USA
| | - Fabien Vincent
- Discovery Sciences, Primary Pharmacology Group, Pfizer, Groton, Connecticut 06340, USA
| | - Jonathan A Lee
- Department of Quantitative Biology, Eli Lilly and Company, Indianapolis, Indiana 46285, USA
| | - Jörg Eder
- Novartis Institutes for Biomedical Research, 4002 Basel, Switzerland
| | - Marco Prunotto
- Phenotype and Target ID, Chemical Biology, pRED, Roche, 4070 Basel, Switzerland. Present address: Office of Innovation, Immunology, Infectious Diseases &Ophthalmology (I2O), Roche Late Stage Development, 124 Grenzacherstrasse, 4070 Basel, Switzerland
| |
Collapse
|
28
|
Antonucci I, Crowley MG, Stuppia L. Amniotic fluid stem cell models: A tool for filling the gaps in knowledge for human genetic diseases. Brain Circ 2017; 3:167-174. [PMID: 30276320 PMCID: PMC6057697 DOI: 10.4103/bc.bc_23_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 09/06/2017] [Accepted: 09/12/2017] [Indexed: 12/21/2022] Open
Abstract
Induced pluripotent stem (iPS) cells have attracted attention in recent years as a model of human genetic diseases. Starting from the diseased somatic cells isolated from an affected patient, iPS cells can be created and subsequently differentiated into various cell types that can be used to gain a better understanding of the disease at a cellular and molecular level. There are limitations of iPS cell generation, however, due to low efficiency, high costs, and lengthy protocols. The use of amniotic fluid stem cells (AFS) presents a worthy alternative as a stem cell source for modeling of human genetic diseases. Prenatal identification of chromosomal or Mendelian diseases may require the collection of amniotic fluid which is not only useful for the sake of diagnosis but also from this, AFS cells can be isolated and cultured. Since AFS cells show some characteristics of pluripotency, having the capacity to differentiate into various cell types derived from all three germ layers in vitro, they are a well-suited model for investigations regarding alterations in the molecular biology of a cell due to a specific genetic disease. This readily accessible source of stem cells can replace the necessity for generating iPS cells. Here, we expand on the applicability and importance of AFS cells as a model for discovery in the field of human genetic disease research. This paper is a review article. Referred literature in this paper has been listed in the references section. The data sets supporting the conclusions of this article are available online by searching various databases, including PubMed. Some original points in this article come from the laboratory practice in our research center and the authors’ experiences.
Collapse
Affiliation(s)
- Ivana Antonucci
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, Annunzio University, Chieti-Pescara, Italy
| | - Marci G Crowley
- Center of Excellence for Aging and Brain Repair, University of South Florida, 12901, USA
| | - Liborio Stuppia
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, Annunzio University, Chieti-Pescara, Italy
| |
Collapse
|
29
|
Traub S, Stahl H, Rosenbrock H, Simon E, Florin L, Hospach L, Hörer S, Heilker R. Pharmaceutical Characterization of Tropomyosin Receptor Kinase B-Agonistic Antibodies on Human Induced Pluripotent Stem (hiPS) Cell-Derived Neurons. J Pharmacol Exp Ther 2017; 361:355-365. [PMID: 28351853 DOI: 10.1124/jpet.117.240184] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/23/2017] [Indexed: 03/08/2025] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a central modulator of neuronal development and synaptic plasticity in the central nervous system. This renders the BDNF-modulated tropomyosin receptor kinase B (TrkB) a promising drug target to treat synaptic dysfunctions. Using GRowth factor-driven expansion and INhibition of NotCH (GRINCH) during maturation, the so-called GRINCH neurons were derived from human-induced pluripotent stem cells. These GRINCH neurons were used as model cells for pharmacologic profiling of two TrkB-agonistic antibodies, hereafter referred to as AB2 and AB20 In next-generation sequencing studies, AB2 and AB20 stimulated transcriptional changes, which extensively overlapped with BDNF-driven transcriptional modulation. In regard to TrkB phosphorylation, both AB2 and AB20 were only about half as efficacious as BDNF; however, with respect to the TrkB downstream signaling, AB2 and AB20 displayed increased efficacy values, providing a stimulation at least comparable to BDNF in respect to VGF transcription, as well as of AKT and cAMP response element-binding protein phosphorylation. In a complex structure of the TrkB-d5 domain with AB20, determined by X-ray crystallography, the AB20 binding site was found to be allosteric in regard to the BDNF binding site, whereas AB2 was known to act orthosterically with BDNF. In agreement with this finding, AB2 and AB20 acted synergistically at greater concentrations to drive TrkB phosphorylation. Although TrkB downstream signaling declined faster after pulse stimulation with AB20 than with AB2, AB20 restimulated TrkB phosphorylation more efficiently than AB2. In conclusion, both antibodies displayed some limitations and some benefits in regard to future applications as therapeutic agents.
Collapse
Affiliation(s)
- Stefanie Traub
- Trenzyme GmbH, Konstanz (S.T.) Germany, Lead Identification and Optimization Support (L.H., S.H., R.H.), Immunological and Respiratory Diseases Research (H.S.), CNS Diseases Research (H.R.), and Target Discovery Research (E.S.), Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany; and Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (L.F.)
| | - Heiko Stahl
- Trenzyme GmbH, Konstanz (S.T.) Germany, Lead Identification and Optimization Support (L.H., S.H., R.H.), Immunological and Respiratory Diseases Research (H.S.), CNS Diseases Research (H.R.), and Target Discovery Research (E.S.), Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany; and Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (L.F.)
| | - Holger Rosenbrock
- Trenzyme GmbH, Konstanz (S.T.) Germany, Lead Identification and Optimization Support (L.H., S.H., R.H.), Immunological and Respiratory Diseases Research (H.S.), CNS Diseases Research (H.R.), and Target Discovery Research (E.S.), Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany; and Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (L.F.)
| | - Eric Simon
- Trenzyme GmbH, Konstanz (S.T.) Germany, Lead Identification and Optimization Support (L.H., S.H., R.H.), Immunological and Respiratory Diseases Research (H.S.), CNS Diseases Research (H.R.), and Target Discovery Research (E.S.), Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany; and Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (L.F.)
| | - Lore Florin
- Trenzyme GmbH, Konstanz (S.T.) Germany, Lead Identification and Optimization Support (L.H., S.H., R.H.), Immunological and Respiratory Diseases Research (H.S.), CNS Diseases Research (H.R.), and Target Discovery Research (E.S.), Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany; and Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (L.F.)
| | - Lisa Hospach
- Trenzyme GmbH, Konstanz (S.T.) Germany, Lead Identification and Optimization Support (L.H., S.H., R.H.), Immunological and Respiratory Diseases Research (H.S.), CNS Diseases Research (H.R.), and Target Discovery Research (E.S.), Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany; and Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (L.F.)
| | - Stefan Hörer
- Trenzyme GmbH, Konstanz (S.T.) Germany, Lead Identification and Optimization Support (L.H., S.H., R.H.), Immunological and Respiratory Diseases Research (H.S.), CNS Diseases Research (H.R.), and Target Discovery Research (E.S.), Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany; and Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (L.F.)
| | - Ralf Heilker
- Trenzyme GmbH, Konstanz (S.T.) Germany, Lead Identification and Optimization Support (L.H., S.H., R.H.), Immunological and Respiratory Diseases Research (H.S.), CNS Diseases Research (H.R.), and Target Discovery Research (E.S.), Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany; and Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (L.F.)
| |
Collapse
|
30
|
Hosoya M, Fujioka M, Sone T, Okamoto S, Akamatsu W, Ukai H, Ueda HR, Ogawa K, Matsunaga T, Okano H. Cochlear Cell Modeling Using Disease-Specific iPSCs Unveils a Degenerative Phenotype and Suggests Treatments for Congenital Progressive Hearing Loss. Cell Rep 2017; 18:68-81. [DOI: 10.1016/j.celrep.2016.12.020] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 09/20/2016] [Accepted: 12/06/2016] [Indexed: 12/29/2022] Open
|
31
|
Chapter 17 Sterile Plate-Based Vitrification of Adherent Human Pluripotent Stem Cells and Their Derivatives Using the TWIST Method. Methods Mol Biol 2017; 1568:231-241. [PMID: 28421501 DOI: 10.1007/978-1-4939-6828-2_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Due to their high biological complexity, e.g., their close cell-to-cell contacts, cryopreservation of human pluripotent stem cells with standard slow-rate protocols often is inefficient and can hardly be standardized. Vitrification that means ultrafast freezing already showed very good viability and recovery rates for this sensitive cell system, but is only applicable for low cell numbers, bears a high risk of contamination, and can hardly be implemented under GxP regulations. In this chapter, a sterile plate-based vitrification method for adherent pluripotent stem cells and their derivatives is presented based on a procedure and device for human embryonic stem cells developed by Beier et al. (Cryobiology 66:8-16, 2013). This protocol overcomes the limitations of conventional vitrification procedures resulting in the highly efficient preservation of ready-to-use adherent pluripotent stem cells with the possibility of vitrifying cells in multi-well formats for direct application in high-throughput screenings.
Collapse
|
32
|
Traub S, Stahl H, Rosenbrock H, Simon E, Heilker R. Upscaling of hiPS Cell-Derived Neurons for High-Throughput Screening. SLAS DISCOVERY 2016; 22:274-286. [PMID: 28231034 DOI: 10.1177/1087057116678161] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The advent of human-induced pluripotent stem (hiPS) cell-derived neurons promised to provide better model cells for drug discovery in the context of the central nervous system. This work demonstrates both the upscaling of cellular expansion and the acceleration of neuronal differentiation to accommodate the immense material needs of a high-throughput screening (HTS) approach. Using GRowth factor-driven expansion and INhibition of NotCH (GRINCH) during maturation, the derived cells are here referred to as GRINCH neurons. GRINCH cells displayed neuronal markers, and their functional activity could be demonstrated by electrophysiological recordings. In an application of GRINCH neurons, the brain-derived neurotrophic factor (BDNF)-mediated activation of tropomyosin receptor kinase (TrkB) was investigated as a promising drug target to treat synaptic dysfunctions. To assess the phosphorylation of endogenous TrkB in the GRINCH cells, the highly sensitive amplified luminescent proximity homogeneous assay LISA (AlphaLISA) format was established as a primary screen. A high-throughput reverse transcription (RT)-PCR format was employed as a secondary assay to analyze TrkB-mediated downstream target gene expression. In summary, an optimized differentiation protocol, highly efficient cell upscaling, and advanced assay miniaturization, combined with increased detection sensitivity, pave the way for a new generation of predictive cell-based drug discovery.
Collapse
Affiliation(s)
- Stefanie Traub
- 1 Lead Identification and Optimization Support, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Heiko Stahl
- 2 Immunological and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Holger Rosenbrock
- 3 CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Eric Simon
- 4 Target Discovery Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Ralf Heilker
- 1 Lead Identification and Optimization Support, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| |
Collapse
|
33
|
Szabo A, Kovacs A, Riba J, Djurovic S, Rajnavolgyi E, Frecska E. The Endogenous Hallucinogen and Trace Amine N,N-Dimethyltryptamine (DMT) Displays Potent Protective Effects against Hypoxia via Sigma-1 Receptor Activation in Human Primary iPSC-Derived Cortical Neurons and Microglia-Like Immune Cells. Front Neurosci 2016; 10:423. [PMID: 27683542 PMCID: PMC5021697 DOI: 10.3389/fnins.2016.00423] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 08/30/2016] [Indexed: 12/11/2022] Open
Abstract
N,N-dimethyltryptamine (DMT) is a potent endogenous hallucinogen present in the brain of humans and other mammals. Despite extensive research, its physiological role remains largely unknown. Recently, DMT has been found to activate the sigma-1 receptor (Sig-1R), an intracellular chaperone fulfilling an interface role between the endoplasmic reticulum (ER) and mitochondria. It ensures the correct transmission of ER stress into the nucleus resulting in the enhanced production of antistress and antioxidant proteins. Due to this function, the activation of Sig-1R can mitigate the outcome of hypoxia or oxidative stress. In this paper, we aimed to test the hypothesis that DMT plays a neuroprotective role in the brain by activating the Sig-1R. We tested whether DMT can mitigate hypoxic stress in in vitro cultured human cortical neurons (derived from induced pluripotent stem cells, iPSCs), monocyte-derived macrophages (moMACs), and dendritic cells (moDCs). Results showed that DMT robustly increases the survival of these cell types in severe hypoxia (0.5% O2) through the Sig-1R. Furthermore, this phenomenon is associated with the decreased expression and function of the alpha subunit of the hypoxia-inducible factor 1 (HIF-1) suggesting that DMT-mediated Sig-1R activation may alleviate hypoxia-induced cellular stress and increase survival in a HIF-1-independent manner. Our results reveal a novel and important role of DMT in human cellular physiology. We postulate that this compound may be endogenously generated in situations of stress, ameliorating the adverse effects of hypoxic/ischemic insult to the brain.
Collapse
Affiliation(s)
- Attila Szabo
- NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of OsloOslo, Norway; Division of Mental Health and Addiction, Oslo University HospitalOslo, Norway; Department of Immunology, Faculty of Medicine, University of DebrecenDebrecen, Hungary
| | - Attila Kovacs
- Department of Psychiatry, Faculty of Medicine, University of Debrecen Debrecen, Hungary
| | - Jordi Riba
- Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical ResearchBarcelona, Spain; Centro de Investigación Biomédica en Red de Salud MentalBarcelona, Spain
| | - Srdjan Djurovic
- NORMENT, KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of BergenBergen, Norway; Department of Medical Genetics, Oslo University HospitalOslo, Norway
| | - Eva Rajnavolgyi
- Department of Immunology, Faculty of Medicine, University of Debrecen Debrecen, Hungary
| | - Ede Frecska
- Department of Psychiatry, Faculty of Medicine, University of Debrecen Debrecen, Hungary
| |
Collapse
|
34
|
Physiological maturation and drug responses of human induced pluripotent stem cell-derived cortical neuronal networks in long-term culture. Sci Rep 2016; 6:26181. [PMID: 27188845 PMCID: PMC4870631 DOI: 10.1038/srep26181] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 04/27/2016] [Indexed: 11/09/2022] Open
Abstract
The functional network of human induced pluripotent stem cell (hiPSC)-derived neurons is a potentially powerful in vitro model for evaluating disease mechanisms and drug responses. However, the culture time required for the full functional maturation of individual neurons and networks is uncertain. We investigated the development of spontaneous electrophysiological activity and pharmacological responses for over 1 year in culture using multi-electrode arrays (MEAs). The complete maturation of spontaneous firing, evoked responses, and modulation of activity by glutamatergic and GABAergic receptor antagonists/agonists required 20–30 weeks. At this stage, neural networks also demonstrated epileptiform synchronized burst firing (SBF) in response to pro-convulsants and SBF suppression using clinical anti-epilepsy drugs. Our results reveal the feasibility of long-term MEA measurements from hiPSC-derived neuronal networks in vitro for mechanistic analyses and drug screening. However, developmental changes in electrophysiological and pharmacological properties indicate the necessity for the international standardization of culture and evaluation procedures.
Collapse
|
35
|
Amniotic Fluid Stem Cells: A Novel Source for Modeling of Human Genetic Diseases. Int J Mol Sci 2016; 17:ijms17040607. [PMID: 27110774 PMCID: PMC4849058 DOI: 10.3390/ijms17040607] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/01/2016] [Accepted: 04/11/2016] [Indexed: 12/15/2022] Open
Abstract
In recent years, great interest has been devoted to the use of Induced Pluripotent Stem cells (iPS) for modeling of human genetic diseases, due to the possibility of reprogramming somatic cells of affected patients into pluripotent cells, enabling differentiation into several cell types, and allowing investigations into the molecular mechanisms of the disease. However, the protocol of iPS generation still suffers from technical limitations, showing low efficiency, being expensive and time consuming. Amniotic Fluid Stem cells (AFS) represent a potential alternative novel source of stem cells for modeling of human genetic diseases. In fact, by means of prenatal diagnosis, a number of fetuses affected by chromosomal or Mendelian diseases can be identified, and the amniotic fluid collected for genetic testing can be used, after diagnosis, for the isolation, culture and differentiation of AFS cells. This can provide a useful stem cell model for the investigation of the molecular basis of the diagnosed disease without the necessity of producing iPS, since AFS cells show some features of pluripotency and are able to differentiate in cells derived from all three germ layers “in vitro”. In this article, we describe the potential benefits provided by using AFS cells in the modeling of human genetic diseases.
Collapse
|
36
|
Abstract
Conversion of one cell type into another cell type by forcibly expressing specific cocktails of transcription factors (TFs) has demonstrated that cell fates are not fixed and that cellular differentiation can be a two-way street with many intersections. These experiments also illustrated the sweeping potential of TFs to “read” genetically hardwired regulatory information even in cells where they are not normally expressed and to access and open up tightly packed chromatin to execute gene expression programs. Cellular reprogramming enables the modeling of diseases in a dish, to test the efficacy and toxicity of drugs in patient-derived cells and ultimately, could enable cell-based therapies to cure degenerative diseases. Yet, producing terminally differentiated cells that fully resemble their in vivo counterparts in sufficient quantities is still an unmet clinical need. While efforts are being made to reprogram cells nongenetically by using drug-like molecules, defined TF cocktails still dominate reprogramming protocols. Therefore, the optimization of TFs by protein engineering has emerged as a strategy to enhance reprogramming to produce functional, stable and safe cells for regenerative biomedicine. Engineering approaches focused on Oct4, MyoD, Sox17, Nanog and Mef2c and range from chimeric TFs with added transactivation domains, designer transcription activator-like effectors to activate endogenous TFs to reprogramming TFs with rationally engineered DNA recognition principles. Possibly, applying the complete toolkit of protein design to cellular reprogramming can help to remove the hurdles that, thus far, impeded the clinical use of cells derived from reprogramming technologies.
Collapse
Affiliation(s)
| | | | - Ralf Jauch
- Genome Regulation Laboratory, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, China
| |
Collapse
|
37
|
Suzuki IK, Vanderhaeghen P. Is this a brain which I see before me? Modeling human neural development with pluripotent stem cells. Development 2016; 142:3138-50. [PMID: 26395142 DOI: 10.1242/dev.120568] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The human brain is arguably the most complex structure among living organisms. However, the specific mechanisms leading to this complexity remain incompletely understood, primarily because of the poor experimental accessibility of the human embryonic brain. Over recent years, technologies based on pluripotent stem cells (PSCs) have been developed to generate neural cells of various types. While the translational potential of PSC technologies for disease modeling and/or cell replacement therapies is usually put forward as a rationale for their utility, they are also opening novel windows for direct observation and experimentation of the basic mechanisms of human brain development. PSC-based studies have revealed that a number of cardinal features of neural ontogenesis are remarkably conserved in human models, which can be studied in a reductionist fashion. They have also revealed species-specific features, which constitute attractive lines of investigation to elucidate the mechanisms underlying the development of the human brain, and its link with evolution.
Collapse
Affiliation(s)
- Ikuo K Suzuki
- Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), and ULB Institute of Neuroscience (UNI), 808 Route de Lennik, Brussels B-1070, Belgium
| | - Pierre Vanderhaeghen
- Université Libre de Bruxelles (ULB), Institute for Interdisciplinary Research (IRIBHM), and ULB Institute of Neuroscience (UNI), 808 Route de Lennik, Brussels B-1070, Belgium WELBIO, Université Libre de Bruxelles, 808 Route de Lennik, Brussels B-1070, Belgium
| |
Collapse
|
38
|
Odawara A, Katoh H, Matsuda N, Suzuki I. Induction of long-term potentiation and depression phenomena in human induced pluripotent stem cell-derived cortical neurons. Biochem Biophys Res Commun 2015; 469:856-62. [PMID: 26718408 DOI: 10.1016/j.bbrc.2015.12.087] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 12/20/2015] [Indexed: 11/18/2022]
Abstract
Plasticity such as long-term potentiation (LTP) and long-term potentiation depression (LTD) in neuronal networks has been analyzed using in vitro and in vivo techniques in simple animals to understand learning, memory, and development in brain function. Human induced pluripotent stem cell (hiPSC)-derived neurons may be effectively used for understanding the plasticity mechanism in human neuronal networks, thereby elucidating disease mechanisms and drug discoveries. In this study, we attempted the induction of LTP and LTD phenomena in a cultured hiPSC-derived cerebral cortical neuronal network using multi-electrode array (MEA) systems. High-frequency stimulation (HFS) produced a potentiated and depressed transmission in a neuronal circuit for 1 h in the evoked responses by test stimulus. The cross-correlation of responses revealed that spike patterns with specific timing were generated during LTP induction and disappeared during LTD induction and that the hiPSC-derived cortical neuronal network has the potential to repeatedly express the spike pattern with a precise timing change within 0.5 ms. We also detected the phenomenon for late-phase LTP (L-LTP) like plasticity and the effects for synchronized burst firing (SBF) in spontaneous firings by HFS. In conclusion, we detected the LTP and LTD phenomena in a hiPSC-derived neuronal network as the change of spike pattern. The studies of plasticity using hiPSC-derived neurons and a MEA system may be beneficial for clarifying the functions of human neuronal circuits and for applying to drug screening.
Collapse
Affiliation(s)
- A Odawara
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi 192-0982, Japan; Japan Society for the Promotion of Science, Japan
| | - H Katoh
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi 192-0982, Japan
| | - N Matsuda
- Department of Electronics and Intelligent Systems, Faculty of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi 982-8577, Japan
| | - I Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi 192-0982, Japan; Department of Electronics and Intelligent Systems, Faculty of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi 982-8577, Japan.
| |
Collapse
|
39
|
Georges P, Boissart C, Poulet A, Peschanski M, Benchoua A. Protein Kinase-A Inhibition Is Sufficient to Support Human Neural Stem Cells Self-Renewal. Stem Cells 2015; 33:3666-72. [PMID: 26299682 DOI: 10.1002/stem.2194] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/06/2015] [Indexed: 12/14/2022]
Abstract
Human pluripotent stem cell-derived neural stem cells offer unprecedented opportunities for producing specific types of neurons for several biomedical applications. However, to achieve it, protocols of production and amplification of human neural stem cells need to be standardized, cost effective, and safe. This means that small molecules should progressively replace the use of media containing cocktails of protein-based growth factors. Here we have conducted a phenotypical screening to identify pathways involved in the regulation of hNSC self-renewal. We analyzed 80 small molecules acting as kinase inhibitors and identified compounds of the 5-isoquinolinesulfonamide family, described as protein kinase A (PKA) and protein kinase G inhibitors, as candidates to support hNSC self-renewal. Investigating the mode of action of these compounds, we found that modulation of PKA activity was central in controlling the choice between self-renewal or terminal neuronal differentiation of hNSC. We finally demonstrated that the pharmacological inhibition of PKA using the small molecule HA1004 was sufficient to support the full derivation, propagation, and long-term maintenance of stable hNSC in absence of any other extrinsic signals. Our results indicated that tuning of PKA activity is a core mechanism regulating hNSC self-renewal and differentiation and delineate the minimal culture media requirement to maintain undifferentiated hNSC in vitro.
Collapse
Affiliation(s)
| | | | | | - Marc Peschanski
- INSERM UMR 861, 91030, Evry Cedex, France.,UEVE UMR 861, I-STEM, AFM, 91030, Evry Cedex, France
| | | |
Collapse
|
40
|
Ohara Y, Koganezawa N, Yamazaki H, Roppongi RT, Sato K, Sekino Y, Shirao T. Early-stage development of human induced pluripotent stem cell-derived neurons. J Neurosci Res 2015; 93:1804-13. [PMID: 26346430 PMCID: PMC5049656 DOI: 10.1002/jnr.23666] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 08/03/2015] [Accepted: 08/22/2015] [Indexed: 01/16/2023]
Abstract
Recent advances in human induced pluripotent stem cells (hiPSCs) offer new possibilities for biomedical research and clinical applications. Differentiated neurons from hiPSCs are expected to be useful for developing novel methods of treatment for various neurological diseases. However, the detailed process of functional maturation of hiPSC-derived neurons (hiPS neurons) remains poorly understood. This study analyzes development of hiPS neurons, focusing specifically on early developmental stages through 48 hr after cell seeding; development was compared with that of primary cultured neurons derived from the rat hippocampus. At 5 hr after cell seeding, neurite formation occurs in a similar manner in both neuronal populations. However, very few neurons with axonal polarization were observed in the hiPS neurons even after 48 hr, indicating that hiPS neurons differentiate more slowly than rat neurons. We further investigated the elongation speed of axons and found that hiPS neuronal axons were slower. In addition, we characterized the growth cones. The localization patterns of skeletal proteins F-actin, microtubule, and drebrin were similar to those of rat neurons, and actin depolymerization by cytochalasin D induced similar changes in cytoskeletal distribution in the growth cones between hiPS neurons and rat neurons. These results indicate that, during the very early developmental stage, hiPS neurons develop comparably to rat hippocampal neurons with regard to axonal differentiation, but the growth of axons is slower.
Collapse
Affiliation(s)
- Yuki Ohara
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Noriko Koganezawa
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Hiroyuki Yamazaki
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Reiko T Roppongi
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Kaoru Sato
- Division of Pharmacology, National Institute of Health Sciences, Tokyo, Japan
| | - Yuko Sekino
- Division of Pharmacology, National Institute of Health Sciences, Tokyo, Japan
| | - Tomoaki Shirao
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| |
Collapse
|
41
|
Elanzew A, Sommer A, Pusch-Klein A, Brüstle O, Haupt S. A reproducible and versatile system for the dynamic expansion of human pluripotent stem cells in suspension. Biotechnol J 2015; 10:1589-99. [PMID: 26110829 DOI: 10.1002/biot.201400757] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 03/13/2015] [Accepted: 06/23/2015] [Indexed: 11/11/2022]
Abstract
Reprogramming of patient cells to human induced pluripotent stem cells (hiPSC) has facilitated in vitro disease modeling studies aiming at deciphering the molecular and cellular mechanisms that contribute to disease pathogenesis and progression. To fully exploit the potential of hiPSC for biomedical applications, technologies that enable the standardized generation and expansion of hiPSC from large numbers of donors are required. Paralleled automated processes for the expansion of hiPSC could provide an opportunity to maximize the generation of hiPSC collections from patient cohorts while minimizing hands-on time and costs. In order to develop a simple method for the parallel expansion of human pluripotent stem cells (hPSC) we established a protocol for their cultivation as undifferentiated aggregates in a bench-top bioreactor system (BioLevitator™). We show that long-term expansion (10 passages) of hPSCs either in mTeSR or E8 medium preserved a normal karyotype, three-germ-layer differentiation potential and high expression of pluripotency-associated markers. The system enables the expansion from low inoculation densities (0.3 × 10(5) cells/mL) and provides a simplified, cost-efficient and time-saving method for the provision of hiPSC at midi-scale. Implementation of this protocol in cell production schemes has the potential to advance cell manufacturing in many areas of hiPSC-based medical research.
Collapse
Affiliation(s)
- Andreas Elanzew
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, University of Bonn, Bonn, Germany.,LIFE&BRAIN GmbH, Bonn, Germany
| | | | | | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, University of Bonn, Bonn, Germany. .,LIFE&BRAIN GmbH, Bonn, Germany.
| | - Simone Haupt
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, University of Bonn, Bonn, Germany. .,LIFE&BRAIN GmbH, Bonn, Germany.
| |
Collapse
|
42
|
Human Pluripotent Stem Cell-Derived Retinal Ganglion Cells: Applications for the Study and Treatment of Optic Neuropathies. CURRENT OPHTHALMOLOGY REPORTS 2015; 3:200-206. [PMID: 26618076 DOI: 10.1007/s40135-015-0081-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
43
|
Tabata Y, Murai N, Sasaki T, Taniguchi S, Suzuki S, Yamazaki K, Ito M. Multiparametric Phenotypic Screening System for Profiling Bioactive Compounds Using Human Fetal Hippocampal Neural Stem/Progenitor Cells. ACTA ACUST UNITED AC 2015; 20:1074-83. [DOI: 10.1177/1087057115598119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 07/07/2015] [Indexed: 01/25/2023]
Abstract
Stem cell research has been progressing rapidly, contributing to regenerative biology and regenerative medicine. In this field, small-molecule compounds affecting stem cell proliferation/differentiation have been explored to understand stem cell biology and support regenerative medicine. In this study, we established a multiparametric screening system to detect bioactive compounds affecting the cell fate of human neural stem/progenitor cells (NSCs/NPCs), using human fetal hippocampal NSCs/NPCs, HIP-009 cells. We examined effects of 410 compounds, which were collected based on mechanisms of action (MOAs) and chemotypes, on HIP-009’s cell fate (self-renewal, neuronal and astrocytic differentiation) and morphology by automated multiparametric assays and profiled induced cellular phenotypes. We found that this screening classified compounds with the same MOAs into subgroups according to additional pharmacological effects (e.g., mammalian target of rapamycin complex 1 [mTORC1] inhibitors and mTORC1/mTORC2 dual inhibitors among mTOR inhibitors). Moreover, it identified compounds that have off-target effects under matrix analyses of MOAs and structure similarities (e.g., neurotropic effects of amitriptyline among tri- and tetracyclic compounds). Therefore, this automated, medium-throughput and multiparametric screening system is useful for finding compounds that affect the cell fate of human NSCs/NPCs for supporting regenerative medicine and to fingerprint compounds based on human stem cells’ multipotency, leading to understanding of stem cell biology.
Collapse
Affiliation(s)
- Yoshikuni Tabata
- Next Generation Systems CFU, Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki, Japan
| | - Norio Murai
- Next Generation Systems CFU, Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki, Japan
| | - Takeo Sasaki
- Global Discovery Research, Neuroscience and General Medicine PCU, Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki, Japan
| | - Sachie Taniguchi
- Next Generation Systems CFU, Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki, Japan
| | - Shuichi Suzuki
- Next Generation Systems CFU, Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki, Japan
| | - Kazuto Yamazaki
- Next Generation Systems CFU, Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki, Japan
| | - Masashi Ito
- Next Generation Systems CFU, Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki, Japan
| |
Collapse
|
44
|
Wan W, Cao L, Kalionis B, Xia S, Tai X. Applications of Induced Pluripotent Stem Cells in Studying the Neurodegenerative Diseases. Stem Cells Int 2015; 2015:382530. [PMID: 26240571 PMCID: PMC4512612 DOI: 10.1155/2015/382530] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 12/05/2014] [Indexed: 12/21/2022] Open
Abstract
Neurodegeneration is the umbrella term for the progressive loss of structure or function of neurons. Incurable neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD) show dramatic rising trends particularly in the advanced age groups. However, the underlying mechanisms are not yet fully elucidated, and to date there are no biomarkers for early detection or effective treatments for the underlying causes of these diseases. Furthermore, due to species variation and differences between animal models (e.g., mouse transgenic and knockout models) of neurodegenerative diseases, substantial debate focuses on whether animal and cell culture disease models can correctly model the condition in human patients. In 2006, Yamanaka of Kyoto University first demonstrated a novel approach for the preparation of induced pluripotent stem cells (iPSCs), which displayed similar pluripotency potential to embryonic stem cells (ESCs). Currently, iPSCs studies are permeating many sectors of disease research. Patient sample-derived iPSCs can be used to construct patient-specific disease models to elucidate the pathogenic mechanisms of disease development and to test new therapeutic strategies. Accordingly, the present review will focus on recent progress in iPSC research in the modeling of neurodegenerative disorders and in the development of novel therapeutic options.
Collapse
Affiliation(s)
- Wenbin Wan
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lan Cao
- State Key Laboratory of Medical Neurobiology, Department of Neurobiology and Institutes of Brain Science, School of Basic Medical Science, Fudan University, Shanghai 200032, China
| | - Bill Kalionis
- Department of Perinatal Medicine, Pregnancy Research Centre and University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, VIC 3052, Australia
| | - Shijin Xia
- Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Xiantao Tai
- School of Acupuncture, Massage and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming 650500, China
| |
Collapse
|
45
|
Distinct Neurodegenerative Changes in an Induced Pluripotent Stem Cell Model of Frontotemporal Dementia Linked to Mutant TAU Protein. Stem Cell Reports 2015; 5:83-96. [PMID: 26143746 PMCID: PMC4618448 DOI: 10.1016/j.stemcr.2015.06.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 06/08/2015] [Accepted: 06/08/2015] [Indexed: 01/06/2023] Open
Abstract
Frontotemporal dementia (FTD) is a frequent form of early-onset dementia and can be caused by mutations in MAPT encoding the microtubule-associated protein TAU. Because of limited availability of neural cells from patients' brains, the underlying mechanisms of neurodegeneration in FTD are poorly understood. Here, we derived induced pluripotent stem cells (iPSCs) from individuals with FTD-associated MAPT mutations and differentiated them into mature neurons. Patient iPSC-derived neurons demonstrated pronounced TAU pathology with increased fragmentation and phospho-TAU immunoreactivity, decreased neurite extension, and increased but reversible oxidative stress response to inhibition of mitochondrial respiration. Furthermore, FTD neurons showed an activation of the unfolded protein response, and a transcriptome analysis demonstrated distinct, disease-associated gene expression profiles. These findings indicate distinct neurodegenerative changes in FTD caused by mutant TAU and highlight the unique opportunity to use neurons differentiated from patient-specific iPSCs to identify potential targets for drug screening purposes and therapeutic intervention.
Collapse
|
46
|
Properties of human central nervous system neurons in a glia-depleted (isolated) culture system. J Neurosci Methods 2015; 253:142-50. [PMID: 26093165 DOI: 10.1016/j.jneumeth.2015.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 06/09/2015] [Accepted: 06/11/2015] [Indexed: 01/14/2023]
Abstract
BACKGROUND Current methods for studying human neurons depend on a feeder layer of astroglia supplemented with animal serum to support the growing neurons. These requirements undermine many of the advantages provided by in vitro cell culture approaches when compared with more complex in vivo techniques. NEW METHOD Here, we identified a reliable marker (MHCI) that allows for direct isolation of primary neurons from fetal human brain. We utilized a magnetic labeling and isolation technique to separate neurons from other neural cells. We established a defined condition, omitting the astroglial supports that could maintain the human neurons for varying amounts of time. RESULTS We showed that the new method significantly improved the purity of human neurons in culture without the need for further chemical/mechanical enrichment. We demonstrated the suitability of these neurons for functional studies including Rho-kinase dependent regulation of neurite outgrowth and ensheathment in co-cultures with oligodendrocyte progenitor cells derived from fetal human brain. COMPARISON WITH EXISTING METHODS The accountability for neuron-only seeding and the controllable density allows for better neuronal maturation and better visualization of the different neuronal compartments. The higher purity culture constitutes an effective system to study and screen for compounds that impact neuron biology without potential confounding effects from glial crowding. CONCLUSIONS High purity human neurons generated using the improved method will enable enhanced reliability in the discovery and development of drugs with neuroregenerative and neuroprotective activity.
Collapse
|
47
|
Corti S, Faravelli I, Cardano M, Conti L. Human pluripotent stem cells as tools for neurodegenerative and neurodevelopmental disease modeling and drug discovery. Expert Opin Drug Discov 2015; 10:615-29. [PMID: 25891144 DOI: 10.1517/17460441.2015.1037737] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Although intensive efforts have been made, effective treatments for neurodegenerative and neurodevelopmental diseases have not been yet discovered. Possible reasons for this include the lack of appropriate disease models of human neurons and a limited understanding of the etiological and neurobiological mechanisms. Recent advances in pluripotent stem cell (PSC) research have now opened the path to the generation of induced pluripotent stem cells (iPSCs) starting from somatic cells, thus offering an unlimited source of patient-specific disease-relevant neuronal cells. AREAS COVERED In this review, the authors focus on the use of human PSC-derived cells in modeling neurological disorders and discovering of new drugs and provide their expert perspectives on the field. EXPERT OPINION The advent of human iPSC-based disease models has fuelled renewed enthusiasm and enormous expectations for insights of disease mechanisms and identification of more disease-relevant and novel molecular targets. Human PSCs offer a unique tool that is being profitably exploited for high-throughput screening (HTS) platforms. This process can lead to the identification and optimization of molecules/drugs and thus move forward new pharmacological therapies for a wide range of neurodegenerative and neurodevelopmental conditions. It is predicted that improvements in the production of mature neuronal subtypes, from patient-specific human-induced pluripotent stem cells and their adaptation to culture, to HTS platforms will allow the increased exploitation of human pluripotent stem cells in drug discovery programs.
Collapse
Affiliation(s)
- Stefania Corti
- University of Milan, Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation, Neurology Unit, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico , via Francesco Sforza 35, Milan 20122 , Italy +39 02 55033817 ;
| | | | | | | |
Collapse
|
48
|
Challenges and opportunities toward enabling phenotypic screening of complex and 3D cell models. Future Med Chem 2015; 7:513-25. [DOI: 10.4155/fmc.14.163] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Increasingly, organotypic cellular platforms are being recognized as useful tools in drug discovery. This review offers an industry-centric perspective on the benefits of emerging complex cell models over conventional 2D systems, as well as the challenges and opportunities for incorporating these multidimensional platforms into high-density formats. We particularly highlight the need for novel chemical sensors to noninvasively quantitate 3D structures in real time, and we contend that the use of more focused chemical and genomics libraries will enable screening of complex cell models derived from primary and induced pluripotent stem cells. Finally, we offer outlooks on several emerging technologies that show great potential for future integration of complex cell systems into contemporary drug screening.
Collapse
|