1
|
Park JW, Bae SJ, Yun JH, Kim S, Park M. Assessment of Genetic Stability in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes by Using Droplet Digital PCR. Int J Mol Sci 2024; 25:1101. [PMID: 38256178 PMCID: PMC10815998 DOI: 10.3390/ijms25021101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/04/2024] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
Unintended genetic modifications that occur during the differentiation and proliferation of human induced pluripotent stem cells (hiPSCs) can lead to tumorigenicity. This is a crucial concern in the development of stem cell-based therapies to ensure the safety and efficacy of the final product. Moreover, conventional genetic stability testing methods are limited by low sensitivity, which is an issue that remains unsolved. In this study, we assessed the genetic stability of hiPSCs and hiPSC-derived cardiomyocytes using various testing methods, including karyotyping, CytoScanHD chip analysis, whole-exome sequencing, and targeted sequencing. Two specific genetic mutations in KMT2C and BCOR were selected from the 17 gene variants identified by whole-exome and targeted sequencing methods, which were validated using droplet digital PCR. The applicability of this approach to stem cell-based therapeutic products was further demonstrated with associated validation according to the International Council for Harmonisation (ICH) guidelines, including specificity, precision, robustness, and limit of detection. Our droplet digital PCR results showed high sensitivity and accuracy for quantitatively detecting gene mutations, whereas conventional qPCR could not avoid false positives. In conclusion, droplet digital PCR is a highly sensitive and precise method for assessing the expression of mutations with tumorigenic potential for the development of stem cell-based therapeutics.
Collapse
Affiliation(s)
| | | | | | | | - Misun Park
- Advanced Bioconvergence Product Research Division, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju-si 28159, Republic of Korea; (J.W.P.); (S.J.B.); (J.H.Y.); (S.K.)
| |
Collapse
|
2
|
Yang M, Liu M, Sánchez YF, Avazzadeh S, Quinlan LR, Liu G, Lu Y, Yang G, O'Brien T, Henshall DC, Hardiman O, Shen S. A novel protocol to derive cervical motor neurons from induced pluripotent stem cells for amyotrophic lateral sclerosis. Stem Cell Reports 2023; 18:1870-1883. [PMID: 37595581 PMCID: PMC10545486 DOI: 10.1016/j.stemcr.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/20/2023] Open
Abstract
Sporadic amyotrophic lateral sclerosis (sALS) is the majority of ALS, and the lack of appropriate disease models has hindered its research. Induced pluripotent stem cell (iPSC) technology now permits derivation of iPSCs from somatic cells of sALS patients to investigate disease phenotypes and mechanisms. Most existing differentiation protocols are time-consuming or low efficient in generating motor neurons (MNs). Here we report a rapid and simple protocol to differentiate MNs in monolayer culture using small molecules, which led to nearly pure neural stem cells in 6 days, robust OLIG2+ pMNs (73%-91%) in 12 days, enriched CHAT+ cervical spinal MNs (sMNs) (88%-97%) in 18 days, and functionally mature sMNs in 28 days. This simple and reproducible protocol permitted the identification of hyperexcitability phenotypes in our sALS iPSC-derived sMNs, and its application in neurodegenerative diseases should facilitate in vitro disease modeling, drug screening, and the development of cell therapy.
Collapse
Affiliation(s)
- Meimei Yang
- Regenerative Medicine Institute, School of Medicine, University of Galway, H91 W2TY Galway, Ireland; FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases and Department of Physiology & Medical Physics, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
| | - Min Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Molecular and Cellular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, China
| | - Yajaira Feller Sánchez
- Cellular Physiology Research Laboratory and CÚRAM SFI Centre for Research in Medical Devices, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Sahar Avazzadeh
- Cellular Physiology Research Laboratory and CÚRAM SFI Centre for Research in Medical Devices, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Leo R Quinlan
- Cellular Physiology Research Laboratory and CÚRAM SFI Centre for Research in Medical Devices, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Hebei Key Laboratory of Heart and Metabolism, Hebei Engineering Research Center of Intelligent Medical Clinical Application, Hebei International Joint Research Center for Structural Heart Disease, Shijiazhuang, Hebei, China
| | - Yin Lu
- College of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Guangming Yang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Timothy O'Brien
- Regenerative Medicine Institute, School of Medicine, University of Galway, H91 W2TY Galway, Ireland
| | - David C Henshall
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases and Department of Physiology & Medical Physics, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland; Department of Physiology and Medical Physics, RCSI University of Medicine & Health Sciences, D02 YN77 Dublin, Ireland.
| | - Orla Hardiman
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases and Department of Physiology & Medical Physics, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland; Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland.
| | - Sanbing Shen
- Regenerative Medicine Institute, School of Medicine, University of Galway, H91 W2TY Galway, Ireland; FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases and Department of Physiology & Medical Physics, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland.
| |
Collapse
|
3
|
Yang X, Chen D, Sun Q, Wang Y, Xia Y, Yang J, Lin C, Dang X, Cen Z, Liang D, Wei R, Xu Z, Xi G, Xue G, Ye C, Wang LP, Zou P, Wang SQ, Rivera-Fuentes P, Püntener S, Chen Z, Liu Y, Zhang J, Zhao Y. A live-cell image-based machine learning strategy for reducing variability in PSC differentiation systems. Cell Discov 2023; 9:53. [PMID: 37280224 DOI: 10.1038/s41421-023-00543-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 03/13/2023] [Indexed: 06/08/2023] Open
Abstract
The differentiation of pluripotent stem cells (PSCs) into diverse functional cell types provides a promising solution to support drug discovery, disease modeling, and regenerative medicine. However, functional cell differentiation is currently limited by the substantial line-to-line and batch-to-batch variabilities, which severely impede the progress of scientific research and the manufacturing of cell products. For instance, PSC-to-cardiomyocyte (CM) differentiation is vulnerable to inappropriate doses of CHIR99021 (CHIR) that are applied in the initial stage of mesoderm differentiation. Here, by harnessing live-cell bright-field imaging and machine learning (ML), we realize real-time cell recognition in the entire differentiation process, e.g., CMs, cardiac progenitor cells (CPCs), PSC clones, and even misdifferentiated cells. This enables non-invasive prediction of differentiation efficiency, purification of ML-recognized CMs and CPCs for reducing cell contamination, early assessment of the CHIR dose for correcting the misdifferentiation trajectory, and evaluation of initial PSC colonies for controlling the start point of differentiation, all of which provide a more invulnerable differentiation method with resistance to variability. Moreover, with the established ML models as a readout for the chemical screen, we identify a CDK8 inhibitor that can further improve the cell resistance to the overdose of CHIR. Together, this study indicates that artificial intelligence is able to guide and iteratively optimize PSC differentiation to achieve consistently high efficiency across cell lines and batches, providing a better understanding and rational modulation of the differentiation process for functional cell manufacturing in biomedical applications.
Collapse
Affiliation(s)
- Xiaochun Yang
- State Key Laboratory of Natural and Biomimetic Drugs, MOE Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Daichao Chen
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Qiushi Sun
- Beijing Key Lab of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| | - Yao Wang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yu Xia
- College of Engineering, Peking University, Beijing, China
| | - Jinyu Yang
- College of Engineering, Peking University, Beijing, China
| | - Chang Lin
- College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, China
| | - Xin Dang
- State Key Laboratory of Natural and Biomimetic Drugs, MOE Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Zimu Cen
- State Key Laboratory of Natural and Biomimetic Drugs, MOE Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Dongdong Liang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Rong Wei
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Ze Xu
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing, China
| | - Guangyin Xi
- State Key Laboratory of Natural and Biomimetic Drugs, MOE Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Gang Xue
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Can Ye
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Li-Peng Wang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing, China
| | - Peng Zou
- College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Shi-Qiang Wang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing, China
| | | | - Salome Püntener
- Department of Chemistry, University of Zurich, Zurich, Switzerland
- Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédéral de Lausanne, Lausanne, Switzerland
| | - Zhixing Chen
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- Institute of Molecular Medicine, National Biomedical Imaging Center, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Yi Liu
- Beijing Key Lab of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China.
| | - Jue Zhang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
- College of Engineering, Peking University, Beijing, China.
| | - Yang Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, MOE Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
4
|
Puigdevall P, Jerber J, Danecek P, Castellano S, Kilpinen H. Somatic mutations alter the differentiation outcomes of iPSC-derived neurons. CELL GENOMICS 2023; 3:100280. [PMID: 37082143 PMCID: PMC10112289 DOI: 10.1016/j.xgen.2023.100280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 11/11/2022] [Accepted: 02/23/2023] [Indexed: 04/22/2023]
Abstract
The use of induced pluripotent stem cells (iPSC) as models for development and human disease has enabled the study of otherwise inaccessible tissues. A remaining challenge in developing reliable models is our limited understanding of the factors driving irregular differentiation of iPSCs, particularly the impact of acquired somatic mutations. We leveraged data from a pooled dopaminergic neuron differentiation experiment of 238 iPSC lines profiled with single-cell RNA and whole-exome sequencing to study how somatic mutations affect differentiation outcomes. We found that deleterious somatic mutations in key developmental genes, notably the BCOR gene, are strongly associated with failure in dopaminergic neuron differentiation and a larger proliferation rate in culture. We further identified broad differences in cell type composition between incorrectly and successfully differentiating lines, as well as significant changes in gene expression contributing to the inhibition of neurogenesis. Our work calls for caution in interpreting differentiation-related phenotypes in disease-modeling experiments.
Collapse
Affiliation(s)
- Pau Puigdevall
- UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Haartmaninkatu 8, PO Box 63, Helsinki 00014, Finland
| | - Julie Jerber
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Petr Danecek
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Sergi Castellano
- UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Helena Kilpinen
- UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Haartmaninkatu 8, PO Box 63, Helsinki 00014, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Viikinkaari 1, PO Box 65, Helsinki 00014, Finland
| |
Collapse
|
5
|
Castillo Bautista CM, Sterneckert J. Progress and challenges in directing the differentiation of human iPSCs into spinal motor neurons. Front Cell Dev Biol 2023; 10:1089970. [PMID: 36684437 PMCID: PMC9849822 DOI: 10.3389/fcell.2022.1089970] [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: 11/04/2022] [Accepted: 12/21/2022] [Indexed: 01/07/2023] Open
Abstract
Motor neuron (MN) diseases, including amyotrophic lateral sclerosis, progressive bulbar palsy, primary lateral sclerosis and spinal muscular atrophy, cause progressive paralysis and, in many cases, death. A better understanding of the molecular mechanisms of pathogenesis is urgently needed to identify more effective therapies. However, studying MNs has been extremely difficult because they are inaccessible in the spinal cord. Induced pluripotent stem cells (iPSCs) can generate a theoretically limitless number of MNs from a specific patient, making them powerful tools for studying MN diseases. However, to reach their potential, iPSCs need to be directed to efficiently differentiate into functional MNs. Here, we review the reported differentiation protocols for spinal MNs, including induction with small molecules, expression of lineage-specific transcription factors, 2-dimensional and 3-dimensional cultures, as well as the implementation of microfluidics devices and co-cultures with other cell types, including skeletal muscle. We will summarize the advantages and disadvantages of each strategy. In addition, we will provide insights into how to address some of the remaining challenges, including reproducibly obtaining mature and aged MNs.
Collapse
Affiliation(s)
| | - Jared Sterneckert
- Center for Regenerative Therapies TU Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany,Medical Faculty Carl Gustav Carus of TU Dresden, Dresden, Germany,*Correspondence: Jared Sterneckert,
| |
Collapse
|
6
|
Benchmarking brain organoid recapitulation of fetal corticogenesis. Transl Psychiatry 2022; 12:520. [PMID: 36539399 PMCID: PMC9767930 DOI: 10.1038/s41398-022-02279-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Brain organoids are becoming increasingly relevant to dissect the molecular mechanisms underlying psychiatric and neurological conditions. The in vitro recapitulation of key features of human brain development affords the unique opportunity of investigating the developmental antecedents of neuropsychiatric conditions in the context of the actual patients' genetic backgrounds. Specifically, multiple strategies of brain organoid (BO) differentiation have enabled the investigation of human cerebral corticogenesis in vitro with increasing accuracy. However, the field lacks a systematic investigation of how closely the gene co-expression patterns seen in cultured BO from different protocols match those observed in fetal cortex, a paramount information for ensuring the sensitivity and accuracy of modeling disease trajectories. Here we benchmark BO against fetal corticogenesis by integrating transcriptomes from in-house differentiated cortical BO (CBO), other BO systems, human fetal brain samples processed in-house, and prenatal cortices from the BrainSpan Atlas. We identified co-expression patterns and prioritized hubs of human corticogenesis and CBO differentiation, highlighting both well-preserved and discordant trends across BO protocols. We evaluated the relevance of identified gene modules for neurodevelopmental disorders and psychiatric conditions finding significant enrichment of disease risk genes especially in modules related to neuronal maturation and synapsis development. The longitudinal transcriptomic analysis of CBO revealed a two-step differentiation composed of a fast-evolving phase, corresponding to the appearance of the main cell populations of the cortex, followed by a slow-evolving one characterized by milder transcriptional changes. Finally, we observed heterochronicity of differentiation across BO models compared to fetal cortex. Our approach provides a framework to directly compare the extent of in vivo/in vitro alignment of neurodevelopmentally relevant processes and their attending temporalities, structured as a resource to query for modeling human corticogenesis and the neuropsychiatric outcomes of its alterations.
Collapse
|
7
|
Cantor EL, Shen F, Jiang G, Tan Z, Cunningham GM, Wu X, Philips S, Schneider BP. Passage number affects differentiation of sensory neurons from human induced pluripotent stem cells. Sci Rep 2022; 12:15869. [PMID: 36151116 PMCID: PMC9508090 DOI: 10.1038/s41598-022-19018-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/23/2022] [Indexed: 11/23/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are a valuable resource for neurological disease-modeling and drug discovery due to their ability to differentiate into neurons reflecting the genetics of the patient from which they are derived. iPSC-derived cultures, however, are highly variable due to heterogeneity in culture conditions. We investigated the effect of passage number on iPSC differentiation to optimize the generation of sensory neurons (iPSC-dSNs). Three iPSC lines reprogrammed from the peripheral blood of three donors were differentiated into iPSC-dSNs at passage numbers within each of the following ranges: low (5-10), intermediate (20-26), and high (30-38). Morphology and pluripotency of the parent iPSCs were assessed prior to differentiation. iPSC-dSNs were evaluated based on electrophysiological properties and expression of key neuronal markers. All iPSC lines displayed similar morphology and were similarly pluripotent across passage numbers. However, the expression levels of neuronal markers and sodium channel function analyses indicated that iPSC-dSNs differentiated from low passage numbers better recapitulated the sensory neuron phenotype than those differentiated from intermediate or high passage numbers. Our results demonstrate that lower passage numbers may be better suited for differentiation into peripheral sensory neurons.
Collapse
Affiliation(s)
- Erica L Cantor
- Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fei Shen
- Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Guanglong Jiang
- Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Zhiyong Tan
- Pharmacology & Toxicology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Geneva M Cunningham
- Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xi Wu
- Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Santosh Philips
- Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bryan P Schneider
- Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
8
|
Vojnits K, Nakanishi M, Porras D, Kim Y, Feng Z, Golubeva D, Bhatia M. Developing CRISPR/Cas9-Mediated Fluorescent Reporter Human Pluripotent Stem-Cell Lines for High-Content Screening. Molecules 2022; 27:molecules27082434. [PMID: 35458632 PMCID: PMC9025795 DOI: 10.3390/molecules27082434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/03/2022] [Accepted: 04/06/2022] [Indexed: 12/22/2022] Open
Abstract
Application of the CRISPR/Cas9 system to knock in fluorescent proteins to endogenous genes of interest in human pluripotent stem cells (hPSCs) has the potential to facilitate hPSC-based disease modeling, drug screening, and optimization of transplantation therapy. To evaluate the capability of fluorescent reporter hPSC lines for high-content screening approaches, we targeted EGFP to the endogenous OCT4 locus. Resulting hPSC–OCT4–EGFP lines generated expressed EGFP coincident with pluripotency markers and could be adapted to multi-well formats for high-content screening (HCS) campaigns. However, after long-term culture, hPSCs transiently lost their EGFP expression. Alternatively, through EGFP knock-in to the AAVS1 locus, we established a stable and consistent EGFP-expressing hPSC–AAVS1–EGFP line that maintained EGFP expression during in vitro hematopoietic and neural differentiation. Thus, hPSC–AAVS1–EGFP-derived sensory neurons could be adapted to a high-content screening platform that can be applied to high-throughput small-molecule screening and drug discovery campaigns. Our observations are consistent with recent findings indicating that high-frequency on-target complexities appear following CRISPR/Cas9 genome editing at the OCT4 locus. In contrast, we demonstrate that the AAVS1 locus is a safe genomic location in hPSCs with high gene expression that does not impact hPSC quality and differentiation. Our findings suggest that the CRISPR/Cas9-integrated AAVS1 system should be applied for generating stable reporter hPSC lines for long-term HCS approaches, and they underscore the importance of careful evaluation and selection of the applied reporter cell lines for HCS purposes.
Collapse
|
9
|
Increased Neuronal Differentiation Efficiency in High Cell Density-Derived Induced Pluripotent Stem Cells. Stem Cells Int 2019; 2019:2018784. [PMID: 31871463 PMCID: PMC6913159 DOI: 10.1155/2019/2018784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 10/23/2019] [Indexed: 01/30/2023] Open
Abstract
Human pluripotent stem cells (hPSCs), including induced pluripotent stem cells (iPSCs), provide access to hard-to-obtain cells for studies under physiological and disease conditions. For the study of neurodegenerative diseases, especially sporadic cases where the “disease condition” might be restricted towards the neuroectodermal lineage, obtaining the affected neurons is important to help unravel the underlying molecular mechanism leading to the diseases. Although differentiation of iPSCs to neural lineage allows acquisition of cell types of interest, the technology suffers from low efficiency leading to low yield of neurons. Here, we investigated the potential of adult neuroprogenitor cells (aNPCs) for iPSC derivation and possible confounders such as cell density of infected NPCs on their subsequent neuronal differentiation potential from reprogrammed cells under isogenic conditions. Characterized hiPSCs of defined cell densities generated from aNPCs were subjected to neuronal differentiation on PA6 stromal cells. The results showed that hiPSC clones obtained from low seeding density (iPSC-aNPCLow) differentiated less efficiently compared to those from higher density (iPSC-aNPCHigh). Our findings might help to further improve the yield and quality of neurons for in vitro modelling of neurodegenerative diseases.
Collapse
|
10
|
Sharlow ER, Koseoglu MM, Bloom GS, Lazo JS. The Promise and Perils of Compound Discovery Screening with Inducible Pluripotent Cell-Derived Neurons. Assay Drug Dev Technol 2019; 18:97-103. [PMID: 31095406 DOI: 10.1089/adt.2019.914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Neurological diseases comprise more than a thousand ailments that adversely affect the brain and nervous system. When grouped together, these neurological conditions impact an estimated 100 million individuals in the United States and up to a billion people worldwide, making drug discovery efforts imperative. However, recent research and development efforts for these neurological diseases, including Alzheimer's disease and amyotrophic lateral sclerosis, have been exceedingly disappointing and typify the challenges associated with translating in vitro and cell-based discoveries to successful preclinical models and subsequent human clinical trials. Our viewpoint is that neuronal progenitor cells and neurons derived from inducible pluripotent stem cells afford an innovative translational bridge, with higher pathological relevancy than previous cellular models. We outline some of the opportunities and challenges associated with their evolving usage in drug discovery and development.
Collapse
Affiliation(s)
- Elizabeth R Sharlow
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia.,Fiske Drug Discovery Laboratory, University of Virginia, Charlottesville, Virginia
| | - Mehmet Murat Koseoglu
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia.,Fiske Drug Discovery Laboratory, University of Virginia, Charlottesville, Virginia
| | - George S Bloom
- Department of Biology, University of Virginia, Charlottesville, Virginia.,Department of Cell Biology, University of Virginia, Charlottesville, Virginia.,Department of Neuroscience, University of Virginia, Charlottesville, Virginia
| | - John S Lazo
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia.,Fiske Drug Discovery Laboratory, University of Virginia, Charlottesville, Virginia.,Department of Chemistry, University of Virginia, Charlottesville, Virginia.,Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
11
|
Ma K, Deng X, Xia X, Fan Z, Qi X, Wang Y, Li Y, Ma Y, Chen Q, Peng H, Ding J, Li C, Huang Y, Tian C, Zheng JC. Direct conversion of mouse astrocytes into neural progenitor cells and specific lineages of neurons. Transl Neurodegener 2018; 7:29. [PMID: 30410751 PMCID: PMC6217767 DOI: 10.1186/s40035-018-0132-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/11/2018] [Indexed: 12/26/2022] Open
Abstract
Background Cell replacement therapy has been envisioned as a promising treatment for neurodegenerative diseases. Due to the ethical concerns of ESCs-derived neural progenitor cells (NPCs) and tumorigenic potential of iPSCs, reprogramming of somatic cells directly into multipotent NPCs has emerged as a preferred approach for cell transplantation. Methods Mouse astrocytes were reprogrammed into NPCs by the overexpression of transcription factors (TFs) Foxg1, Sox2, and Brn2. The generation of subtypes of neurons was directed by the force expression of cell-type specific TFs Lhx8 or Foxa2/Lmx1a. Results Astrocyte-derived induced NPCs (AiNPCs) share high similarities, including the expression of NPC-specific genes, DNA methylation patterns, the ability to proliferate and differentiate, with the wild type NPCs. The AiNPCs are committed to the forebrain identity and predominantly differentiated into glutamatergic and GABAergic neuronal subtypes. Interestingly, additional overexpression of TFs Lhx8 and Foxa2/Lmx1a in AiNPCs promoted cholinergic and dopaminergic neuronal differentiation, respectively. Conclusions Our studies suggest that astrocytes can be converted into AiNPCs and lineage-committed AiNPCs can acquire differentiation potential of other lineages through forced expression of specific TFs. Understanding the impact of the TF sets on the reprogramming and differentiation into specific lineages of neurons will provide valuable strategies for astrocyte-based cell therapy in neurodegenerative diseases.
Collapse
Affiliation(s)
- Kangmu Ma
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China.,3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Xiaobei Deng
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Xiaohuan Xia
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Zhaohuan Fan
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Xinrui Qi
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Yongxiang Wang
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China.,3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Yuju Li
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China.,3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Yizhao Ma
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Qiang Chen
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China.,3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Hui Peng
- 3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Jianqing Ding
- 4Department of Neurology & Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Chunhong Li
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Yunlong Huang
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China.,3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Changhai Tian
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China.,3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Jialin C Zheng
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China.,2Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, 200092 China.,3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA.,5Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| |
Collapse
|
12
|
Gatie MI, Kelly GM. Metabolic profile and differentiation potential of extraembryonic endoderm-like cells. Cell Death Discov 2018; 4:42. [PMID: 30302276 PMCID: PMC6158286 DOI: 10.1038/s41420-018-0102-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 08/20/2018] [Accepted: 08/28/2018] [Indexed: 02/08/2023] Open
Abstract
Glucose metabolism has a crucial role for providing substrates required to generate ATP and regulate the epigenetic landscape. We reported that F9 embryonal carcinoma stem-like cells require cytosolic reactive oxygen species to differentiate into extraembryonic endoderm; however, mitochondrial sources were not examined. To extend these studies, we examined the metabolic profile of early and late-passage F9 cells, and show that their ability to differentiate is similar, even though each population has dramatically different metabolic profiles. Differentiated early-passage cells relied on glycolysis, while differentiated late-passage cells transitioned towards oxidative phosphorylation (OXPHOS). Unexpectedly, electron transport chain protein stoichiometry was disrupted in differentiated late-passage cells, whereas genes encoding mitofusion 1 and 2, which promote mitochondrial fusion and favor OXPHOS, were upregulated in differentiated early-passage cells. Despite this, early-passage cells cultured under conditions to promote glycolysis showed enhanced differentiation, whereas promoting OXPHOS in late-passage cells showed a similar trend. Further analysis revealed that the distinct metabolic profiles seen between the two populations is largely associated with changes in genomic integrity, linking metabolism to passage number. Together, these results indicate that passaging has no effect on the potential for F9 cells to differentiate into extraembryonic endoderm; however, it does impact their metabolic profile. Thus, it is imperative to determine the molecular and metabolic status of a stem cell population before considering its utility as a therapeutic tool for regenerative medicine.
Collapse
Affiliation(s)
- Mohamed I Gatie
- 1Department of Biology, Collaborative Graduate Specialization in Developmental Biology, The University of Western Ontario, London, ON Canada
| | - Gregory M Kelly
- 1Department of Biology, Collaborative Graduate Specialization in Developmental Biology, The University of Western Ontario, London, ON Canada.,2Department of Paediatrics, The University of Western Ontario, London, ON Canada.,3Department of Physiology and Pharmacology, The University of Western Ontario, London, ON Canada.,Child Health Research Institute, London, ON Canada.,5Ontario Institute for Regenerative Medicine, Toronto, ON Canada
| |
Collapse
|
13
|
Schukking M, Miranda HC, Trujillo CA, Negraes PD, Muotri AR. Direct Generation of Human Cortical Organoids from Primary Cells. Stem Cells Dev 2018; 27:1549-1556. [PMID: 30142987 DOI: 10.1089/scd.2018.0112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The study of variations in human neurodevelopment and cognition is limited by the availability of experimental models. While animal models only partially recapitulate the human brain development, genetics, and heterogeneity, human-induced pluripotent stem cells can provide an attractive experimental alternative. However, cellular reprogramming and further differentiation techniques are costly and time-consuming and therefore, studies using this approach are often limited to a small number of samples. In this study, we describe a rapid and cost-effective method to reprogram somatic cells and the direct generation of cortical organoids in a 96-well format. Our data are a proof-of-principle that a large cohort of samples can be generated for experimental assessment of the human neural development.
Collapse
Affiliation(s)
- Monique Schukking
- 1 Department of Pediatrics, Rady Children's Hospital San Diego, University of California San Diego School of Medicine , La Jolla, California.,2 Department of Stem Cell Program, Department of Cellular and Molecular Medicine, University of California San Diego School of Medicine , La Jolla, California
| | - Helen C Miranda
- 1 Department of Pediatrics, Rady Children's Hospital San Diego, University of California San Diego School of Medicine , La Jolla, California.,2 Department of Stem Cell Program, Department of Cellular and Molecular Medicine, University of California San Diego School of Medicine , La Jolla, California
| | - Cleber A Trujillo
- 1 Department of Pediatrics, Rady Children's Hospital San Diego, University of California San Diego School of Medicine , La Jolla, California.,2 Department of Stem Cell Program, Department of Cellular and Molecular Medicine, University of California San Diego School of Medicine , La Jolla, California
| | - Priscilla D Negraes
- 1 Department of Pediatrics, Rady Children's Hospital San Diego, University of California San Diego School of Medicine , La Jolla, California.,2 Department of Stem Cell Program, Department of Cellular and Molecular Medicine, University of California San Diego School of Medicine , La Jolla, California
| | - Alysson R Muotri
- 1 Department of Pediatrics, Rady Children's Hospital San Diego, University of California San Diego School of Medicine , La Jolla, California.,2 Department of Stem Cell Program, Department of Cellular and Molecular Medicine, University of California San Diego School of Medicine , La Jolla, California.,3 Kavli Institute for Brain and Mind, University of California San Diego , La Jolla, California.,4 Center for Academic Research and Training in Anthropogeny (CARTA), University of California San Diego , La Jolla, California
| |
Collapse
|
14
|
Shafa M, Yang F, Fellner T, Rao MS, Baghbaderani BA. Human-Induced Pluripotent Stem Cells Manufactured Using a Current Good Manufacturing Practice-Compliant Process Differentiate Into Clinically Relevant Cells From Three Germ Layers. Front Med (Lausanne) 2018; 5:69. [PMID: 29600249 PMCID: PMC5862873 DOI: 10.3389/fmed.2018.00069] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/28/2018] [Indexed: 01/07/2023] Open
Abstract
The discovery of reprogramming and generation of human-induced pluripotent stem cells (iPSCs) has revolutionized the field of regenerative medicine and opened new opportunities in cell replacement therapies. While generation of iPSCs represents a significant breakthrough, the clinical relevance of iPSCs for cell-based therapies requires generation of high-quality specialized cells through robust and reproducible directed differentiation protocols. We have recently reported manufacturing of human iPSC master cell banks (MCB) under current good manufacturing practices (cGMPs). Here, we describe the clinical potential of human iPSCs generated using this cGMP-compliant process by differentiating them into the cells from all three embryonic germ layers including ectoderm, endoderm, and mesoderm. Most importantly, we have shown that our iPSC manufacturing process and cell culture system is not biased toward a specific lineage. Following controlled induction into a specific differentiation lineage, specialized cells with morphological and cellular characteristics of neural stem cells, definitive endoderm, and cardiomyocytes were developed. We believe that these cGMP-compliant iPSCs have the potential to make various clinically relevant products suitable for cell therapy applications.
Collapse
Affiliation(s)
- Mehdi Shafa
- Lonza Walkersville, Inc., Walkersville, MD, United States
| | - Fan Yang
- Lonza Walkersville, Inc., Walkersville, MD, United States
| | - Thomas Fellner
- Lonza Walkersville, Inc., Walkersville, MD, United States
| | - Mahendra S Rao
- NxCell Inc, Novato, CA, United States.,Q Therapeutics, Salt Lake City, UT, United States
| | | |
Collapse
|
15
|
J Siney E, Kurbatskaya K, Chatterjee S, Prasannan P, Mudher A, Willaime-Morawek S. Modelling neurodegenerative diseases in vitro: Recent advances in 3D iPSC technologies. ACTA ACUST UNITED AC 2018. [DOI: 10.3934/celltissue.2018.1.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
16
|
Zimmerlin L, Park TS, Zambidis ET. Capturing Human Naïve Pluripotency in the Embryo and in the Dish. Stem Cells Dev 2017; 26:1141-1161. [PMID: 28537488 DOI: 10.1089/scd.2017.0055] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Although human embryonic stem cells (hESCs) were first derived almost 20 years ago, it was only recently acknowledged that they share closer molecular and functional identity to postimplantation lineage-primed murine epiblast stem cells than to naïve preimplantation inner cell mass-derived mouse ESCs (mESCs). A myriad of transcriptional, epigenetic, biochemical, and metabolic attributes have now been described that distinguish naïve and primed pluripotent states in both rodents and humans. Conventional hESCs and human induced pluripotent stem cells (hiPSCs) appear to lack many of the defining hallmarks of naïve mESCs. These include important features of the naïve ground state murine epiblast, such as an open epigenetic architecture, reduced lineage-primed gene expression, and chimera and germline competence following injection into a recipient blastocyst-stage embryo. Several transgenic and chemical methods were recently reported that appear to revert conventional human PSCs to mESC-like ground states. However, it remains unclear if subtle deviations in global transcription, cell signaling dependencies, and extent of epigenetic/metabolic shifts in these various human naïve-reverted pluripotent states represent true functional differences or alternatively the existence of distinct human pluripotent states along a spectrum. In this study, we review the current understanding and developmental features of various human pluripotency-associated phenotypes and discuss potential biological mechanisms that may support stable maintenance of an authentic epiblast-like ground state of human pluripotency.
Collapse
Affiliation(s)
- Ludovic Zimmerlin
- 1 Institute for Cell Engineering, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins , Baltimore, Maryland
| | - Tea Soon Park
- 1 Institute for Cell Engineering, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins , Baltimore, Maryland
| | - Elias T Zambidis
- 1 Institute for Cell Engineering, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins , Baltimore, Maryland
| |
Collapse
|
17
|
Jaworska E, Kozlowska E, Switonski PM, Krzyzosiak WJ. Modeling simple repeat expansion diseases with iPSC technology. Cell Mol Life Sci 2016; 73:4085-100. [PMID: 27261369 PMCID: PMC11108530 DOI: 10.1007/s00018-016-2284-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/20/2016] [Accepted: 05/24/2016] [Indexed: 12/20/2022]
Abstract
A number of human genetic disorders, including Huntington's disease, myotonic dystrophy type 1, C9ORF72 form of amyotrophic lateral sclerosis and several spinocerebellar ataxias, are caused by the expansion of various microsatellite sequences in single implicated genes. The neurodegenerative and neuromuscular nature of the repeat expansion disorders considerably limits the access of researchers to appropriate cellular models of these diseases. This limitation, however, can be overcome by the application of induced pluripotent stem cell (iPSC) technology. In this paper, we review the current knowledge on the modeling of repeat expansion diseases with human iPSCs and iPSC-derived cells, focusing on the disease phenotypes recapitulated in these models. In subsequent sections, we provide basic practical knowledge regarding iPSC generation, characterization and differentiation into neurons. We also cover disease modeling in iPSCs, neuronal stem cells and specialized neuronal cultures. Furthermore, we also summarize the therapeutic potential of iPSC technology in repeat expansion diseases.
Collapse
Affiliation(s)
- Edyta Jaworska
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14 Str., 61-704, Poznan, Poland
| | - Emilia Kozlowska
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14 Str., 61-704, Poznan, Poland
| | - Pawel M Switonski
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14 Str., 61-704, Poznan, Poland
| | - Wlodzimierz J Krzyzosiak
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14 Str., 61-704, Poznan, Poland.
| |
Collapse
|
18
|
DeJonge RE, Liu XP, Deig CR, Heller S, Koehler KR, Hashino E. Modulation of Wnt Signaling Enhances Inner Ear Organoid Development in 3D Culture. PLoS One 2016; 11:e0162508. [PMID: 27607106 PMCID: PMC5015985 DOI: 10.1371/journal.pone.0162508] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/01/2016] [Indexed: 01/17/2023] Open
Abstract
Stem cell-derived inner ear sensory epithelia are a promising source of tissues for treating patients with hearing loss and dizziness. We recently demonstrated how to generate inner ear sensory epithelia, designated as inner ear organoids, from mouse embryonic stem cells (ESCs) in a self-organizing 3D culture. Here we improve the efficiency of this culture system by elucidating how Wnt signaling activity can drive the induction of otic tissue. We found that a carefully timed treatment with the potent Wnt agonist CHIR99021 promotes induction of otic vesicles—a process that was previously self-organized by unknown mechanisms. The resulting otic-like vesicles have a larger lumen size and contain a greater number of Pax8/Pax2-positive otic progenitor cells than organoids derived without the Wnt agonist. Additionally, these otic-like vesicles give rise to large inner ear organoids with hair cells whose morphological, biochemical and functional properties are indistinguishable from those of vestibular hair cells in the postnatal mouse inner ear. We conclude that Wnt signaling plays a similar role during inner ear organoid formation as it does during inner ear development in the embryo.
Collapse
Affiliation(s)
- Rachel E. DeJonge
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
| | - Xiao-Ping Liu
- Department of Otolaryngology, F.M. Kirby Neurobiology Center Boston Children’s Hospital, and Harvard Medical School, Boston, MA, 02115, United States of America
| | - Christopher R. Deig
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
| | - Stefan Heller
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Palo Alto, CA, 94305, United States of America
| | - Karl R. Koehler
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
- * E-mail: (EH); (KRK)
| | - Eri Hashino
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
- * E-mail: (EH); (KRK)
| |
Collapse
|
19
|
Miyagishima KJ, Wan Q, Corneo B, Sharma R, Lotfi MR, Boles NC, Hua F, Maminishkis A, Zhang C, Blenkinsop T, Khristov V, Jha BS, Memon OS, D'Souza S, Temple S, Miller SS, Bharti K. In Pursuit of Authenticity: Induced Pluripotent Stem Cell-Derived Retinal Pigment Epithelium for Clinical Applications. Stem Cells Transl Med 2016; 5:1562-1574. [PMID: 27400791 PMCID: PMC5070511 DOI: 10.5966/sctm.2016-0037] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/18/2016] [Indexed: 12/12/2022] Open
Abstract
For effective treatment, induced pluripotent stem cell (iPSC)-retinal pigment epithelium (RPE) must recapitulate the physiology of native human RPE cells. A set of physiologically relevant functional assays that assess the polarized functional activity and maturation state of the intact RPE monolayer is provided. The study data show that donor-to-donor variability exceeds the tissue-to-tissue variability for a given donor and provides, for the first time, criteria necessary to identify iPSC-RPE cells most suitable for clinical application. Induced pluripotent stem cells (iPSCs) can be efficiently differentiated into retinal pigment epithelium (RPE), offering the possibility of autologous cell replacement therapy for retinal degeneration stemming from RPE loss. The generation and maintenance of epithelial apical-basolateral polarity is fundamental for iPSC-derived RPE (iPSC-RPE) to recapitulate native RPE structure and function. Presently, no criteria have been established to determine clonal or donor based heterogeneity in the polarization and maturation state of iPSC-RPE. We provide an unbiased structural, molecular, and physiological evaluation of 15 iPSC-RPE that have been derived from distinct tissues from several different donors. We assessed the intact RPE monolayer in terms of an ATP-dependent signaling pathway that drives critical aspects of RPE function, including calcium and electrophysiological responses, as well as steady-state fluid transport. These responses have key in vivo counterparts that together help determine the homeostasis of the distal retina. We characterized the donor and clonal variation and found that iPSC-RPE function was more significantly affected by the genetic differences between different donors than the epigenetic differences associated with different starting tissues. This study provides a reference dataset to authenticate genetically diverse iPSC-RPE derived for clinical applications. Significance The retinal pigment epithelium (RPE) is essential for maintaining visual function. RPE derived from human induced pluripotent stem cells (iPSC-RPE) offer a promising cell-based transplantation therapy for slowing or rescuing RPE-induced visual function loss. For effective treatment, iPSC-RPE must recapitulate the physiology of native human RPE. A set of physiologically relevant functional assays are provided that assess the polarized functional activity and maturation state of the intact RPE monolayer. The present data show that donor-to-donor variability exceeds the tissue-to-tissue variability for a given donor and provides, for the first time, criteria necessary to identify iPSC-RPE most suitable for clinical application.
Collapse
Affiliation(s)
- Kiyoharu J Miyagishima
- Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Qin Wan
- Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Barbara Corneo
- Columbia Stem Cell Core Facility, Columbia University Medical Center, New York, New York, USA
| | - Ruchi Sharma
- Unit on Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mostafa R Lotfi
- Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Fang Hua
- Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Arvydas Maminishkis
- Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Congxiao Zhang
- Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Timothy Blenkinsop
- Department of Development and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Vladimir Khristov
- Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Balendu S Jha
- Unit on Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Omar S Memon
- Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Sunita D'Souza
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sally Temple
- Neural Stem Cell Institute, Rensselaer, New York, USA
| | - Sheldon S Miller
- Section on Epithelial and Retinal Physiology and Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Kapil Bharti
- Unit on Ocular and Stem Cell Translational Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
20
|
Begum AN, Hong Y. A Novel Method for the Generation of Region-Specific Neurons and Neural Networks from Human Pluripotent Stem Cells. JOURNAL OF STEM CELL RESEARCH & THERAPY 2016; 1:1-3. [PMID: 33409004 PMCID: PMC7784482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Stem cell-based neuronal differentiation has provided a unique opportunity for disease modeling and regenerative medicine. We have reported a novel culture condition and method for generating neuronal progenitors and neural networks from human embryonic and induced pluripotent stem cells without any genetic manipulation. Neurospheres generated under 10% CO2 with Supplemented Knockout Serum Replacement Medium (SKSRM) had doubled the expression of NESTIN, PAX6 and FOXG1 genes compared to the neurospheres generated under 5% CO2. Furthermore, an additional step (AdStep) was introduced to fragment the neurospheres, which increased the expression of neuronal progenitor genes NEUROD1, NEUROG2, TBR1, TBR2, and NOTCH1 and the formation of the neuroepithelial-type cells. With the supplements, neuronal progenitors further differentiated into different layers of cortical, pyramidal, GABAergic, glutamatergic, cholinergic, dopaminergic and purkinje neurons within 27-40 days, which is faster than traditional neurodifferentiation protocols (42-60 days). Furthermore, our in vivo studies indicated that neuronal progenitors derived under our culture conditions with "AdStep" showed significantly increased neurogenesis in Severe Combined Immunodeficiency (SCID) mouse brains. This neurosphere-based neurodifferentiation protocol is a valuable tool for studies neurogenesis, neuronal transplantation and high throughput screening assays.
Collapse
Affiliation(s)
- Aynun N Begum
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, USA
| | - Yiling Hong
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, USA
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
21
|
Peng SP, Copray S. Comparison of Human Primary with Human iPS Cell-Derived Dopaminergic Neuron Grafts in the Rat Model for Parkinson's Disease. Stem Cell Rev Rep 2016; 12:105-20. [PMID: 26438376 PMCID: PMC4720696 DOI: 10.1007/s12015-015-9623-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Neuronal degeneration within the substantia nigra and the loss of the dopaminergic nigro-striatal pathway are the major hallmarks of Parkinson's disease (PD). Grafts of foetal ventral mesencephalic (VM) dopaminergic (DA) neurons into the striatum have been shown to be able to restore striatal dopamine levels and to improve overall PD symptoms. However, human foetus-derived cell grafts are not feasible for clinical application. Autologous induced pluripotent stem cell (iPS cell)-derived DA neurons are emerging as an unprecedented alternative. In this review, we summarize and compare the efficacy of human iPS cell-derived DA neuron grafts to restore normal behaviour in a rat model for PD with that of human foetal primary DA neurons. The differences we observed in the efficacy to restore normal function between the 2 types of DA neuron grafts could be ascribed to intrinsic properties of the iPS cell-derived DA neurons that critically affected survival and proper neurite extension in the striatum after implantation.
Collapse
Affiliation(s)
- Su-Ping Peng
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
- Department of Neuroscience, Medical Physiology, University Medical Center Groningen, University of Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Sjef Copray
- Department of Neuroscience, Medical Physiology, University Medical Center Groningen, University of Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
22
|
Begum AN, Guoynes C, Cho J, Hao J, Lutfy K, Hong Y. Rapid generation of sub-type, region-specific neurons and neural networks from human pluripotent stem cell-derived neurospheres. Stem Cell Res 2015; 15:731-741. [PMID: 26613348 DOI: 10.1016/j.scr.2015.10.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 09/26/2015] [Accepted: 10/21/2015] [Indexed: 10/22/2022] Open
Abstract
Stem cell-based neuronal differentiation has provided a unique opportunity for disease modeling and regenerative medicine. Neurospheres are the most commonly used neuroprogenitors for neuronal differentiation, but they often clump in culture, which has always represented a challenge for neurodifferentiation. In this study, we report a novel method and defined culture conditions for generating sub-type or region-specific neurons from human embryonic and induced pluripotent stem cells derived neurosphere without any genetic manipulation. Round and bright-edged neurospheres were generated in a supplemented knockout serum replacement medium (SKSRM) with 10% CO2, which doubled the expression of the NESTIN, PAX6 and FOXG1 genes compared with those cultured with 5% CO2. Furthermore, an additional step (AdSTEP) was introduced to fragment the neurospheres and facilitate the formation of a neuroepithelial-type monolayer that we termed the "neurosphederm". The large neural tube-type rosette (NTTR) structure formed from the neurosphederm, and the NTTR expressed higher levels of the PAX6, SOX2 and NESTIN genes compared with the neuroectoderm-derived neuroprogenitors. Different layers of cortical, pyramidal, GABAergic, glutamatergic, cholinergic neurons appeared within 27 days using the neurosphederm, which is a shorter period than in traditional neurodifferentiation-protocols (42-60 days). With additional supplements and timeline dopaminergic and Purkinje neurons were also generated in culture too. Furthermore, our in vivo results indicated that the fragmented neurospheres facilitated significantly better neurogenesis in severe combined immunodeficiency (SCID) mouse brains compared with the non-fragmented neurospheres. Therefore, this neurosphere-based neurodifferentiation protocol is a valuable tool for studies of neurodifferentiation, neuronal transplantation and high throughput screening assays.
Collapse
Affiliation(s)
- Aynun N Begum
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Caleigh Guoynes
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Jane Cho
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Jijun Hao
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Kabirullah Lutfy
- College of Pharmacy, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Yiling Hong
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766-1854, USA; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA.
| |
Collapse
|
23
|
Crook JM, Wallace G, Tomaskovic-Crook E. The potential of induced pluripotent stem cells in models of neurological disorders: implications on future therapy. Expert Rev Neurother 2015; 15:295-304. [PMID: 25664599 DOI: 10.1586/14737175.2015.1013096] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
There is an urgent need for new and advanced approaches to modeling the pathological mechanisms of complex human neurological disorders. This is underscored by the decline in pharmaceutical research and development efficiency resulting in a relative decrease in new drug launches in the last several decades. Induced pluripotent stem cells represent a new tool to overcome many of the shortcomings of conventional methods, enabling live human neural cell modeling of complex conditions relating to aberrant neurodevelopment, such as schizophrenia, epilepsy and autism as well as age-associated neurodegeneration. This review considers the current status of induced pluripotent stem cell-based modeling of neurological disorders, canvassing proven and putative advantages, current constraints, and future prospects of next-generation culture systems for biomedical research and translation.
Collapse
Affiliation(s)
- Jeremy Micah Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Squires Way, Fairy Meadow, New South Wales 2519, Australia
| | | | | |
Collapse
|
24
|
Nelakanti RV, Kooreman NG, Wu JC. Teratoma formation: a tool for monitoring pluripotency in stem cell research. CURRENT PROTOCOLS IN STEM CELL BIOLOGY 2015; 32:4A.8.1-4A.8.17. [PMID: 25640819 PMCID: PMC4402211 DOI: 10.1002/9780470151808.sc04a08s32] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
This unit describes protocols for evaluating the pluripotency of embryonic and induced pluripotent stem cells using a teratoma formation assay. Cells are prepared for injection and transplanted into immunodeficient mice at the gastrocnemius muscle, a site well suited for teratoma growth and surgical access. Teratomas that form from the cell transplants are explanted, fixed in paraformaldehyde, and embedded in paraffin. These preserved samples are sectioned, stained, and analyzed. Pluripotency of a cell line is confirmed by whether the teratoma contains tissues derived from each of the embryonic germ layers: endoderm, mesoderm, and ectoderm. Alternatively, explanted and fixed teratomas can be cryopreserved for immunohistochemistry, which allows for more detailed identification of specific tissue types present in the samples.
Collapse
Affiliation(s)
- Raman V Nelakanti
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California
- Departments of Medicine and Radiology (Molecular Imaging Program), Stanford University School of Medicine, Stanford, California
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Nigel G Kooreman
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California
- Departments of Medicine and Radiology (Molecular Imaging Program), Stanford University School of Medicine, Stanford, California
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California
- Departments of Medicine and Radiology (Molecular Imaging Program), Stanford University School of Medicine, Stanford, California
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
25
|
Zare M, Soleimani M, Akbarzadeh A, Bakhshandeh B, Aghaee-Bakhtiari SH, Zarghami N. A Novel Protocol to Differentiate Induced Pluripotent Stem Cells by Neuronal microRNAs to Provide a Suitable Cellular Model. Chem Biol Drug Des 2015; 86:232-8. [DOI: 10.1111/cbdd.12485] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Revised: 11/15/2014] [Accepted: 11/17/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Mehrak Zare
- Department of Cellular Biology; Stem Cell Technology Research Center; Tehran Iran
- Neurosciences Research Center; Tabriz University of Medical Sciences; Tabriz Iran
| | - Masoud Soleimani
- Department of Haematology; School of Medicine; Tarbiat Modares University; Tehran Iran
| | - Abolfazl Akbarzadeh
- Department of Medical Nanotechnology; Faculty of Advanced Medical Science; Tabriz University of Medical Sciences; Tabriz Iran
| | - Behnaz Bakhshandeh
- Department of Biotechnology; College of Science; University of Tehran; Tehran Iran
| | | | - Nosratollah Zarghami
- Department of Clinical Biochemistry and Laboratory; Faculty of Medicine; Tabriz University of Medical Sciences; Tabriz Iran
- Department of Clinical Biochemistry; Radiopharmacy Lab; Drug Applied Research Center; Tabriz University of Medical Sciences; Tabriz Iran
| |
Collapse
|
26
|
Ren X, Ott HC. On the road to bioartificial organs. Pflugers Arch 2014; 466:1847-57. [PMID: 24691559 DOI: 10.1007/s00424-014-1504-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 03/18/2014] [Indexed: 01/08/2023]
Abstract
Biological organs are highly orchestrated systems with well-coordinated positioning, grouping, and interaction of different cell types within their specialized extracellular environment. Bioartificial organs are intended to be functional replacements of native organs generated through bioengineering techniques and hold the potential to alleviate donor organ shortage for transplantation. The development, production, and evaluation of such bioartificial organs require synergistic efforts of biology, material science, engineering, and medicine. In this review, we highlight the emerging platforms enabling structured assembly of multiple cell types into functional grafts and discuss recent advances and challenges in the development of bioartificial organs, including cell sources, in vitro organ culture, in vivo evaluation, and clinical considerations.
Collapse
Affiliation(s)
- X Ren
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | |
Collapse
|
27
|
Srikanth P, Young-Pearse TL. Stem cells on the brain: modeling neurodevelopmental and neurodegenerative diseases using human induced pluripotent stem cells. J Neurogenet 2014; 28:5-29. [PMID: 24628482 PMCID: PMC4285381 DOI: 10.3109/01677063.2014.881358] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Seven years have passed since the initial report of the generation of induced pluripotent stem cells (iPSCs) from adult human somatic cells, and in the intervening time the field of neuroscience has developed numerous disease models using this technology. Here, we review progress in the field and describe both the advantages and potential pitfalls of modeling neurodegenerative and neurodevelopmental diseases using this technology. We include tables with information on neural differentiation protocols and studies that developed human iPSC lines to model neurological diseases. We also discuss how one can: investigate effects of genetic mutations with iPSCs, examine cell fate-specific phenotypes, best determine the specificity of a phenotype, and bring in vivo relevance to this in vitro technique.
Collapse
Affiliation(s)
- Priya Srikanth
- Center for Neurologic Diseases, Brigham and Women's Hospital , Boston, Massachusetts , USA
| | | |
Collapse
|
28
|
Offen N, Flemming J, Kamawal H, Ahmad R, Wolber W, Geis C, Zaehres H, Schöler HR, Ehrenreich H, Müller AM, Sirén AL. Effects of erythropoietin in murine-induced pluripotent cell-derived panneural progenitor cells. Mol Med 2013; 19:399-408. [PMID: 24408113 DOI: 10.2119/molmed.2013.00136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 11/06/2013] [Indexed: 11/06/2022] Open
Abstract
Induced cell fate changes by reprogramming of somatic cells offers an efficient strategy to generate autologous pluripotent stem (iPS) cells from any adult cell type. The potential of iPS cells to differentiate into various cell types is well established, however the efficiency to produce functional neurons from iPS cells remains modest. Here, we generated panneural progenitor cells (pNPCs) from mouse iPS cells and investigated the effect of the neurotrophic growth factor erythropoietin (EPO) on their survival, proliferation and neurodifferentiation. Under neural differentiation conditions, iPS-derived pNPCs gave rise to microtubule-associated protein-2 positive neuronlike cells (34% to 43%) and platelet-derived growth factor receptor positive oligodendrocytelike cells (21% to 25%) while less than 1% of the cells expressed the astrocytic marker glial fibrillary acidic protein. Neuronlike cells generated action potentials and developed active presynaptic terminals. The pNPCs expressed EPO receptor (EPOR) mRNA and displayed functional EPOR signaling. In proliferating cultures, EPO (0.1-3 U/mL) slightly improved pNPC survival but reduced cell proliferation and neurosphere formation in a concentration-dependent manner. In differentiating cultures EPO facilitated neurodifferentiation as assessed by the increased number of β-III-tubulin positive neurons. Our results show that EPO inhibits iPS pNPC self-renewal and promotes neurogenesis.
Collapse
Affiliation(s)
- Nils Offen
- Department of Neurosurgery, University of Würzburg, Würzburg, Germany
| | - Johannes Flemming
- Department of Neurosurgery, University of Würzburg, Würzburg, Germany
| | - Hares Kamawal
- Department of Neurosurgery, University of Würzburg, Würzburg, Germany
| | - Ruhel Ahmad
- Center for Experimental Molecular Medicine (ZEMM), University of Würzburg, Würzburg, Germany
| | - Wanja Wolber
- Department of Neurosurgery, University of Würzburg, Würzburg, Germany
| | - Christian Geis
- Department of Neurology, University of Würzburg, Würzburg, Germany Department of Neurology and Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Holm Zaehres
- Department of Cell and Developmental Biology, Max-Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max-Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Albrecht M Müller
- Center for Experimental Molecular Medicine (ZEMM), University of Würzburg, Würzburg, Germany
| | - Anna-Leena Sirén
- Department of Neurosurgery, University of Würzburg, Würzburg, Germany
| |
Collapse
|
29
|
Later passages of neural progenitor cells from neonatal brain are more permissive for human cytomegalovirus infection. J Virol 2013; 87:10968-79. [PMID: 23903847 DOI: 10.1128/jvi.01120-13] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Congenital human cytomegalovirus (HCMV) infection is the most frequent infectious cause of birth defects, primarily neurological disorders. Neural progenitor/stem cells (NPCs) are the major cell type in the subventricular zone and are susceptible to HCMV infection. In culture, the differentiation status of NPCs may change with passage, which in turn may alter susceptibility to virus infection. Previously, only early-passage (i.e., prior to passage 9) NPCs were studied and shown to be permissive to HCMV infection. In this study, NPC cultures derived at different gestational ages were evaluated after short (passages 3 to 6) and extended (passages 11 to 20) in vitro passages for biological and virological parameters (i.e., cell morphology, expression of NPC markers and HCMV receptors, viral entry efficiency, viral gene expression, virus-induced cytopathic effect, and release of infectious progeny). These parameters were not significantly influenced by the gestational age of the source tissues. However, extended-passage cultures showed evidence of initiation of differentiation, increased viral entry, and more efficient production of infectious progeny. These results confirm that NPCs are fully permissive for HCMV infection and that extended-passage NPCs initiate differentiation and are more permissive for HCMV infection. Later-passage NPCs being differentiated and more permissive for HCMV infection suggest that HCMV infection in fetal brain may cause more neural cell loss and give rise to severe neurological disabilities with advancing brain development.
Collapse
|
30
|
Honda A, Hatori M, Hirose M, Honda C, Izu H, Inoue K, Hirasawa R, Matoba S, Togayachi S, Miyoshi H, Ogura A. Naive-like conversion overcomes the limited differentiation capacity of induced pluripotent stem cells. J Biol Chem 2013; 288:26157-26166. [PMID: 23880763 DOI: 10.1074/jbc.m113.502492] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Although induced pluripotent stem (iPS) cells are indistinguishable from ES cells in their expression of pluripotent markers, their differentiation into targeted cells is often limited. Here, we examined whether the limited capacity of iPS cells to differentiate into neural lineage cells could be mitigated by improving their base-line level of pluripotency, i.e. by converting them into the so-called "naive" state. In this study, we used rabbit iPS and ES cells because of the easy availability of both cell types and their typical primed state characters. Repeated passages of the iPS cells permitted their differentiation into early neural cell types (neural stem cells, neurons, and glial astrocytes) with efficiencies similar to ES cells. However, unlike ES cells, their ability to differentiate later into neural cells (oligodendrocytes) was severely compromised. In contrast, after these iPS cells had been converted to a naive-like state, they readily differentiated into mature oligodendrocytes developing characteristic ramified branches, which could not be attained even with ES cells. These results suggest that the naive-like conversion of iPS cells might endow them with a higher differentiation capacity.
Collapse
Affiliation(s)
- Arata Honda
- From the Organization for Promotion of Tenure Track, University of Miyazaki, 5200, Kibara, Kiyotake, Miyazaki 889-1692,; the RIKEN BioResource Center, Tsukuba, Ibaraki 305-0074,; PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012,.
| | | | | | - Chizumi Honda
- From the Organization for Promotion of Tenure Track, University of Miyazaki, 5200, Kibara, Kiyotake, Miyazaki 889-1692
| | - Haruna Izu
- From the Organization for Promotion of Tenure Track, University of Miyazaki, 5200, Kibara, Kiyotake, Miyazaki 889-1692
| | - Kimiko Inoue
- the RIKEN BioResource Center, Tsukuba, Ibaraki 305-0074,; the Graduate School of Life and Environmental Science, University of Tsukuba, Tsukuba, Ibaraki 305-8572, and
| | | | - Shogo Matoba
- the RIKEN BioResource Center, Tsukuba, Ibaraki 305-0074
| | | | | | - Atsuo Ogura
- the RIKEN BioResource Center, Tsukuba, Ibaraki 305-0074,; the Graduate School of Life and Environmental Science, University of Tsukuba, Tsukuba, Ibaraki 305-8572, and; the Center for Disease Biology and Integrative Medicine, 5 Faculty of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
31
|
Generation of inner ear sensory epithelia from pluripotent stem cells in 3D culture. Nature 2013; 500:217-21. [PMID: 23842490 PMCID: PMC3739998 DOI: 10.1038/nature12298] [Citation(s) in RCA: 273] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Accepted: 05/13/2013] [Indexed: 02/06/2023]
Abstract
The inner ear contains sensory epithelia that detect head movements, gravity and sound. It is unclear how to derive these sensory epithelia from pluripotent stem cells, a process which will be critical for modeling inner ear disorders or developing cell-based therapies for profound hearing loss and balance disorders1,2. To date, attempts to derive inner ear mechanosensitive hair cells and sensory neurons have resulted in inefficient or incomplete phenotypic conversion of stem cells into inner ear-like cells3–7. A key insight lacking from these previous studies is the importance of the non-neural and pre-placodal ectoderm, two critical precursors during inner ear development8–11. Here we report the step-wise differentiation of inner ear sensory epithelia from mouse embryonic stem cells (ESCs) in three-dimensional culture12,13. We show that by recapitulating in vivo development with precise temporal control of BMP, TGFβ and FGF signaling, ESC aggregates transform sequentially into non-neural, pre-placodal and otic placode-like epithelia. Remarkably, in a self-organized process that mimics normal development, vesicles containing prosensory cells emerge from the presumptive otic placodes and give rise to hair cells bearing stereocilia bundles and a kinocilium. Moreover, these stem cell-derived hair cells exhibit functional properties of native mechanosensitive hair cells and form specialized synapses with sensory neurons that have also arisen from ESCs in the culture. Finally, we demonstrate how these vesicles are structurally and biochemically comparable to developing vestibular end organs. Our data thus establish a novel in vitro model of inner ear differentiation that can be used to gain deeper insight into inner ear development and disorder.
Collapse
|
32
|
Chung CY, Yang JT, Kuo YC. Polybutylcyanoacrylate nanoparticle-mediated neurotrophin-3 gene delivery for differentiating iPS cells into neurons. Biomaterials 2013; 34:5562-70. [PMID: 23623427 DOI: 10.1016/j.biomaterials.2013.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 04/03/2013] [Indexed: 12/12/2022]
Abstract
Guided neuronal differentiation of induced pluripotent stem cells (iPSCs) with genetic regulation is an important issue in biomedical research and in clinical practice for nervous regeneration and repair. To enhance the intracellular delivery of plasmid DNA (pDNA), polybutylcyanoacrylate (PBCA) nanoparticles (NPs) were employed to mediate the transport of neurotrophin-3 (NT-3) into iPSCs. The ability of iPSCs to differentiate into neuronal lineages was shown by immunofluorescent staining, western blotting, and flow cytometry. By transmission electron microscopy, we found that PBCA NPs could efficiently grasp pDNA, thereby increasing the particle size and conferring a negative surface charge. In addition, the treatments with PBCA NP/NT-3 complexes enhanced the expression of NT-3, TrkC, NH-H, NSE, and PSD95 by differentiating iPSCs. Neurons produced from iPSCs were incapable of returning to pluripotency, demonstrating with a series of differentiation scheme for adipogenesis and osteogenesis. The pretreatment with PBCA NP/NT-3 complexes can be one of critical biotechnologies and effective delivery systems in gene transfection to accelerate the differentiation of iPSCs into neurons.
Collapse
Affiliation(s)
- Chiu-Yen Chung
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi 62102, Taiwan, ROC
| | | | | |
Collapse
|
33
|
Zeng X, Couture LA. Pluripotent stem cells for Parkinson's disease: progress and challenges. Stem Cell Res Ther 2013; 4:25. [PMID: 23672848 PMCID: PMC3707048 DOI: 10.1186/scrt173] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is a common debilitating neurodegenerative disease. The motor symptoms of PD are caused mainly by a progressive loss of dopaminergic neurons from the substania nigra, resulting in a loss of dopamine production. Current therapies are palliative and, in the long term, ineffective. In addition, some can result in significant clinical side effects. The relatively localized pathology of PD makes it an ideal candidate for cell replacement therapy. Initial efforts focused on fetal cell transplantation, and significant clinical benefit lasting more than 10 years has been reported in some cases. However, the approach is controversial and results have been inconsistent. Inherent limitations of this approach for widespread use are the limited availability and variability of transplant material. In contrast, the self-renewal and differentiation potential of human pluripotent stem cells (hPSCs) make them a promising alternative cell source for cell replacement therapy for PD. Efforts in the past decade have demonstrated that hPSCs can be induced to differentiate in culture to functional dopaminergic neurons. Studies in delivering these cells into PD animal models have demonstrated survival, engraftment, and behavioral deficit improvements. Several groups are developing these cells with clinical trials in mind. Here, we review the state of the technology and consider the suitability of current manufacturing processes, cell purity, and tumorgenicity for clinical testing.
Collapse
|
34
|
Kramer AS, Harvey AR, Plant GW, Hodgetts SI. Systematic Review of Induced Pluripotent Stem Cell Technology as a Potential Clinical Therapy for Spinal Cord Injury. Cell Transplant 2013; 22:571-617. [DOI: 10.3727/096368912x655208] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Transplantation therapies aimed at repairing neurodegenerative and neuropathological conditions of the central nervous system (CNS) have utilized and tested a variety of cell candidates, each with its own unique set of advantages and disadvantages. The use and popularity of each cell type is guided by a number of factors including the nature of the experimental model, neuroprotection capacity, the ability to promote plasticity and guided axonal growth, and the cells' myelination capability. The promise of stem cells, with their reported ability to give rise to neuronal lineages to replace lost endogenous cells and myelin, integrate into host tissue, restore functional connectivity, and provide trophic support to enhance and direct intrinsic regenerative ability, has been seen as a most encouraging step forward. The advent of the induced pluripotent stem cell (iPSC), which represents the ability to “reprogram” somatic cells into a pluripotent state, hails the arrival of a new cell transplantation candidate for potential clinical application in therapies designed to promote repair and/or regeneration of the CNS. Since the initial development of iPSC technology, these cells have been extensively characterized in vitro and in a number of pathological conditions and were originally reported to be equivalent to embryonic stem cells (ESCs). This review highlights emerging evidence that suggests iPSCs are not necessarily indistinguishable from ESCs and may occupy a different “state” of pluripotency with differences in gene expression, methylation patterns, and genomic aberrations, which may reflect incomplete reprogramming and may therefore impact on the regenerative potential of these donor cells in therapies. It also highlights the limitations of current technologies used to generate these cells. Moreover, we provide a systematic review of the state of play with regard to the use of iPSCs in the treatment of neurodegenerative and neuropathological conditions. The importance of balancing the promise of this transplantation candidate in the light of these emerging properties is crucial as the potential application in the clinical setting approaches. The first of three sections in this review discusses (A) the pathophysiology of spinal cord injury (SCI) and how stem cell therapies can positively alter the pathology in experimental SCI. Part B summarizes (i) the available technologies to deliver transgenes to generate iPSCs and (ii) recent data comparing iPSCs to ESCs in terms of characteristics and molecular composition. Lastly, in (C) we evaluate iPSC-based therapies as a candidate to treat SCI on the basis of their neurite induction capability compared to embryonic stem cells and provide a summary of available in vivo data of iPSCs used in SCI and other disease models.
Collapse
Affiliation(s)
- Anne S. Kramer
- Spinal Cord Repair Laboratory, School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, Western Australia
| | - Alan R. Harvey
- Spinal Cord Repair Laboratory, School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, Western Australia
| | - Giles W. Plant
- Stanford Partnership for Spinal Cord Injury and Repair, Stanford Institute for Neuro-Innovation and Translational Neurosciences, Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Stuart I. Hodgetts
- Spinal Cord Repair Laboratory, School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, Western Australia
| |
Collapse
|
35
|
Yang JY, Mumaw JL, Liu Y, Stice SL, West FD. SSEA4-positive pig induced pluripotent stem cells are primed for differentiation into neural cells. Cell Transplant 2012; 22:945-59. [PMID: 23043799 DOI: 10.3727/096368912x657279] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Neural cells derived from induced pluripotent stem cells (iPSCs) have the potential for autologous cell therapies in treating patients with severe neurological disorders or injury. However, further study of efficacy and safety are needed in large animal preclinical models that have similar neural anatomy and physiology to humans such as the pig. The pig model for pluripotent stem cell therapy has been made possible for the first time with the development of pig iPSCs (piPSCs) capable of in vitro and in vivo differentiation into tissues of all three germ layers. Still, the question remains if piPSCs are capable of undergoing robust neural differentiation using a system similar to those being used with human iPSCs. In this study, we generated a new line of piPSCs from fibroblast cells that expressed pluripotency markers and were capable of embryoid body differentiation into all three germ layers. piPSCs demonstrated robust neural differentiation forming βIII-TUB/MAP2+ neurons, GFAP+ astrocytes, and O4+ oligodendrocytes and demonstrated strong upregulation of neural cell genes representative of all three major neural lineages of the central nervous system. In the presence of motor neuron signaling factors, piPSC-derived neurons showed expression of transcription factors associated with motor neuron differentiation (HB9 and ISLET1). Our findings demonstrate that SSEA4 expression is required for piPSCs to differentiate into neurons, astrocytes, and oligodendrocytes and furthermore develop specific neuronal subtypes. This indicates that the pigs can fill the need for a powerful model to study autologous neural iPSC therapies in a system similar to humans.
Collapse
Affiliation(s)
- Jeong-Yeh Yang
- Regenerative Bioscience Center, University of Georgia, Rhodes Center for Animal and Dairy Science, Athens, GA 30602-2771, USA
| | | | | | | | | |
Collapse
|