1
|
Okubo C, Nakamura M, Sato M, Shichino Y, Mito M, Takashima Y, Iwasaki S, Takahashi K. EIF3D safeguards the homeostasis of key signaling pathways in human primed pluripotency. SCIENCE ADVANCES 2025; 11:eadq5484. [PMID: 40203091 PMCID: PMC11980838 DOI: 10.1126/sciadv.adq5484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Although pluripotent stem cell (PSC) properties, such as differentiation and infinite proliferation, have been well documented within the frameworks of transcription factor networks, epigenomes, and signal transduction, they remain unclear and fragmented. Directing attention toward translational regulation as a bridge between these events can yield additional insights into previously unexplained mechanisms. Our functional CRISPR interference screen-based approach revealed that EIF3D, a translation initiation factor, is crucial for maintaining primed pluripotency. Loss of EIF3D disrupted the balance of pluripotency-associated signaling pathways, thereby compromising primed pluripotency. Moreover, EIF3D ensured robust proliferation by controlling the translation of various p53 regulators, which maintain low p53 activity in the undifferentiated state. In this way, EIF3D-mediated translation contributes to tuning the homeostasis of the primed pluripotency networks, ensuring the maintenance of an undifferentiated state with high proliferative potential. This study provides further insights into the translation network in maintaining pluripotency.
Collapse
Affiliation(s)
- Chikako Okubo
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Michiko Nakamura
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Masae Sato
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Yuichi Shichino
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Saitama, 351-0198, Japan
| | - Mari Mito
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Saitama, 351-0198, Japan
| | - Yasuhiro Takashima
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Shintaro Iwasaki
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Saitama, 351-0198, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, 277-8561, Japan
| | - Kazutoshi Takahashi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| |
Collapse
|
2
|
Iida J, Kotani K, Murata K, Hakamada K, Maihemuti W, Mandai Y, Hiraoka Y, Minatoya K, Masumoto H. Retention of locally injected human iPS cell-derived cardiomyocytes into the myocardium using hydrolyzed gelatin. Sci Rep 2025; 15:4635. [PMID: 39920228 PMCID: PMC11806045 DOI: 10.1038/s41598-025-87885-w] [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: 10/06/2024] [Accepted: 01/22/2025] [Indexed: 02/09/2025] Open
Abstract
This study explored the impact of hydrolyzed gelatin (HG) concentration on the retention and therapeutic efficacy of human iPS cell-derived cardiomyocytes (hiPSC-CMs) when injected into the myocardium. The solubility of HG allows precise control over its concentration, influencing the distribution and leakage of injected solutions, which may affect therapeutic outcomes. Using both ex vivo and in vivo rat models, we investigated how varying HG concentrations affect the retention of solution and diffusion within the myocardium. In ex vivo static rat hearts, 10% HG minimized leakage but allowed significant diffusion. However, in pulsating in vivo hearts, 20% HG provided the best retention. In a rat myocardial infarction model, hiPSC-CMs suspended in 20% HG resulted in the highest cell retention. Echocardiogram showed a significant increase in the ejection fraction two weeks after transplantation compared to before transplantation. Additionally, cardiac magnetic resonance imaging (MRI) revealed that the ejection fraction was significantly higher than that of the sham group four weeks after transplantation. These findings suggest that optimizing HG concentration is crucial for enhancing the retention and therapeutic efficacy of hiPSC-CM transplants in treating heart disease.
Collapse
Affiliation(s)
- Jun Iida
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojimaminami-cho, Chuo-ku, Kobe, 650-0047, Japan
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuki Kotani
- Biomedical Department, R&D Center, Nitta Gelatin Inc, Yao, Japan
| | - Kozue Murata
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojimaminami-cho, Chuo-ku, Kobe, 650-0047, Japan
| | - Keisuke Hakamada
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Wusiman Maihemuti
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojimaminami-cho, Chuo-ku, Kobe, 650-0047, Japan
| | - Yoshinobu Mandai
- Biomedical Department, R&D Center, Nitta Gelatin Inc, Yao, Japan
| | - Yosuke Hiraoka
- Biomedical Department, R&D Center, Nitta Gelatin Inc, Yao, Japan
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidetoshi Masumoto
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojimaminami-cho, Chuo-ku, Kobe, 650-0047, Japan.
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
3
|
Teale MA, Schneider SL, Seidel S, Krasenbrink J, Poggel M, Eibl D, Sousa MFQ, Eibl R. Expansion of induced pluripotent stem cells under consideration of bioengineering aspects: part 2. Appl Microbiol Biotechnol 2025; 109:38. [PMID: 39912924 PMCID: PMC11802622 DOI: 10.1007/s00253-024-13373-2] [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: 07/26/2024] [Revised: 11/21/2024] [Accepted: 11/29/2024] [Indexed: 02/07/2025]
Abstract
The manufacturing of allogeneic cell therapeutics based on human-induced pluripotent stem cells (hiPSCs) holds considerable potential to revolutionize the accessibility and affordability of modern healthcare. However, achieving the cell yields necessary to ensure robust production hinges on identifying suitable and scalable single-use (SU) bioreactor systems. While specific stirred SU bioreactor types have demonstrated proficiency in supporting hiPSC expansion at L-scale, others, notably instrumented SU multiplate and fixed-bed bioreactors, remain relatively unexplored. By characterizing these bioreactors using both computational fluid dynamics and experimental bioengineering methods, operating ranges were identified for the Xpansion® 10 and Ascent™ 1 m2 bioreactors in which satisfactory hiPSC expansion under serum-free conditions was achieved. These operating ranges were shown not only to effectively limit cell exposure to wall shear stress but also facilitated sufficient oxygen transfer and mixing. Through their application, almost 5 × 109 viable cells could be produced within 5 days, achieving expansion factors of up to 35 without discernable impact on cell viability, identity, or differentiation potential. Key Points •Bioengineering characterizations allowed the identification of operating ranges that supported satisfactory hiPSC expansion •Both the Xpansion® 10 multiplate and Ascent™ 1 m2 fixed-bed reactor accommodated the production of almost 5 × 109 viable cells within 5 days •Exposing the hiPSCs to a median wall shear stress of up to 8.2 × 10-5 N cm-2 did not impair quality.
Collapse
Affiliation(s)
- Misha Alexander Teale
- Centre for Cell Cultivation Techniques, Tissue Engineering, and Medical Biology, Institute of Chemistry and Biotechnology, ZHAW Zurich University of Applied Sciences, Grüentalstrasse 14, 8820, Wädenswil, Switzerland
| | - Samuel Lukas Schneider
- Centre for Cell Cultivation Techniques, Tissue Engineering, and Medical Biology, Institute of Chemistry and Biotechnology, ZHAW Zurich University of Applied Sciences, Grüentalstrasse 14, 8820, Wädenswil, Switzerland
| | - Stefan Seidel
- Centre for Cell Cultivation Techniques, Tissue Engineering, and Medical Biology, Institute of Chemistry and Biotechnology, ZHAW Zurich University of Applied Sciences, Grüentalstrasse 14, 8820, Wädenswil, Switzerland
| | - Jürgen Krasenbrink
- Advanced Manufacturing-Platform Engineering and Support, Bayer AG, Kaiser-Wilhelm-Allee 1, 51373, Leverkusen, Germany
| | - Martin Poggel
- Advanced Manufacturing-Platform Engineering and Support, Bayer AG, Kaiser-Wilhelm-Allee 1, 51373, Leverkusen, Germany
| | - Dieter Eibl
- Centre for Cell Cultivation Techniques, Tissue Engineering, and Medical Biology, Institute of Chemistry and Biotechnology, ZHAW Zurich University of Applied Sciences, Grüentalstrasse 14, 8820, Wädenswil, Switzerland
| | - Marcos F Q Sousa
- Advanced Manufacturing-Platform Engineering and Support, Bayer AG, Kaiser-Wilhelm-Allee 1, 51373, Leverkusen, Germany.
| | - Regine Eibl
- Centre for Cell Cultivation Techniques, Tissue Engineering, and Medical Biology, Institute of Chemistry and Biotechnology, ZHAW Zurich University of Applied Sciences, Grüentalstrasse 14, 8820, Wädenswil, Switzerland
| |
Collapse
|
4
|
Hashizume R, Wakita S, Sawada H, Takebayashi SI, Kitabatake Y, Miyagawa Y, Hirokawa YS, Imai H, Kurahashi H. Trisomic rescue via allele-specific multiple chromosome cleavage using CRISPR-Cas9 in trisomy 21 cells. PNAS NEXUS 2025; 4:pgaf022. [PMID: 39967679 PMCID: PMC11832276 DOI: 10.1093/pnasnexus/pgaf022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/07/2025] [Indexed: 02/20/2025]
Abstract
Human trisomy 21, responsible for Down syndrome, is the most prevalent genetic cause of cognitive impairment and remains a key focus for prenatal and preimplantation diagnosis. However, research directed toward eliminating supernumerary chromosomes from trisomic cells is limited. The present study demonstrates that allele-specific multiple chromosome cleavage by clustered regularly interspaced palindromic repeats Cas9 can achieve trisomy rescue by eliminating the target chromosome from human trisomy 21 induced pluripotent stem cells and fibroblasts. Unlike previously reported allele-nonspecific strategies, we have developed a comprehensive allele-specific (AS) Cas9 target sequence extraction method that efficiently removes the target chromosome. The temporary knockdown of DNA damage response genes increases the chromosome loss rate, while chromosomal rescue reversibly restores gene signatures and ameliorates cellular phenotypes. Additionally, this strategy proves effective in differentiated, nondividing cells. We anticipate that an AS approach will lay the groundwork for more sophisticated medical interventions targeting trisomy 21.
Collapse
Affiliation(s)
- Ryotaro Hashizume
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
- Department of Genomic Medicine, Mie University Hospital, Tsu, Mie 514-8507, Japan
| | - Sachiko Wakita
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Hirofumi Sawada
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Shin-ichiro Takebayashi
- Laboratory of Molecular and Cellular Biology, Graduate School of Bioresources, Mie University, Tsu, Mie 514-8507, Japan
| | - Yasuji Kitabatake
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yoshitaka Miyagawa
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Yoshifumi S Hirokawa
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Hiroshi Imai
- Department of Genomic Medicine, Mie University Hospital, Tsu, Mie 514-8507, Japan
- Pathology Division, Mie University Hospital, Tsu, Mie 514-8507, Japan
| | - Hiroki Kurahashi
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake 470-1192, Japan
| |
Collapse
|
5
|
Tang J, Huang X. Transcriptome analysis of human dental pulp cells cultured on a novel cell-adhesive fragment by RNA sequencing. Gene 2024; 927:148709. [PMID: 38901533 DOI: 10.1016/j.gene.2024.148709] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
AIM The aim of the present work was to find an efficient method for safe and reliable expansion of human dental pulp cells (hDPCs) in vitro. Here, we examined the effect of a novel recombinant E8 fragment of Laminin-511 (iMatrix-511) in hDPCs regarding viability and cell spreading. Further, we investigated the underlying mechanisms governing its effects in hDPCs using RNA sequencing (RNA-seq). METHODOLOGY hDPCs were obtained from caries-free maxilla third molars (n = 3). CCK-8 assay was conducted to measure the viability of cells cultured on iMatrix-511 and two other ECM proteins. Cell morphology was observed by phase contrast microscope. RNA-seq of hDPCs cultured on iMatrix-511 or noncoated control was performed on Illumina NovaseqTM 6000 platform. RESULTS iMatrix-511 (0.5 μg/cm2) enhanced the viability of hDPCs to an extent better than COL-1 and gelatin. Short term culture of hDPCs on iMatrix-511 resulted in 233 differentially expressed genes (DEGs). The top 12 most upregulated genes were XIAP, AL354740, MRFAP1, AC012321, KCND3, TMEM120B, AC009812, GET1-SH3BGR, CNTN3, AC090409, GEN1 and PIK3IP1, whereas the top 12 most downregulated genes were SFN, KRT17, RAB4B-EGLN2, CSTA, KCTD11, ATP6V1G2-DDX39B, AC010323, SBSN, LYPD3, FOSB, AC022400 and CHI3L1. qPCR validation confirmed the significant upregulation of GEN1, KCND3, PIK3IP1 and MRFAP1. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed, with genes enriched in various extracellular matrix interaction, estrogen and fat metabolism-related functions and pathways. CONCLUSIONS iMatrix-511 facilitated spreading and proliferation of hDPCs. It enhances expression of anti-apoptotic genes, while inhibits expression of epidermis development-related genes.
Collapse
Affiliation(s)
- Jia Tang
- School and Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, Shanghai 200072, PR China
| | - Xiaofeng Huang
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China.
| |
Collapse
|
6
|
Maihemuti W, Murata K, Abulaiti M, Minatoya K, Masumoto H. Simultaneous electro-dynamic stimulation accelerates maturation of engineered cardiac tissues generated by human iPS cells. Biochem Biophys Res Commun 2024; 733:150605. [PMID: 39197194 DOI: 10.1016/j.bbrc.2024.150605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/09/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
Electrical and dynamic stimulation are commonly employed to enhance the maturation of engineered cardiac tissue (ECT) derived from human induced pluripotent stem cells (iPSCs), reflecting the physiological environment of the heart. While electrical stimulation mimics natural bioelectrical signals and dynamic stimulation replicates mechanical forces, the combined effects of these stimuli on ECT maturation have not been thoroughly explored. We hypothesized that simultaneous electro-dynamic stimulation would enhance ECT maturation and function more effectively than either stimulus alone. Human iPSC-derived cardiovascular cells were co-cultured with Collagen I and Matrigel for 2 weeks, followed by a comparative analysis of four groups: no stimulation, dynamic stimulation, electrical stimulation, and simultaneous electro-dynamic stimulation. The functionality of ECTs was assessed by measuring contractile capacity and calcium indicators, and histological assessments examined structural maturation. Our results demonstrated that simultaneous electro-dynamic stimulation significantly increased the CM component, elevated TNNT2 mRNA expression levels, and enhanced calcium transient capacity. Additionally, ECTs subjected to simultaneous stimulation exhibited a positive force-frequency relationship in contractility and an elevation in peak calcium flux, indicative of advanced tissue maturation. Moreover, simultaneous stimulation promoted vascular network formation within the ECTs, suggesting improved structural organization. These findings underscore the importance of simultaneous stimulation for developing effective cardiac tissue engineering strategies.
Collapse
Affiliation(s)
- Wusiman Maihemuti
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan; Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kozue Murata
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan; Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mosha Abulaiti
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidetoshi Masumoto
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan; Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
7
|
Song HW, Solomon JN, Masri F, Mack A, Durand N, Cameau E, Dianat N, Hunter A, Oh S, Schoen B, Marsh M, Bravery C, Sumen C, Clarke D, Bharti K, Allickson JG, Lakshmipathy U. Bioprocessing considerations for generation of iPSCs intended for clinical application: perspectives from the ISCT Emerging Regenerative Medicine Technology working group. Cytotherapy 2024; 26:1275-1284. [PMID: 38970614 DOI: 10.1016/j.jcyt.2024.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 07/08/2024]
Abstract
Approval of induced pluripotent stem cells (iPSCs) for the manufacture of cell therapies to support clinical trials is now becoming realized after 20 years of research and development. In 2022 the International Society for Cell and Gene Therapy (ISCT) established a Working Group on Emerging Regenerative Medicine Technologies, an area in which iPSCs-derived technologies are expected to play a key role. In this article, the Working Group surveys the steps that an end user should consider when generating iPSCs that are stable, well-characterised, pluripotent, and suitable for making differentiated cell types for allogeneic or autologous cell therapies. The objective is to provide the reader with a holistic view of how to achieve high-quality iPSCs from selection of the starting material through to cell banking. Key considerations include: (i) intellectual property licenses; (ii) selection of the raw materials and cell sources for creating iPSC intermediates and master cell banks; (iii) regulatory considerations for reprogramming methods; (iv) options for expansion in 2D vs. 3D cultures; and (v) available technologies and equipment for harvesting, washing, concentration, filling, cryopreservation, and storage. Some key process limitations are highlighted to help drive further improvement and innovation, and includes recommendations to close and automate current open and manual processes.
Collapse
Affiliation(s)
- Hannah W Song
- Center for Cellular Engineering, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | - Emmanuelle Cameau
- Cytiva, Pall Life Sciences 24-26 avenue de Winchester, CS5005, 78100 St. Germain-en-Laye, France
| | | | | | - Steve Oh
- Cellvec Pte. Ltd. 100 Pasir Panjang, #04-01/02, Singapore 118518 Singapore
| | - Brianna Schoen
- Charles River Laboratories Cell Solutions, Inc. 8500 Balboa Blvd. Suite 230 Northridge, CA 91320, USA
| | | | | | | | | | - Kapil Bharti
- National Eye Institute, National Institutes of Health, Bethsda, MD, USA
| | - Julie G Allickson
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
8
|
Köhnke T, Karigane D, Hilgart E, Fan AC, Kayamori K, Miyauchi M, Collins CT, Suchy FP, Rangavajhula A, Feng Y, Nakauchi Y, Martinez-Montes E, Fowler JL, Loh KM, Nakauchi H, Koldobskiy MA, Feinberg AP, Majeti R. DNMT3A R882H Is Not Required for Disease Maintenance in Primary Human AML, but Is Associated With Increased Leukemia Stem Cell Frequency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.26.620318. [PMID: 39553934 PMCID: PMC11565803 DOI: 10.1101/2024.10.26.620318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Genetic mutations are being thoroughly mapped in human cancers, yet a fundamental question in cancer biology is whether such mutations are functionally required for cancer initiation, maintenance of established cancer, or both. Here, we study this question in the context of human acute myeloid leukemia (AML), where DNMT3A R882 missense mutations often arise early, in pre-leukemic clonal hematopoiesis, and corrupt the DNA methylation landscape to initiate leukemia. We developed CRISPR-based methods to directly correct DNMT3A R882 mutations in leukemic cells obtained from patients. Surprisingly, DNMT3A R882 mutations were largely dispensable for disease maintenance. Replacing DNMT3A R882 mutants with wild-type DNMT3A did not impair the ability of AML cells to engraft in vivo, and minimally altered DNA methylation. Taken together, DNMT3A R882 mutations are initially necessary for AML initiation, but are largely dispensable for disease maintenance. The notion that initiating oncogenes differ from those that maintain cancer has important implications for cancer evolution and therapy.
Collapse
Affiliation(s)
- Thomas Köhnke
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University; Stanford, CA, 94305, USA
| | - Daiki Karigane
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University; Stanford, CA, 94305, USA
| | - Eleanor Hilgart
- Center for Epigenetics and Department of Medicine, Johns Hopkins University School of Medicine; Baltimore, MD, 21205, USA
| | - Amy C. Fan
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University; Stanford, CA, 94305, USA
| | - Kensuke Kayamori
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University; Stanford, CA, 94305, USA
| | - Masashi Miyauchi
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University; Stanford, CA, 94305, USA
| | - Cailin T. Collins
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University; Stanford, CA, 94305, USA
| | - Fabian P. Suchy
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University; Stanford, CA, 94305, USA
| | - Athreya Rangavajhula
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University; Stanford, CA, 94305, USA
| | - Yang Feng
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University; Stanford, CA, 94305, USA
| | - Yusuke Nakauchi
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University; Stanford, CA, 94305, USA
| | - Eduardo Martinez-Montes
- Center for Epigenetics and Department of Medicine, Johns Hopkins University School of Medicine; Baltimore, MD, 21205, USA
| | - Jonas L. Fowler
- Department of Developmental Biology and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University; Stanford, CA, 94305, USA
| | - Kyle M. Loh
- Department of Developmental Biology and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University; Stanford, CA, 94305, USA
| | - Hiromitsu Nakauchi
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University; Stanford, CA, 94305, USA
| | - Michael A. Koldobskiy
- Center for Epigenetics and Department of Medicine, Johns Hopkins University School of Medicine; Baltimore, MD, 21205, USA
| | - Andrew P. Feinberg
- Center for Epigenetics and Department of Medicine, Johns Hopkins University School of Medicine; Baltimore, MD, 21205, USA
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University; Stanford, CA, 94305, USA
| |
Collapse
|
9
|
Lu X, Perr E, Naqvi T, Galitz D, Andersen M, Grabowski D, Person A, Kalyuzhny A, Flynn KC. A Novel Recombinant Vitronectin Variant Supports the Expansion and Differentiation of Pluripotent Stem Cells in Defined Animal-Free Workflows. Cells 2024; 13:1566. [PMID: 39329750 PMCID: PMC11429963 DOI: 10.3390/cells13181566] [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: 06/12/2024] [Revised: 09/04/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
An essential aspect of harnessing the potential of pluripotent stem cells (PSCs) and their derivatives for regenerative medicine is the development of animal-free and chemically defined conditions for ex vivo cultivation. PSCs, including embryonic and induced PSCs (iPSCs), are in the early stages of clinical trials for various indications, including degenerative diseases and traumatic injury. A key step in the workflows generating these cells for more widespread clinical use is their safe and robust ex vivo cultivation. This entails optimization of cell culture media and substrates that are safe and consistent while maintaining robust functionality. Here, we describe the design of a human vitronectin (hVTN) variant with improved manufacturability in a bacterial expression system along with improved function in comparison to wild-type VTN and other previously characterized polypeptide fragments. In conjunction with an animal component-free media formulation, our hVTN fragment provides animal-free conditions for the enhanced expansion of iPSCs. This hVTN variant also supports the reprogramming of PBMCs into iPSCs. Furthermore, we show that these iPSCs can be efficiently differentiated into the three major germ layers and cortical neurons, thereby closing the loop on a completely defined animal-free workflow for cell types relevant for regenerative medicine.
Collapse
Affiliation(s)
- Xi Lu
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA; (X.L.); (E.P.); (T.N.); (D.G.); (M.A.)
| | - Eli Perr
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA; (X.L.); (E.P.); (T.N.); (D.G.); (M.A.)
| | - Tahmina Naqvi
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA; (X.L.); (E.P.); (T.N.); (D.G.); (M.A.)
| | - David Galitz
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA; (X.L.); (E.P.); (T.N.); (D.G.); (M.A.)
| | - Marnelle Andersen
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA; (X.L.); (E.P.); (T.N.); (D.G.); (M.A.)
| | - David Grabowski
- Protein Development, Bio-Techne, Minneapolis, MN 55413, USA; (D.G.); (A.P.)
| | - Anthony Person
- Protein Development, Bio-Techne, Minneapolis, MN 55413, USA; (D.G.); (A.P.)
| | - Alex Kalyuzhny
- Antibody Applications, Bio-Techne, Minneapolis, MN 55413, USA;
| | - Kevin C. Flynn
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA; (X.L.); (E.P.); (T.N.); (D.G.); (M.A.)
- Department of Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
10
|
Takahashi K, Aritomi S, Honkawa F, Asari S, Hirose K, Konishi A. Efficient and cost-effective differentiation of induced neural crest cells from induced pluripotent stem cells using laminin 211. Regen Ther 2024; 26:749-759. [PMID: 39290629 PMCID: PMC11406167 DOI: 10.1016/j.reth.2024.08.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction Neural crest cells (NCCs) are cell populations that originate during the formation of neural crest in developmental stages. They are characterized by their multipotency, self-renewal and migration potential. Given their ability to differentiate into various types of cells such as neurons and Schwann cells, NCCs hold promise for cell therapy applications. The conventional method for obtaining NCCs involves inducing them from stem cells like induced pluripotent stem cells (iPSCs), followed by a long-term passage or purification using fluorescence-activated cell sorting (FACS). Although FACS allows high purity induced neural crest cells (iNCCs) to be obtained quickly, it is complex and costly. Therefore, there is a need for a simpler, cost-effective and less time-consuming method for cell therapy application. Methods To select differentiated iNCCs from heterogeneous cell populations quickly without using FACS, we adopted the use of scaffold material full-length laminin 211 (LN211), a recombinant, xeno-free protein suitable for cell therapy. After fist passage on LN211, iNCCs characterization was performed using polymerase chain reaction and flow cytometry. Additionally, proliferation and multipotency to various cells were evaluated. Result The iNCCs obtained using our new method expressed cranial NCC- related genes and exhibited stable proliferation ability for at least 57 days, while maintaining high expression level of the NCCs marker CD271. They demonstrated differentiation ability into several cell types: neurons, astrocytes, melanocytes, smooth muscle cells, osteoblasts, adipocytes and chondrocytes. Furthermore, they could be induced to differentiate into induced mesenchymal stem cells (iMSCs) which retain the essential functions of somatic MSCs. Conclusion In this study, we have developed novel method for obtaining high purity iNCCs differentiated from iPSCs in a short time using LN211 under xeno-free condition. Compared with traditional methods, like FACS or long-term passage, this approach enables the acquisition of a large amount of cells at a lower cost and labor, and it is expected to contribute to stable supply of large scale iNCCs for future cell therapy applications.
Collapse
Affiliation(s)
- Kazuma Takahashi
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kanagawa, Kawasaki, 210-8681, Japan
| | - Shizuka Aritomi
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kanagawa, Kawasaki, 210-8681, Japan
| | - Fumie Honkawa
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kanagawa, Kawasaki, 210-8681, Japan
| | - Sayaka Asari
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kanagawa, Kawasaki, 210-8681, Japan
| | - Ken Hirose
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kanagawa, Kawasaki, 210-8681, Japan
| | - Atsushi Konishi
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kanagawa, Kawasaki, 210-8681, Japan
| |
Collapse
|
11
|
Watanabe T, Yasuda S, Kusakawa S, Kuroda T, Furukawa H, Futamura M, Shimizu S, Morishita A, Hata S, Koeda A, Komatsu K, Sato Y. Multisite studies for optimization of a highly efficient culture assay used for in vitro detection of residual undifferentiated human pluripotent stem cells intermingled in cell therapy products. Regen Ther 2024; 26:315-323. [PMID: 38983832 PMCID: PMC11231703 DOI: 10.1016/j.reth.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/21/2024] [Accepted: 06/09/2024] [Indexed: 07/11/2024] Open
Abstract
Introduction MEASURE2 (Multisite Evaluation Study on Analytical Methods for Non-clinical Safety Assessment of HUman-derived REgenerative Medical Products 2) is a Japanese experimental public-private partnership initiative that aims to standardize testing methods for tumorigenicity evaluation of human pluripotent stem cell (hPSC)-derived cell therapy products (CTPs). MEASURE2 organized multisite studies to optimize the methodology of the highly efficient culture (HEC) assay, a sensitive culture-based in vitro assay for detecting residual undifferentiated hPSCs in CTPs. Methods In these multisite studies, 1) the efficiency of colony formation by human induced pluripotent stem cells (hiPSCs) under two different culture conditions and 2) the sorting efficiency of microbeads conjugated to various anti-hPSC markers during hiPSC enrichment were evaluated using samples in which hiPSCs were spiked into hiPSC-derived mesenchymal stem cells. Results The efficiency of colony formation was significantly higher under culture conditions with the combination of Chroman 1, Emricasan, Polyamines, and Trans-ISRIB (CEPT) than with Y-27632, which is widely used for the survival of hPSCs. Between-laboratory variance was also smaller under the condition with CEPT than with Y-27632. The sorting efficiency of microbeads conjugated with the anti-Tra-1-60 antibody was sufficiently higher (>80%) than those of the other various microbeads investigated. Conclusions Results of these multisite studies are expected to contribute to improvements in the sensitivity and robustness of the HEC assay, as well as to the future standardization of the tumorigenicity risk assessment of hPSC-derived CTPs.
Collapse
Affiliation(s)
- Takeshi Watanabe
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
| | - Shinji Kusakawa
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
| | - Takuya Kuroda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
| | - Hatsue Furukawa
- Safety Business Unit, Axcelead Drug Discovery Partners, Inc., Kanagawa, Japan
| | - Mayumi Futamura
- Drug Discovery Support Division, Tsukuba Research Institute, BoZo Research Center Inc., Ibaraki, Japan
| | - Shigekazu Shimizu
- CMIC Bioresearch Center, CMIC Pharma Science Co., Ltd., Yamanashi, Japan
| | | | | | - Akiko Koeda
- Research Administration Department, Ina Research Inc., Nagano, Japan
| | - Kana Komatsu
- Clinical Laboratory Department, Ina Research Inc., Nagano, Japan
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kanagawa, Japan
- Division of Drugs, National Institute of Health Sciences, Kanagawa, Japan
| |
Collapse
|
12
|
Fujisaki H, Watanabe T, Yoshihara S, Fukuda H, Tomono Y, Tometsuka C, Mizuno K, Nishiyama T, Hattori S. Laminin 511 E8 fragment promotes to form basement membrane-like structure in human skin equivalents. Regen Ther 2024; 26:717-728. [PMID: 39286641 PMCID: PMC11403260 DOI: 10.1016/j.reth.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/22/2024] [Accepted: 08/18/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction Laminin 511 (LM511), a component of the skin basement membrane (BM), is known to enhance the adhesion of some cell types and it has been reported to affect cell behavior. A recombinant fragment consisting of the integrin recognition site; E8 region of LM511 (511E8) has also been studied. 511E8 has been reported by many as a superior culture substrate. However, the effects of 511E8 on human skin cells remain unclear. In this study, we added 511E8 during the culture period of a reconstituted skin equivalent (SE) and investigated its effect on the formation of BM-like structures. Methods SEs were formed by air-liquid culture of human foreskin keratinocytes (HFKs) on contracted type I collagen (Col-I) gels containing human fibroblasts. We compared the BM-like structures formed with and without 511E8 during HFKs culture periods. Morphological analysis, gene expression analysis of extracellular matrix components, and localization analysis of 511E8 in order to identify where 511E8 works were performed. Results Immunohistochemical observation by light microscopy showed an accumulation of BM components between the gels and cell layers regardless of the addition of 511E8. There was a stronger and more continuous positive staining for LM α3, type IV collagen, and type VII collagen in the 511E8-added group compared to the no-added group. Transmission electron microscopic observation showed that the continuity of BM-like structures was increased with the addition of 511E8. Furthermore, gene expression analysis showed that the 511E8 addition increased some BM component genes expression, with collagen type IV and type VII α1 chains showing significant increases. His-tagged 511E8 was stained around the basal cells of HFK layers, not in basal regions. Co-staining with anti-His-tag and anti-integrin β1 antibodies revealed the co-localization of theses in some intercellular regions among basal cells. Conclusion These results suggest that 511E8 effected on HFKs, enhancing the production of BM components and strengthening the anchoring between the Col-I gels and the HFK layers.
Collapse
Affiliation(s)
- Hitomi Fujisaki
- Nippi Research Institute of Biomatrix, 520-11 Kuwabara, Toride, Ibaraki 302-0017, Japan
| | - Takafumi Watanabe
- Laboratory of Veterinary Anatomy, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - Shusuke Yoshihara
- Laboratory of Veterinary Anatomy, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - Hideki Fukuda
- Laboratory of Veterinary Anatomy, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | | | - Chisa Tometsuka
- Nippi Research Institute of Biomatrix, 520-11 Kuwabara, Toride, Ibaraki 302-0017, Japan
| | - Kazunori Mizuno
- Nippi Research Institute of Biomatrix, 520-11 Kuwabara, Toride, Ibaraki 302-0017, Japan
| | - Toshio Nishiyama
- Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| | - Shunji Hattori
- Nippi Research Institute of Biomatrix, 520-11 Kuwabara, Toride, Ibaraki 302-0017, Japan
| |
Collapse
|
13
|
Shoji M, Ohashi T, Nagase S, Yuri H, Ichihashi K, Takagishi T, Nagata Y, Nomura Y, Fukunaka A, Kenjou S, Miyake H, Hara T, Yoshigai E, Fujitani Y, Sakurai H, Dos Santos HG, Fukada T, Kuzuhara T. Possible involvement of zinc transporter ZIP13 in myogenic differentiation. Sci Rep 2024; 14:8052. [PMID: 38609428 PMCID: PMC11014994 DOI: 10.1038/s41598-024-56912-7] [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: 11/09/2022] [Accepted: 03/12/2024] [Indexed: 04/14/2024] Open
Abstract
Ehlers-Danlos syndrome spondylodysplastic type 3 (EDSSPD3, OMIM 612350) is an inherited recessive connective tissue disorder that is caused by loss of function of SLC39A13/ZIP13, a zinc transporter belonging to the Slc39a/ZIP family. We previously reported that patients with EDSSPD3 harboring a homozygous loss of function mutation (c.221G > A, p.G64D) in ZIP13 exon 2 (ZIP13G64D) suffer from impaired development of bone and connective tissues, and muscular hypotonia. However, whether ZIP13 participates in the early differentiation of these cell types remains unclear. In the present study, we investigated the role of ZIP13 in myogenic differentiation using a murine myoblast cell line (C2C12) as well as patient-derived induced pluripotent stem cells (iPSCs). We found that ZIP13 gene expression was upregulated by myogenic stimulation in C2C12 cells, and its knockdown disrupted myotubular differentiation. Myocytes differentiated from iPSCs derived from patients with EDSSPD3 (EDSSPD3-iPSCs) also exhibited incomplete myogenic differentiation. Such phenotypic abnormalities of EDSSPD3-iPSC-derived myocytes were corrected by genomic editing of the pathogenic ZIP13G64D mutation. Collectively, our findings suggest the possible involvement of ZIP13 in myogenic differentiation, and that EDSSPD3-iPSCs established herein may be a promising tool to study the molecular basis underlying the clinical features caused by loss of ZIP13 function.
Collapse
Affiliation(s)
- Masaki Shoji
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan.
| | - Takuto Ohashi
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Saki Nagase
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Haato Yuri
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Kenta Ichihashi
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Teruhisa Takagishi
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Yuji Nagata
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Yuki Nomura
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Ayako Fukunaka
- Laboratory of Developmental Biology and Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi-City, Gunma, Japan
| | - Sae Kenjou
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Hatsuna Miyake
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Takafumi Hara
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Emi Yoshigai
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan
| | - Yoshio Fujitani
- Laboratory of Developmental Biology and Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi-City, Gunma, Japan
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto-City, Kyoto, Japan
| | | | - Toshiyuki Fukada
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan.
| | - Takashi Kuzuhara
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima, 770-8514, Japan.
| |
Collapse
|
14
|
Tsujimoto H, Hoshina A, Mae SI, Araoka T, Changting W, Ijiri Y, Nakajima-Koyama M, Sakurai S, Okita K, Mizuta K, Niwa A, Saito MK, Saitou M, Yamamoto T, Graneli C, Woollard KJ, Osafune K. Selective induction of human renal interstitial progenitor-like cell lineages from iPSCs reveals development of mesangial and EPO-producing cells. Cell Rep 2024; 43:113602. [PMID: 38237600 DOI: 10.1016/j.celrep.2023.113602] [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: 05/11/2022] [Revised: 06/13/2023] [Accepted: 12/05/2023] [Indexed: 03/02/2024] Open
Abstract
Recent regenerative studies using human pluripotent stem cells (hPSCs) have developed multiple kidney-lineage cells and organoids. However, to further form functional segments of the kidney, interactions of epithelial and interstitial cells are required. Here we describe a selective differentiation of renal interstitial progenitor-like cells (IPLCs) from human induced pluripotent stem cells (hiPSCs) by modifying our previous induction method for nephron progenitor cells (NPCs) and analyzing mouse embryonic interstitial progenitor cell (IPC) development. Our IPLCs combined with hiPSC-derived NPCs and nephric duct cells form nephrogenic niche- and mesangium-like structures in vitro. Furthermore, we successfully induce hiPSC-derived IPLCs to differentiate into mesangial and erythropoietin-producing cell lineages in vitro by screening differentiation-inducing factors and confirm that p38 MAPK, hypoxia, and VEGF signaling pathways are involved in the differentiation of mesangial-lineage cells. These findings indicate that our IPC-lineage induction method contributes to kidney regeneration and developmental research.
Collapse
Affiliation(s)
- Hiraku Tsujimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Azusa Hoshina
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shin-Ichi Mae
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Toshikazu Araoka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Wang Changting
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yoshihiro Ijiri
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - May Nakajima-Koyama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Satoko Sakurai
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kazusa Okita
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Ken Mizuta
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Akira Niwa
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Megumu K Saito
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Mitinori Saitou
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto 606-8501, Japan
| | - Takuya Yamamoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto 606-8501, Japan; Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto 606-8507, Japan
| | - Cecilia Graneli
- BioPharmaceuticals R&D Cell Therapy, Research and Early Development, Cardiovascular, Renal and Metabolic (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Kevin J Woollard
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
15
|
Murata K, Makino A, Tomonaga K, Masumoto H. Predicted risk of heart failure pandemic due to persistent SARS-CoV-2 infection using a three-dimensional cardiac model. iScience 2024; 27:108641. [PMID: 38299028 PMCID: PMC10829886 DOI: 10.1016/j.isci.2023.108641] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 11/13/2023] [Accepted: 12/01/2023] [Indexed: 02/02/2024] Open
Abstract
Patients with chronic cardiomyopathy may have persistent viral infections in their hearts, particularly with SARS-CoV-2, which targets the ACE2 receptor highly expressed in human hearts. This raises concerns about a potential global heart failure pandemic stemming from COVID-19, an SARS-CoV-2 pandemic in near future. Although faced with this healthcare caveat, there is limited research on persistent viral heart infections, and no models have been established. In this study, we created an SARS-CoV-2 persistent infection model using human iPS cell-derived cardiac microtissues (CMTs). Mild infections sustained viral presence without significant dysfunction for a month, indicating persistent infection. However, when exposed to hypoxic conditions mimicking ischemic heart diseases, cardiac function deteriorated alongside intracellular SARS-CoV-2 reactivation in cardiomyocytes and disrupted vascular network formation. This study demonstrates that SARS-CoV-2 persistently infects the heart opportunistically causing cardiac dysfunction triggered by detrimental stimuli such as ischemia, potentially predicting a post COVID-19 era heart failure pandemic.
Collapse
Affiliation(s)
- Kozue Murata
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akiko Makino
- Laboratory of RNA Viruses, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Keizo Tomonaga
- Laboratory of RNA Viruses, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hidetoshi Masumoto
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
16
|
Netsrithong R, Garcia-Perez L, Themeli M. Engineered T cells from induced pluripotent stem cells: from research towards clinical implementation. Front Immunol 2024; 14:1325209. [PMID: 38283344 PMCID: PMC10811463 DOI: 10.3389/fimmu.2023.1325209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/15/2023] [Indexed: 01/30/2024] Open
Abstract
Induced pluripotent stem cell (iPSC)-derived T (iT) cells represent a groundbreaking frontier in adoptive cell therapies with engineered T cells, poised to overcome pivotal limitations associated with conventional manufacturing methods. iPSCs offer an off-the-shelf source of therapeutic T cells with the potential for infinite expansion and straightforward genetic manipulation to ensure hypo-immunogenicity and introduce specific therapeutic functions, such as antigen specificity through a chimeric antigen receptor (CAR). Importantly, genetic engineering of iPSC offers the benefit of generating fully modified clonal lines that are amenable to rigorous safety assessments. Critical to harnessing the potential of iT cells is the development of a robust and clinically compatible production process. Current protocols for genetic engineering as well as differentiation protocols designed to mirror human hematopoiesis and T cell development, vary in efficiency and often contain non-compliant components, thereby rendering them unsuitable for clinical implementation. This comprehensive review centers on the remarkable progress made over the last decade in generating functional engineered T cells from iPSCs. Emphasis is placed on alignment with good manufacturing practice (GMP) standards, scalability, safety measures and quality controls, which constitute the fundamental prerequisites for clinical application. In conclusion, the focus on iPSC as a source promises standardized, scalable, clinically relevant, and potentially safer production of engineered T cells. This groundbreaking approach holds the potential to extend hope to a broader spectrum of patients and diseases, leading in a new era in adoptive T cell therapy.
Collapse
Affiliation(s)
- Ratchapong Netsrithong
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Laura Garcia-Perez
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Maria Themeli
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
17
|
Fiacco E, Landi S, Zasso J, Ambrosini C, Faga G. Optimized and Scalable Precoating-Free Reprogramming of Human Peripheral Blood Mononuclear Cells into iPSCs. Curr Protoc 2024; 4:e979. [PMID: 38265186 DOI: 10.1002/cpz1.979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Human disease modeling has been profoundly transformed by the introduction of human induced pluripotent stem cells (iPSCs), marking the onset of a new era. This ground-breaking development offers a tailored framework for generating pluripotent cells from any individual, effectively enabling the development of cellular models for the study of human physiology and diseases on an unprecedented scale. Although technologies for iPSCs generation have advanced rapidly over the past two decades, protocols for reprogramming patient-derived somatic cells into stem cells still pose a major challenge for the development of automated pipelines capable of generating iPSCs at scales that are cost-effective, reproducible, and easy to implement. Most methods commonly rely on extracellular matrix protein mixtures or synthetic substrates to promote efficient proliferation of iPSCs. Nonetheless, employing these substances entails a laborious and time-consuming process, as the culture surface requires coating treatments before cell seeding. Here we describe a method for reprogramming blood-derived mononucleated cells that eliminates the need to precoat culture surfaces for the entire experimental flow. This procedure is suitable for fresh or frozen purified peripheral blood mononuclear cells (PBMCs) and allows seeding of reprogrammed cells in a culture medium containing a fragment of laminin-511, regardless of the method of reprogramming employed. Our protocol incorporates a streamlined workflow that optimizes key factors, including cell density, culture medium composition, and iPSC culture propagation techniques. Using a precoating-free approach, we eliminate the time-consuming steps, while our optimized subcloning method improves the scalability of the protocol, making it suitable for large-scale applications. Additionally, the automation-friendly nature of our protocol allows for high-throughput processing, reducing the labor and costs associated with manual handling. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Miniaturized and time efficient precoating-free reprogramming of fresh or frozen PBMCs Alternate Protocol: Erythroid progenitor cells (EPCs) enrichment and reprogramming into iPSCs using Sendai viral vectors Basic Protocol 2: Picking and precoating-free optimized expansion of iPSC clones.
Collapse
|
18
|
Lin CY, Ching YY, Wu SF, Lee YK, Fan HC, Su LY, Tsai SY, Chen YC, Shen CI, Su HL. Coating-Free Culture Medium for Establishing and Maintaining Human Induced Pluripotent Stem Cells. Cell Transplant 2023; 32:9636897231198172. [PMID: 37698258 PMCID: PMC10498698 DOI: 10.1177/09636897231198172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 09/13/2023] Open
Abstract
Cell expansion of human pluripotent stem cells (hPSCs) commonly depends on Matrigel as a coating matrix on two-dimensional (2D) culture plates and 3D microcarriers. However, the xenogenic Matrigel requires sophisticated quality-assurance processes to meet clinical requirements. In this study, we develop an innovative coating-free medium for expanding hPSCs. The xenofree medium supports the weekend-free culture and competitive growth of hPSCs on several cell culture plastics without an additional pre-coating process. The pluripotent stemness of the expanded cells is stably sustained for more than 10 passages, featured with high pluripotent marker expressions, normal karyotyping, and differentiating capacity for three germ layers. The expression levels of some integrins are reduced, compared with those of the hPSCs on Matrigel. This medium also successfully supports the clonal expansion and induced pluripotent stem cell establishment from mitochondrial-defective MELAS (mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes) patient's peripheral blood mononuclear cells. This innovative hPSC medium provides a straightforward scale-up process for producing clinical-orientated hPSCs by excluding the conventional coating procedure.
Collapse
Affiliation(s)
- Chih-Yao Lin
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yu-Yun Ching
- Duogenic StemCells Corporation, Taichung, Taiwan
| | - Shih-Fang Wu
- The Joint Program of Tissue Engineering and Regenerative Medicine, National Chung Hsing University and National Health Research Institutes, Taichung, Taiwan
| | - Yi-Ko Lee
- Duogenic StemCells Corporation, Taichung, Taiwan
| | - Hueng-Chuen Fan
- Department of Pediatrics, Tungs’ Taichung MetroHarbor Hospital, Wuchi, Taichung, Taiwan
- Department of Nursing, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| | - Liang-Yu Su
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Su-Yi Tsai
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yu-Ching Chen
- The Joint Program of Tissue Engineering and Regenerative Medicine, National Chung Hsing University and National Health Research Institutes, Taichung, Taiwan
- Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua City, Taiwan
| | - Ching-I Shen
- Duogenic StemCells Corporation, Taichung, Taiwan
| | - Hong-Lin Su
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
19
|
Kamishibahara Y, Okamoto S, Ohkuma T, Taniguchi H. Stabilized generation of human iPSC-derived liver organoids using a modified coating approach. Biol Methods Protoc 2022; 8:bpac034. [PMID: 36694573 PMCID: PMC9869720 DOI: 10.1093/biomethods/bpac034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/24/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
Human-induced pluripotent stem cell (hiPSC)-derived hepatic cells are useful tools for regenerative medicine, and various culture substrates are currently used for their differentiation. We differentiated hiPSC-derived hepatic endoderm (HE), endothelial cells (ECs), and mesenchymal cells (MCs) using Laminin-511 (LN) coating to generate liver organoids, hiPSC-liver buds (hiPSC-LBs), which exhibited therapeutic effects when transplanted into disease model animals. Stably producing significant amounts of hiPSC-LBs is necessary for sufficient therapeutic effects. However, general precoating (standard coating) requires quick manipulation, often causing failure for inexperienced cell cultures, we thus tested direct LN addition to the culture medium (Direct coating). Using quantitative gene expression, flow cytometry, albumin secretion, and ammonia metabolism, we demonstrated that Standard and Direct coating similarly induce hiPSC-derived hepatocyte, mesodermal cell, EC, and MC differentiation. Standard and Direct coating-differentiated cells generated iPSC-LBs with equivalent hepatic functions. Furthermore, Direct coating enabled stable induction of differentiation independent of individual culture skills and reduced total amount of LN use as the same differentiated cell quality can be obtained upon LN supplementation at lower concentrations. In summary, the results of this study suggest that Direct coating could enable stable hiPSC-LB production at a low cost, thereby yielding mass cell production using hiPSCs.
Collapse
Affiliation(s)
- Yu Kamishibahara
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Satoshi Okamoto
- Correspondence address. (S.O.) Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan. Tel/: +81 45 787 8963; E-mail: . (H.T.) Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, the University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan. Tel: +81 3 5449 5698; E-mail:
| | - Takuya Ohkuma
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Hideki Taniguchi
- Correspondence address. (S.O.) Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan. Tel/: +81 45 787 8963; E-mail: . (H.T.) Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, the University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan. Tel: +81 3 5449 5698; E-mail:
| |
Collapse
|
20
|
Tsujimoto H, Katagiri N, Ijiri Y, Sasaki B, Kobayashi Y, Mima A, Ryosaka M, Furuyama K, Kawaguchi Y, Osafune K. In vitro methods to ensure absence of residual undifferentiated human induced pluripotent stem cells intermingled in induced nephron progenitor cells. PLoS One 2022; 17:e0275600. [PMID: 36378656 PMCID: PMC9665373 DOI: 10.1371/journal.pone.0275600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Cell therapies using human induced pluripotent stem cell (hiPSC)-derived nephron progenitor cells (NPCs) are expected to ameliorate acute kidney injury (AKI). However, using hiPSC-derived NPCs clinically is a challenge because hiPSCs themselves are tumorigenic. LIN28A, ESRG, CNMD and SFRP2 transcripts have been used as a marker of residual hiPSCs for a variety of cell types undergoing clinical trials. In this study, by reanalyzing public databases, we found a baseline expression of LIN28A, ESRG, CNMD and SFRP2 in hiPSC-derived NPCs and several other cell types, suggesting LIN28A, ESRG, CNMD and SFRP2 are not always reliable markers for iPSC detection. As an alternative, we discovered a lncRNA marker gene, MIR302CHG, among many known and unknown iPSC markers, as highly differentially expressed between hiPSCs and NPCs, by RNA sequencing and quantitative RT-PCR (qRT-PCR) analyses. Using MIR302CHG as an hiPSC marker, we constructed two assay methods, a combination of magnetic bead-based enrichment and qRT-PCR and digital droplet PCR alone, to detect a small number of residual hiPSCs in NPC populations. The use of these in vitro assays could contribute to patient safety in treatments using hiPSC-derived cells.
Collapse
Affiliation(s)
- Hiraku Tsujimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, Kyoto, Japan
- * E-mail: (KO); (HT)
| | - Naoko Katagiri
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, Kyoto, Japan
| | - Yoshihiro Ijiri
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, Kyoto, Japan
| | - Ben Sasaki
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yoshifumi Kobayashi
- Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, Kyoto, Japan
| | - Akira Mima
- Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, Kyoto, Japan
| | - Makoto Ryosaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, Kyoto, Japan
| | - Kenichiro Furuyama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yoshiya Kawaguchi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- * E-mail: (KO); (HT)
| |
Collapse
|
21
|
Huang WC, Hung CH, Lin YW, Zheng YC, Lei WL, Lu HE. Electrically Copolymerized Polydopamine Melanin/Poly(3,4-ethylenedioxythiophene) Applied for Bioactive Multimodal Neural Interfaces with Induced Pluripotent Stem Cell-Derived Neurons. ACS Biomater Sci Eng 2022; 8:4807-4818. [PMID: 36222713 DOI: 10.1021/acsbiomaterials.2c00822] [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: 11/29/2022]
Abstract
Multimodal neural interfaces include combined functions of electrical neuromodulation and synchronic monitoring of neurochemical and physiological signals in one device. The remarkable biocompatibility and electrochemical performance of polystyrene sulfonate-doped poly(3,4-ethylenedioxythiophene) (PEDOT:PSS) have made it the most recommended conductive polymer neural electrode material. However, PEDOT:PSS formed by electrochemical deposition, called PEDOT/PSS, often need multiple doping to improve structural instability in moisture, resolve the difficulties of functionalization, and overcome the poor cellular affinity. In this work, inspired by the catechol-derived adhesion and semiconductive properties of polydopamine melanin (PDAM), we used electrochemical oxidation polymerization to develop PDAM-doped PEDOT (PEDOT/PDAM) as a bioactive multimodal neural interface that permits robust electrochemical performance, structural stability, analyte-trapping capacity, and neural stem cell affinity. The use of potentiodynamic scans resolved the problem of copolymerizing 3,4-ethylenedioxythiophene (EDOT) and dopamine (DA), enabling the formation of PEDOT/PDAM self-assembled nanodomains with an ideal doping state associated with remarkable current storage and charge transfer capacity. Owing to the richness of hydrogen bond donors/acceptors provided by the hydroxyl groups of PDAM, PEDOT/PDAM presented better electrochemical and mechanical stability than PEDOT/PSS. It has also enabled high sensitivity and selectivity in the electrochemical detection of DA. Different from PEDOT/PSS, which inhibited the survival of human induced pluripotent stem cell-derived neural progenitor cells, PEDOT/PDAM maintained cell proliferation and even promoted cell differentiation into neuronal networks. Finally, PEDOT/PDAM was modified on a commercialized microelectrode array system, which resulted in the reduction of impedance by more than one order of magnitude; this significantly improved the resolution and reduced the noise of neuronal signal recording. With these advantages, PEDOT/PDAM is anticipated to be an efficient bioactive multimodal neural electrode material with potential application to brain-machine interfaces.
Collapse
Affiliation(s)
- Wei-Chen Huang
- Department of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| | - Ching-Heng Hung
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Yueh-Wen Lin
- Department of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| | - Yu-Cheng Zheng
- Department of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| | - Wan-Lou Lei
- Department of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| | - Huai-En Lu
- Food Industry Research and Development Institute, Hsinchu 300, Taiwan.,Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| |
Collapse
|
22
|
Alarfaj AA, Hirad AH, Munusamy MA, Kumar SS, Higuchi A. Human embryonic stem cells cultured on hydrogels grafted with extracellular matrix protein-derived peptides with polyethylene glycol joint nanosegments. IET Nanobiotechnol 2022; 16:295-304. [PMID: 36200801 DOI: 10.1049/nbt2.12091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/01/2022] [Accepted: 06/24/2022] [Indexed: 11/19/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) can be proliferated on completely synthetic materials under xeno-free cultivation conditions using biomaterials grafted with extracellular matrix protein (ECM)-derived peptides. However, cell culture biomaterials grafted with ECM-derived peptides must be prepared using a high concentration of peptide reaction solution (e.g. 1000 μg/ml), whereas the ECM concentration of the ECM-coated surface for hPSC culture is typically 5 μg/ml. We designed a polyethylene glycol (PEG) joint nanosegment (linker) to be used between base cell culture biomaterials and bioactive ECM-derived peptides to enhance the probability of contact between ECM-derived peptides and cell binding receptors of hPSCs. Vitronectin-derived peptides with glycine joint nanosegments (GCGG) were conjugated onto poly (vinyl alcohol-co-itaconic acid) hydrogels via PEG joint nanosegments, and human embryonic stem cells (hESCs) were cultivated on these hydrogels. hESCs could successfully be cultivated on hydrogels while maintaining their pluripotency and differentiation potential to differentiate into cells that are induced from three germ layers in vitro and in vivo, where only a 50 μg/ml ECM-derived peptide concentration was used when the PEG joint nanosegments were introduced into peptides that were grafted onto hydrogel surfaces. The joint nanosegments between bioactive peptides and base cell culture biomaterials were found to contribute to efficient hESC attachment and proliferation.
Collapse
Affiliation(s)
- Abdullah A Alarfaj
- Department of Botany and Microbiology, King Saud University, Riyadh, Saudi Arabia
| | - Abdurahman H Hirad
- Department of Botany and Microbiology, King Saud University, Riyadh, Saudi Arabia
| | - Murugan A Munusamy
- Department of Botany and Microbiology, King Saud University, Riyadh, Saudi Arabia
| | - S Suresh Kumar
- Department of Biotechnology, Bharath Institute of Higher Education and Research, Chennai-73, India
| | - Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan.,Department of Reproduction, National Center for Child Health and Development, Tokyo, Japan.,School of Ophthalmology and Optometry, The Eye Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, China.,R&D Center for Membrane Technology, Chung Yuan Christian University, Taoyuan, Taiwan
| |
Collapse
|
23
|
Ryosaka M, Mae SI, Osafune K. Protocol for the generation and expansion of human iPS cell-derived ureteric bud organoids. STAR Protoc 2022; 3:101484. [PMID: 35769929 PMCID: PMC9234152 DOI: 10.1016/j.xpro.2022.101484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The ureteric bud (UB) is a kidney precursor tissue that repeats branching morphogenesis and gives rise to the collecting ducts (CDs) and lower urinary tract. Here, we describe protocols to generate iUB organoids from human iPSCs; iUB organoids repeat branching morphogenesis. We describe how to expand iUB-organoid-derived tip colonies and how to induce CD progenitors from iUB organoids. These organoids can be used to study CD development and potentially as a model of kidney and urinary tract diseases. For complete details on the use and execution of this protocol, please refer to Mae et al. (2020). Protocol for ureteric bud (UB) organoids with repeated branching potential An expansion culture approach for UB tip colonies using hydrogel Differentiation of UB organoids into 2D and 3D collecting duct progenitors
Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics.
Collapse
|
24
|
Kitajima K, Shingai M, Ando H, Hamasaki M, Hara T. An interferon-γ/FLT3 axis positively regulates hemopoietic progenitor cell expansion from human pluripotent stem cells. Stem Cells 2022; 40:906-918. [PMID: 35901509 DOI: 10.1093/stmcls/sxac052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/14/2022] [Indexed: 11/12/2022]
Abstract
Since it became possible to differentiate human pluripotent stem cells (hPSCs) into hematopoietic cells in vitro, great efforts have been made to obtain highly potent hematopoietic stem/progenitor cells (HSPCs) from hPSCs. Immunophenotypical HSPCs can be obtained from hPSCs, but their repopulating potential in vivo is low. Here, we developed a novel hematopoietic differentiation method for human induced pluripotent stem cells (hiPSCs) to determine why the existing hPSC differentiation systems are inadequate. hiPSC-derived CD45 +CD34 + cells in our system were mostly CD38 - immunophenotypical HSPCs. The vast majority of human CD45 +CD34 + cells in umbilical cord blood, fetal liver, and bone marrow are CD38 + hematopoietic progenitor cells (HPCs); therefore, the poor production of CD38 + HPCs was indicative of a systematic problem. hiPSC-derived CD45 +CD34 + cells did not express FLT3, a receptor tyrosine kinase. Exogenous FLT3 activity significantly enhanced production of CD38 + HPCs from hiPSCs. Thus, poor production of CD38 + HPCs was due to a lack of FLT3 expression. Interferon-γ upregulated expression of FLT3 and increased the number of CD38 + HPCs among hiPSC-derived CD45 +CD34 + cells. These results suggest that poor production of CD38 + HPCs with hPSC differentiation systems is due to a lack of FLT3 expression, and that addition of interferon-γ can solve this problem.
Collapse
Affiliation(s)
- Kenji Kitajima
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Minako Shingai
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hikaru Ando
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mako Hamasaki
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Graduate School of Science, Department of Biological Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Takahiko Hara
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Graduate School of Science, Department of Biological Science, Tokyo Metropolitan University, Tokyo, Japan
| |
Collapse
|
25
|
Takahashi K, Okubo C, Nakamura M, Iwasaki M, Kawahara Y, Tabata T, Miyamoto Y, Woltjen K, Yamanaka S. A stress-reduced passaging technique improves the viability of human pluripotent cells. CELL REPORTS METHODS 2022; 2:100155. [PMID: 35474962 PMCID: PMC9017214 DOI: 10.1016/j.crmeth.2021.100155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/13/2021] [Accepted: 12/23/2021] [Indexed: 06/14/2023]
Abstract
Xeno-free culture systems have expanded the clinical and industrial application of human pluripotent stem cells (PSCs). However, reproducibility issues, often arising from variability during passaging steps, remain. Here, we describe an improved method for the subculture of human PSCs. The revised method significantly enhances the viability of human PSCs by lowering DNA damage and apoptosis, resulting in more efficient and reproducible downstream applications such as gene editing and directed differentiation. Furthermore, the method does not alter PSC characteristics after long-term culture and attenuates the growth advantage of abnormal subpopulations. This robust passaging method minimizes experimental error and reduces the rate of PSCs failing quality control of human PSC research and application.
Collapse
Affiliation(s)
- Kazutoshi Takahashi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Chikako Okubo
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Michiko Nakamura
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Mio Iwasaki
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Yuka Kawahara
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Tsuyoshi Tabata
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Yousuke Miyamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Knut Woltjen
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Shinya Yamanaka
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
26
|
Nishimura T, Niizuma K, Nakauchi H. Bioluminescent Tracking of Human Induced Pluripotent Stem Cells In Vitro and In Vivo. Methods Mol Biol 2022; 2524:291-297. [PMID: 35821480 DOI: 10.1007/978-1-0716-2453-1_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The discovery and development of induced pluripotent stem cells (iPSCs) opened a novel venue for disease modeling, drug discovery, and personalized medicine. Additionally, iPSCs have been utilized for a wide variety of research and clinical applications without immunological and ethical concerns that arise from using embryonic stem cells. Understanding the in vivo behavior of iPSCs, as well as their derivatives, requires the monitoring of their localization, proliferation, and viability after transplantation. Bioluminescence imaging (BLI) gives investigators a non-invasive and sensitive means for spatio-temporal tracking in vivo. For scientists working within the field of iPSCs, this protocol provides a walk-through on how to conduct in vitro and in vivo experiments with an iPSCs constitutively expressing luciferase.
Collapse
Affiliation(s)
- Toshinobu Nishimura
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Kouta Niizuma
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiromitsu Nakauchi
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
27
|
Yoshiba N, Edanami N, Ohkura N, Maekawa T, Takahashi N, Tsuzuno T, Maeda T, Tabeta K, Izumi K, Noiri Y, Yoshiba K. Laminin Isoforms in Human Dental Pulp: Lymphatic Vessels Express Laminin-332, and Schwann Cell-Associated Laminin-211 Modulates CD163 Expression of M2-like Macrophages. Immunohorizons 2021; 5:1008-1020. [PMID: 34965967 DOI: 10.4049/immunohorizons.2100110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 11/19/2022] Open
Abstract
Laminin, a basement membrane heterotrimeric glycoprotein composed of α/β/γ subunits, has important tissue-specific functions in the control of cellular behavior. Our recent study showed the colocalization of CD163+ M2-like macrophages with Schwann cells in human dental pulp, leading us to hypothesize that the laminin isoform of Schwann cells is associated with CD163 expression. The present study investigated the distribution of laminin isoforms in human dental pulp and the underlying mechanisms that affect macrophage phenotypes. Immunofluorescence analysis indicated that blood vessels were exclusively positive for laminin α4 and α5, whereas laminin α2 was associated with Schwann cells. Unexpectedly, laminin α3/laminin-332 (α3β3γ2) was detected on lymphatic vessels. In intact and carious teeth, CD163+ cells were associated with laminin α2, whereas CD206 single-positive cells were present inside, outside, and along blood vessels. In vitro incubation of THP-1 macrophages in plates coated with laminin-211/511 or its functionally analogous E8 fragments of α-chain (E8-α) indicated that cell shapes differed between macrophages grown on laminin-211/E8-α2 and macrophages grown on laminin-511/E8-α5. Laminin-211/E8-α2-coated plates upregulated CD163 expression, compared with laminin-511/E8-α5-coated plates. Integrin α3- and integrin α6-neutralizing Abs altered the shape of THP-1 macrophages and upregulated mRNA levels of CD206 and CD163 in macrophages grown on laminin-511; the neutralizing Abs did not affect macrophages grown on laminin-211. These findings suggest that laminin isoforms differentially regulate macrophage behavior via distinct integrin-laminin affinities. Of note, laminin-332 is expressed by pulpal lymphatic vessels, the existence of which has been debated; laminin-211 might have a role in maintaining CD163 expression on macrophages.
Collapse
Affiliation(s)
- Nagako Yoshiba
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Naoki Edanami
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Naoto Ohkura
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tomoki Maekawa
- Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Naoki Takahashi
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takahiro Tsuzuno
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takeyasu Maeda
- Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Koichi Tabeta
- Division of Periodontology, Department of Oral Biological Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kenji Izumi
- Division of Biomimetics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; and
| | - Yuichiro Noiri
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kunihiko Yoshiba
- Division of Oral Science for Health Promotion, Department of Oral Health and Welfare, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
28
|
Liu YC, Ban LK, Lee HHC, Lee HT, Chang YT, Lin YT, Su HY, Hsu ST, Higuchi A. Laminin-511 and recombinant vitronectin supplementation enables human pluripotent stem cell culture and differentiation on conventional tissue culture polystyrene surfaces in xeno-free conditions. J Mater Chem B 2021; 9:8604-8614. [PMID: 34605523 DOI: 10.1039/d1tb01878g] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Human pluripotent stem cells (hPSCs) are typically cultivated on extracellular matrix (ECM) protein-coated dishes in xeno-free culture conditions. We supplemented mixed ECM proteins (laminin-511 and recombinant vitronectin, rVT) in culture medium for hPSC culture on conventional polystyrene dishes. Three hPSC cell lines were successfully cultivated on uncoated polystyrene dishes in medium supplemented with optimal conditions of laminin-511 and rVT. Excellent colony shape and colony size as well as high expansion fold of hPSCs were found under these conditions, whereas the colony size was small and poor expansion fold was found solely on L-511-coated dishes. A small portion of L-511 in the culture medium supported hPSC adhesion and prevented the adhesion from being too strong on the uncoated dishes, and rVT in the culture medium further supported adhesion of hPSCs on the dishes by maintaining their pluripotency. Having the optimal composition of L-511 and rVT in the culture medium was important for generating good hPSC colony shapes and sizes as well as a high expansion fold. After long-term culture of hPSCs on uncoated dishes supplemented with the mixed proteins, the hPSCs successfully showed pluripotent markers and could differentiate into a specific lineage of cells, cardiomyocytes, with high efficiency.
Collapse
Affiliation(s)
- Ya-Chu Liu
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda Rd, Jhongli, Taoyuan, 32001, Taiwan.
| | - Lee-Kiat Ban
- Department of Surgery, Hsinchu Cathay General Hospital, No. 678, Sec 2, Zhonghua Rd, Hsinchu, 30060, Taiwan
| | - Henry Hsin-Chung Lee
- Department of Surgery, Hsinchu Cathay General Hospital, No. 678, Sec 2, Zhonghua Rd, Hsinchu, 30060, Taiwan.,Graduate Institute of Translational and Interdisciplinary Medicine, National Central University, No. 300, Jhongda Rd, Jhongli, Taoyuan, 32001, Taiwan
| | - Hsin-Ting Lee
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda Rd, Jhongli, Taoyuan, 32001, Taiwan.
| | - Yu-Tang Chang
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda Rd, Jhongli, Taoyuan, 32001, Taiwan.
| | - Yun-Ting Lin
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda Rd, Jhongli, Taoyuan, 32001, Taiwan.
| | - Her-Young Su
- Department of Obstetrics and Gynecology, Bobson Yuho Women and Children's Clinic, No. 182, Zhuangjing S. Rd, Zhubei City, Hsinchu 302, Taiwan
| | - Shih-Tien Hsu
- Department of Internal Medicine, Taiwan Landseed Hospital, 77, Kuangtai Road, Pingjen City, Taoyuan 32405, Taiwan
| | - Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda Rd, Jhongli, Taoyuan, 32001, Taiwan. .,R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taoyuan 320, Taiwan
| |
Collapse
|
29
|
Sung TC, Lu MW, Tian Z, Lee HHC, Pan J, Ling QD, Higuchi A. Poly(vinyl alcohol- co-itaconic acid) hydrogels grafted with several designed peptides for human pluripotent stem cell culture and differentiation into cardiomyocytes. J Mater Chem B 2021; 9:7662-7673. [PMID: 34586153 DOI: 10.1039/d1tb01555a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We developed poly(vinyl alcohol-co-itaconic acid) (PV) hydrogels grafted with laminin-derived peptides that had different joint segments and several specific designs, including dual chain motifs. PV hydrogels grafted with a peptide derived from laminin-β4 (PMQKMRGDVFSP) containing a joint segment, dual chain motif and cationic amino acid insertion could attach human pluripotent stem (hPS) cells and promoted high expansion folds in long-term culture (over 10 passages) with low differentiation rates, whereas hPS cells attached poorly on PV hydrogels grafted with laminin-α5 peptides that had joint segments with and without a cationic amino acid or on PV hydrogels grafted with laminin-β4 peptides containing the joint segment only. The inclusion of a cationic amino acid in the laminin-β4 peptide was critical for hPS cell attachment on PV hydrogels, which contributed to the zeta potential shifting to higher values (3-4 mV enhancement). The novel peptide segment-grafted PV hydrogels developed in this study supported hPS cell proliferation, which induced better hPS cell expansion than recombinant vitronectin-coated dishes (gold standard of hPS cell culture dishes) in xeno-free culture conditions. After long-term culture on peptide-grafted hydrogels, hPS cells could be induced to differentiate into specific lineages of cells, such as cardiomyocytes, with high efficiency.
Collapse
Affiliation(s)
- Tzu-Cheng Sung
- School of Ophthalmology and Optometry, The Eye Hospital of Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Ming-Wei Lu
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan.
| | - Zeyu Tian
- School of Ophthalmology and Optometry, The Eye Hospital of Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Henry Hsin-Chung Lee
- Department of Surgery, Hsinchu Cathay General Hospital, No. 678, Sec 2, Zhonghua Rd., Hsinchu, 30060, Taiwan.,Graduate Institute of Translational and Interdisciplinary Medicine, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan
| | - Jiandong Pan
- School of Ophthalmology and Optometry, The Eye Hospital of Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Qing-Dong Ling
- Cathay Medical Research Institute, Cathay General Hospital, No. 32, Ln 160, Jian-Cheng Road, Hsi-Chi City, Taipei 221, Taiwan
| | - Akon Higuchi
- School of Ophthalmology and Optometry, The Eye Hospital of Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China. .,Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan. .,R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taoyuan 320, Taiwan.,Nano Medical Engineering Laboratory, Riken Cluster for Pioneering Research, Riken, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.,Department of Reproduction, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| |
Collapse
|
30
|
Feeder-Free Human Induced Pluripotent Stem Cell Culture Using a DNA Aptamer-Based Mimic of Basic Fibroblast Growth Factor. Methods Mol Biol 2021; 2312:301-305. [PMID: 34228298 DOI: 10.1007/978-1-0716-1441-9_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Cell culture media are often supplemented with recombinant growth factors and cytokines to reproduce biological conditions in vitro. Basic fibroblast growth factor (bFGF) has been widely used to support the pluripotency and self-renewal activity of human induced pluripotent stem cells (hiPSCs). We had previously developed a synthetic surrogate for bFGF on the basis of a DNA aptamer that binds to one of the FGF receptors. Since DNA aptamers have advantages over recombinant proteins in terms of thermal stability and production cost, replacing recombinant growth factors in cell culture media with DNA aptamers would be of great interest. Herein, we describe our protocol for feeder-free hiPSC culture using a DNA aptamer-based mimic of bFGF.
Collapse
|
31
|
Skorik C, Mullin NK, Shi M, Zhang Y, Hunter P, Tang Y, Hilton B, Schlaeger TM. Xeno-Free Reprogramming of Peripheral Blood Mononuclear Erythroblasts on Laminin-521. ACTA ACUST UNITED AC 2021; 52:e103. [PMID: 31977148 DOI: 10.1002/cpsc.103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Translating human induced pluripotent stem cell (hiPSC)-derived cells and tissues into the clinic requires streamlined and reliable production of clinical-grade hiPSCs. This article describes an entirely animal component-free procedure for the reliable derivation of stable hiPSC lines from donor peripheral blood mononuclear cells (PBMCs) using only autologous patient materials and xeno-free reagents. PBMCs are isolated from a whole blood donation, from which a small amount of patient serum is also generated. The PBMCs are then expanded prior to reprogramming in an animal component-free erythroblast growth medium supplemented with autologous patient serum, thereby eliminating the need for animal serum. After expansion, the erythroblasts are reprogrammed using either cGMP-grade Sendai viral particles (CytoTune™ 2.1 kit) or episomally replicating reprogramming plasmids (Epi5™ kit), both commercially available. Expansion of emerging hiPSCs on a recombinant cGMP-grade human laminin substrate is compatible with a number of xeno-free or chemically defined media (some available as cGMP-grade reagents), such as E8, Nutristem, Stemfit, or mTeSR Plus. hiPSC lines derived using this method display expression of expected surface markers and transcription factors, loss of the reprogramming agent-derived nucleic acids, genetic stability, and the ability to robustly differentiate in vitro to multiple lineages. © 2020 by John Wiley & Sons, Inc. Basic Protocol 1: Isolating peripheral blood mononuclear cells using CPT tubes Support Protocol 1: Removal of clotting factors to produce serum from autologous plasma collected in Basic Protocol 1 Basic Protocol 2: PBMC expansion in an animal-free erythroblast expansion medium containing autologous serum Basic Protocol 3: Reprogramming of expanded PBMCs with Sendai viral reprogramming particles Alternate Protocol: Reprogramming of expanded PBMCs with episomal plasmids Basic Protocol 4: Picking, expanding, and cryopreserving hiPSC clones Support Protocol 2: Testing Sendai virus kit-reprogrammed hiPSC for absence of Sendai viral RNA Support Protocol 3: Testing Epi5 kit-reprogrammed hiPSC for absence of episomal plasmid DNA Support Protocol 4: Assessing the undifferentiated state of human pluripotent stem cell cultures by multi-color immunofluorescent staining and confocal imaging Support Protocol 5: Coating plates with extracellular matrices to support hiPSC attachment and expansion.
Collapse
Affiliation(s)
- Christian Skorik
- Stem Cell Core Facility, Boston Children's Hospital, Stem Cell Program, Boston, Massachusetts.,Stemcell Technologies, Cambridge, Massachusetts
| | - Nathaniel K Mullin
- Stem Cell Core Facility, Boston Children's Hospital, Stem Cell Program, Boston, Massachusetts.,Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Michael Shi
- Stem Cell Core Facility, Boston Children's Hospital, Stem Cell Program, Boston, Massachusetts.,School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Yosra Zhang
- Stem Cell Core Facility, Boston Children's Hospital, Stem Cell Program, Boston, Massachusetts.,Stemcell Technologies, Cambridge, Massachusetts
| | - Phoebe Hunter
- Stem Cell Core Facility, Boston Children's Hospital, Stem Cell Program, Boston, Massachusetts
| | - Yang Tang
- Stem Cell Core Facility, Boston Children's Hospital, Stem Cell Program, Boston, Massachusetts
| | - Brianna Hilton
- Stem Cell Core Facility, Boston Children's Hospital, Stem Cell Program, Boston, Massachusetts
| | - Thorsten M Schlaeger
- Stem Cell Core Facility, Boston Children's Hospital, Stem Cell Program, Boston, Massachusetts.,Harvard Stem Cell Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
32
|
Fukunaga I, Oe Y, Danzaki K, Ohta S, Chen C, Shirai K, Kawano A, Ikeda K, Kamiya K. Modeling gap junction beta 2 gene-related deafness with human iPSC. Hum Mol Genet 2021; 30:1429-1442. [PMID: 33997905 DOI: 10.1093/hmg/ddab097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023] Open
Abstract
There are >120 forms of non-syndromic deafness associated with identified genetic loci. In particular, mutation of the gap junction beta 2 gene (GJB2), which encodes connexin (CX)26 protein, is the most frequent cause of hereditary deafness worldwide. We previously described an induction method to develop functional CX26 gap junction-forming cells from mouse-induced pluripotent stem cells (iPSCs) and generated in vitro models for GJB2-related deafness. However, functional CX26 gap junction-forming cells derived from human iPSCs or embryonic stem cells (ESCs) have not yet been reported. In this study, we generated human iPSC-derived functional CX26 gap junction-forming cells (iCX26GJCs), which have the characteristics of cochlear supporting cells. These iCX26GJCs had gap junction plaque-like formations at cell-cell borders and co-expressed several markers that are expressed in cochlear supporting cells. Furthermore, we generated iCX26GJCs derived from iPSCs from two patients with the most common GJB2 mutation in Asia, and these cells reproduced the pathology of GJB2-related deafness. These in vitro models may be useful for establishing optimal therapies and drug screening for various mutations in GJB2-related deafness.
Collapse
Affiliation(s)
- Ichiro Fukunaga
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 1138421, Japan
| | - Yoko Oe
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 1138421, Japan
| | - Keiko Danzaki
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 1138421, Japan
| | - Sayaka Ohta
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 1138421, Japan
| | - Cheng Chen
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 1138421, Japan
| | - Kyoko Shirai
- Department of Otolaryngology, Head and Neck Surgery, Tokyo Medical University, Tokyo 1600023, Japan
| | - Atsushi Kawano
- Department of Otolaryngology, Head and Neck Surgery, Tokyo Medical University, Tokyo 1600023, Japan
| | - Katsuhisa Ikeda
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 1138421, Japan
| | - Kazusaku Kamiya
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 1138421, Japan
| |
Collapse
|
33
|
Hattori A, Ohta E, Nagai M, Iwabuchi K, Okano H. A new approach to analysis of intracellular proteins and subcellular localization using cellprofiler and imageJ in combination. Methods 2021; 203:233-241. [PMID: 33915291 DOI: 10.1016/j.ymeth.2021.04.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 04/13/2021] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
Analytical pipeline, which is used for various analysis application, of CellProfiler, an open-source software for cell imaging analysis, is very important. In the present study, to examine whether intracellular proteins can be discriminated using a combination of CellProfiler and ImageJ, we analyzed neuroblastoma and monocytic cell lines, and disease-specific induced pluripotent stem cell (iPSC)-derived neurons. This revealed that scattered puncta of Rab7 and transferrin in neuroblastoma lines were clearly detectable by created analytical pipelines in CellProfiler. We then constructed pipelines for measuring the distance from the center of the nucleus to allow investigation of the intracellular localization of Rab7 or transferrin. Using CellProfiler and ImageJ in combination, we confirmed that our pipelines were applicable both quantitatively and objectively to analysis of membrane trafficking of proteins such as Rab proteins and transferrin. In addition, when applied to quantitative measurement of phagocytosis, our pipelines clearly detected monocytic cell lines that had engulfed bioparticles. Finally, we developed new pipelines for analysis of disease phenotype using iPSCs from a patient with familial Parkinson's disease (PD), harboring the I2020T LRRK2 mutation (PARK8). These were able to successfully detect Rab5 puncta and Rab7 puncta in PARK8 patient iPSC-derived neurons. Interestingly, in long-term culture, we found that the numbers of Rab7 puncta in a single PARK8 patient iPSC-derived neurons were lower than that of control iPSC-derived neurons. On the other hands, at 14 days in vitro, the numbers of Rab5 puncta in PARK8 patient iPSC-derived neurons were lower than those of isogenic iPSC-derived neurons, but not Rab7 puncta. Furthermore, Rab5 puncta of PARK8 patient iPSC-derived neurons exhibited distinct localization pattern relative to isogenic iPSC-derived neurons. These present results suggest that this new analytical tool can be used as a supporting method for quantification of intracellular protein.
Collapse
Affiliation(s)
- Akito Hattori
- Program in Cellular Immunology, Graduate School of Medical Science, Kitasato University, Kanagawa, Japan
| | - Etsuro Ohta
- R & D Center for Cell Design, Institute for Regenerative Medicine and Cell Design, Kitasato University School of Allied Health Sciences,Kanagawa, Japan; Department of ImmunologyⅡ, Kitasato University of Allied Health Science, Kanagawa, Japan; Division of Clinical Immunology, Graduate School of Medical Science, Kitasato University, Kanagawa, Japan; Department of Physiology, Keio University School of Medicine, Tokyo, Japan.
| | - Makiko Nagai
- Department of Neurology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Kazuya Iwabuchi
- Program in Cellular Immunology, Graduate School of Medical Science, Kitasato University, Kanagawa, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
34
|
Efficient Induction of Primate iPS Cells Using a Combination of RNA Transfection and Chemical Compounds. Methods Mol Biol 2021. [PMID: 33772459 DOI: 10.1007/7651_2021_373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Non-human primate induced pluripotent cells (iPS cells) are useful for preclinical studies of iPS cell-based therapies and the research of primate developments. Since the initial report of iPS cells in 2006, various iPS cell induction methods have been reported. Here, we describe an efficient method for inducing iPS cells using a combination of RNA transfection and chemical compounds without using transgenes. Many kinds of marmoset cells, including difficult-to-reprogram cells, can be converted into iPS cells using this combinatorial method. Furthermore, this method can be applied to other primates, including humans.
Collapse
|
35
|
Yuzuriha A, Nakamura S, Sugimoto N, Kihara S, Nakagawa M, Yamamoto T, Sekiguchi K, Eto K. Extracellular laminin regulates hematopoietic potential of pluripotent stem cells through integrin β1-ILK-β-catenin-JUN axis. Stem Cell Res 2021; 53:102287. [PMID: 33813173 DOI: 10.1016/j.scr.2021.102287] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 12/21/2022] Open
Abstract
Recombinant matrices have enabled feeder cell-free maintenance cultures of human pluripotent stem cells (hPSCs), with laminin 511-E8 fragment (LM511-E8) being widely used. However, we herein report that hPSCs maintained on LM511-E8 resist differentiating to multipotent hematopoietic progenitor cells (HPCs), unlike hPSCs maintained on LM421-E8 or LM121-E8. The latter two LM-E8s bound weakly to hPSCs compared with LM511-E8 and activated the canonical Wnt/β-catenin signaling pathway. Moreover, the extracellular LM-E8-dependent preferential hematopoiesis was associated with a higher expression of integrin β1 (ITGB1) and downstream integrin-linked protein kinase (ILK), β-catenin and phosphorylated JUN. Accordingly, the lower coating concentration of LM511-E8 or addition of a Wnt/β-catenin signaling activator, CHIR99021, facilitated higher HPC yield. In contrast, the inhibition of ILK, Wnt or JNK by inhibitors or mRNA knockdown suppressed the HPC yield. These findings suggest that extracellular laminin scaffolds modulate the hematopoietic differentiation potential of hPSCs by activating the ITGB1-ILK-β-catenin-JUN axis at the undifferentiated stage. Finally, the combination of low-concentrated LM511-E8 and a revised hPSC-sac method, which adds bFGF, SB431542 and heparin to the conventional method, enabled a higher yield of HPCs and higher rate for definitive hematopoiesis, suggesting a useful protocol for obtaining differentiated hematopoietic cells from hPSCs in general.
Collapse
Affiliation(s)
- Akinori Yuzuriha
- Department of Clinical Application, CiRA, Kyoto University, Kyoto, Japan
| | - Sou Nakamura
- Department of Clinical Application, CiRA, Kyoto University, Kyoto, Japan
| | - Naoshi Sugimoto
- Department of Clinical Application, CiRA, Kyoto University, Kyoto, Japan
| | - Shunsuke Kihara
- Department of Fundamental Cell Technology, CiRA, Kyoto University, Kyoto, Japan
| | - Masato Nakagawa
- Department of Life Science Frontiers, CiRA, Kyoto University, Kyoto, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, CiRA, Kyoto University, Kyoto, Japan; Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto 606-8501, Japan; AMED-CREST, AMED 1-7-1 Otemachi, Chiyodaku, Tokyo 100-0004, Japan
| | - Kiyotoshi Sekiguchi
- Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Japan
| | - Koji Eto
- Department of Clinical Application, CiRA, Kyoto University, Kyoto, Japan; Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba, Japan.
| |
Collapse
|
36
|
Anzai T, Hara H, Chanthra N, Sadahiro T, Ieda M, Hanazono Y, Uosaki H. Generation of Efficient Knock-in Mouse and Human Pluripotent Stem Cells Using CRISPR-Cas9. Methods Mol Biol 2021; 2320:247-259. [PMID: 34302663 DOI: 10.1007/978-1-0716-1484-6_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
A knock-in can generate fluorescent or Cre-reporter under the control of an endogenous promoter. It also generates knock-out or tagged-protein with fluorescent protein and short tags for tracking and purification. Recent advances in genome editing with clustered regularly interspaced short palindromic repeat (CRISPR) and CRISPR-associated protein 9 (Cas9) significantly increased the efficiencies of making knock-in cells. Here we describe the detailed protocols of generating knock-in mouse and human pluripotent stem cells (PSCs) by electroporation and lipofection, respectively.
Collapse
Affiliation(s)
- Tatsuya Anzai
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan
| | - Hiromasa Hara
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
- Animal Resource Laboratory, Center for Development of Advanced Medical Technology, Jichi Medical University, Tochigi, Japan
| | - Nawin Chanthra
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Taketaro Sadahiro
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masaki Ieda
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yutaka Hanazono
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
- Translational Research Laboratory, Center for Development of Advanced Medical Technology, Jichi Medical University, Tochigi, Japan
| | - Hideki Uosaki
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan.
- Translational Research Laboratory, Center for Development of Advanced Medical Technology, Jichi Medical University, Tochigi, Japan.
| |
Collapse
|
37
|
Abulaiti M, Yalikun Y, Murata K, Sato A, Sami MM, Sasaki Y, Fujiwara Y, Minatoya K, Shiba Y, Tanaka Y, Masumoto H. Establishment of a heart-on-a-chip microdevice based on human iPS cells for the evaluation of human heart tissue function. Sci Rep 2020. [DOI: 10.1201/9781420010138] [Citation(s) in RCA: 1419] [Impact Index Per Article: 283.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Abstract
Human iPS cell (iPSC)-derived cardiomyocytes (CMs) hold promise for drug discovery for heart diseases and cardiac toxicity tests. To utilize human iPSC-derived CMs, the establishment of three-dimensional (3D) heart tissues from iPSC-derived CMs and other heart cells, and a sensitive bioassay system to depict physiological heart function are anticipated. We have developed a heart-on-a-chip microdevice (HMD) as a novel system consisting of dynamic culture-based 3D cardiac microtissues derived from human iPSCs and microelectromechanical system (MEMS)-based microfluidic chips. The HMDs could visualize the kinetics of cardiac microtissue pulsations by monitoring particle displacement, which enabled us to quantify the physiological parameters, including fluidic output, pressure, and force. The HMDs demonstrated a strong correlation between particle displacement and the frequency of external electrical stimulation. The transition patterns were validated by a previously reported versatile video-based system to evaluate contractile function. The patterns are also consistent with oscillations of intracellular calcium ion concentration of CMs, which is a fundamental biological component of CM contraction. The HMDs showed a pharmacological response to isoproterenol, a β-adrenoceptor agonist, that resulted in a strong correlation between beating rate and particle displacement. Thus, we have validated the basic performance of HMDs as a resource for human iPSC-based pharmacological investigations.
Collapse
|
38
|
Abulaiti M, Yalikun Y, Murata K, Sato A, Sami MM, Sasaki Y, Fujiwara Y, Minatoya K, Shiba Y, Tanaka Y, Masumoto H. Establishment of a heart-on-a-chip microdevice based on human iPS cells for the evaluation of human heart tissue function. Sci Rep 2020; 10:19201. [PMID: 33154509 PMCID: PMC7645446 DOI: 10.1038/s41598-020-76062-w] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/23/2020] [Indexed: 12/04/2022] Open
Abstract
Human iPS cell (iPSC)-derived cardiomyocytes (CMs) hold promise for drug discovery for heart diseases and cardiac toxicity tests. To utilize human iPSC-derived CMs, the establishment of three-dimensional (3D) heart tissues from iPSC-derived CMs and other heart cells, and a sensitive bioassay system to depict physiological heart function are anticipated. We have developed a heart-on-a-chip microdevice (HMD) as a novel system consisting of dynamic culture-based 3D cardiac microtissues derived from human iPSCs and microelectromechanical system (MEMS)-based microfluidic chips. The HMDs could visualize the kinetics of cardiac microtissue pulsations by monitoring particle displacement, which enabled us to quantify the physiological parameters, including fluidic output, pressure, and force. The HMDs demonstrated a strong correlation between particle displacement and the frequency of external electrical stimulation. The transition patterns were validated by a previously reported versatile video-based system to evaluate contractile function. The patterns are also consistent with oscillations of intracellular calcium ion concentration of CMs, which is a fundamental biological component of CM contraction. The HMDs showed a pharmacological response to isoproterenol, a β-adrenoceptor agonist, that resulted in a strong correlation between beating rate and particle displacement. Thus, we have validated the basic performance of HMDs as a resource for human iPSC-based pharmacological investigations.
Collapse
Affiliation(s)
- Mosha Abulaiti
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, 650-0047, Japan.,Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Laboratory for Integrated Biodevice, RIKEN Center for Biosystems Dynamics Research, Suita, Japan
| | - Yaxiaer Yalikun
- Laboratory for Integrated Biodevice, RIKEN Center for Biosystems Dynamics Research, Suita, Japan
| | - Kozue Murata
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, 650-0047, Japan.,Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Asako Sato
- Laboratory for Integrated Biodevice, RIKEN Center for Biosystems Dynamics Research, Suita, Japan
| | - Mustafa M Sami
- Laboratory for Morphogenetic Signaling, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Yuko Sasaki
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, 650-0047, Japan
| | - Yasue Fujiwara
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, 650-0047, Japan.,Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuji Shiba
- Department of Regenerative Science and Medicine, Institute for Biomedical Sciences, Shinshu University, Matsumoto, Japan
| | - Yo Tanaka
- Laboratory for Integrated Biodevice, RIKEN Center for Biosystems Dynamics Research, Suita, Japan
| | - Hidetoshi Masumoto
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, 650-0047, Japan. .,Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
39
|
Sekine K, Ogawa S, Tsuzuki S, Kobayashi T, Ikeda K, Nakanishi N, Takeuchi K, Kanai E, Otake Y, Okamoto S, Kobayashi T, Takebe T, Taniguchi H. Generation of human induced pluripotent stem cell-derived liver buds with chemically defined and animal origin-free media. Sci Rep 2020; 10:17937. [PMID: 33087763 PMCID: PMC7578079 DOI: 10.1038/s41598-020-73908-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 09/23/2020] [Indexed: 01/09/2023] Open
Abstract
Advances in organoid technology have broadened the number of target diseases and conditions in which human induced pluripotent stem cell (iPSC)-based regenerative medicine can be applied; however, mass production of organoids and the development of chemically defined, animal origin-free (CD-AOF) media and supplements are unresolved issues that hamper the clinical applicability of these approaches. CD-AOF media and supplements ensure the quality and reproducibility of culture systems by lowering lot-to-lot variations and the risk of contamination with viruses or toxins. We previously generated liver organoids from iPSCs, namely iPSC-liver buds (iPSC-LBs), by mimicking the organogenic interactions among hepatocytes, endothelial cells (ECs), and mesenchymal cells (MCs) and recently reported the mass production of iPSC-LBs derived entirely from iPSCs (all iPSC-LBs), which should facilitate their large-scale production for the treatment of liver failure. However, in previous studies we used media originating from animals for differentiation except for the maintenance of undifferentiated iPSCs. Therefore, we developed a CD-AOF medium to generate all iPSC-LBs. We first developed a CD-AOF medium for hepatocytes, ECs, and stage-matched MCs, i.e., septum transversum mesenchyme (STM), in 2D cultures. We next generated all iPSC-LBs by incubating individual cell types in ultra-low attachment micro-dimple plates. The hepatic functions of all iPSC-LBs generated using the CD-AOF medium were equivalent to those of all iPSC-LBs generated using the conventional medium both in vitro and in vivo. Furthermore, we found that this CD-AOF medium could be used in several cell culture settings. Taken together, these results demonstrate the successful development of a CD-AOF medium suitable for all iPSC-LBs. The protocol developed in this study will facilitate the clinical applicability of all iPSC-LBs in the treatment of liver diseases.
Collapse
Affiliation(s)
- Keisuke Sekine
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan. .,Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan.
| | - Shimpei Ogawa
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co., Inc., Kawasaki, 210-8681, Japan
| | - Syusaku Tsuzuki
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.,Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Tatsuya Kobayashi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Kazuki Ikeda
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Noriko Nakanishi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Kenta Takeuchi
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.,Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Eriko Kanai
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.,Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Yugo Otake
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Satoshi Okamoto
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Tsuyoshi Kobayashi
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co., Inc., Kawasaki, 210-8681, Japan
| | - Takanori Takebe
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan.,Advanced Medical Research Center, Yokohama City University, Yokohama, 236-0004, Japan.,Division of Gastroenterology, Hepatology and Nutrition, Developmental Biology, Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229-3039, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229-3039, USA
| | - Hideki Taniguchi
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan. .,Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan. .,Advanced Medical Research Center, Yokohama City University, Yokohama, 236-0004, Japan.
| |
Collapse
|
40
|
Kawase E, Takada K, Suemori H. Kyoto hESC cell resource for regenerative medicine. Stem Cell Res 2020; 49:102020. [PMID: 33059130 DOI: 10.1016/j.scr.2020.102020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 05/03/2020] [Accepted: 09/28/2020] [Indexed: 10/23/2022] Open
Affiliation(s)
- Eihachiro Kawase
- Division of Clinical Basis for ES Cell Research, Center for Human ES Cell Research, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Kei Takada
- Division of Clinical Basis for ES Cell Research, Center for Human ES Cell Research, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hirofumi Suemori
- Division of Clinical Basis for ES Cell Research, Center for Human ES Cell Research, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
41
|
Watanabe T, Yasuda S, Kusakawa S, Kuroda T, Futamura M, Ogawa M, Mochizuki H, Kikkawa E, Furukawa H, Nagaoka M, Sato Y. Multisite studies for validation and improvement of a highly efficient culture assay for detection of undifferentiated human pluripotent stem cells intermingled in cell therapy products. Cytotherapy 2020; 23:176-183. [PMID: 32978066 DOI: 10.1016/j.jcyt.2020.07.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND AIMS The Multisite Evaluation Study on Analytical Methods for Non-Clinical Safety Assessment of Human-Derived Regenerative Medical Products (MEASURE) is a Japanese experimental public-private partnership initiative, which aims to standardize methodology for tumorigenicity evaluation of human pluripotent stem cell (hPSC)-derived cell therapy products (CTPs). Undifferentiated hPSCs possess tumorigenic potential, and thus residual undifferentiated hPSCs are one of the major hazards for the risk of tumor formation from hPSC-derived CTPs. Among currently available assays, a highly efficient culture (HEC) assay is reported to be one of the most sensitive for the detection of residual undifferentiated hPSCs. METHODS MEASURE first validated the detection sensitivity of HEC assay and then investigated the feasibility of magnetic-activated cell sorting (MACS) to improve sensitivity. RESULTS The multisite experiments confirmed that the lower limit of detection under various conditions to which the human induced pluripotent stem cell lines and culture medium/substrate were subjected was 0.001%. In addition, MACS concentrated cells expressing undifferentiated cell markers and consequently achieved a detection sensitivity of 0.00002%. CONCLUSIONS These results indicate that HEC assay is highly sensitive and robust and that the application of MACS on this assay is a promising tool for further mitigation of the potential tumorigenicity risk of hPSC-derived CTPs.
Collapse
Affiliation(s)
- Takeshi Watanabe
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, Fujisawa, Japan; The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan.
| | - Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| | - Shinji Kusakawa
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| | - Takuya Kuroda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| | - Mayumi Futamura
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Drug Discovery Support Division, Tsukuba Research Institute, BoZo Research Center Inc, Tsukuba, Japan
| | - Mitsuhide Ogawa
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; CMIC Bioresearch Center, CMIC Pharma Science Co, Ltd, Hokuto, Japan
| | - Hidemi Mochizuki
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Research Planning Section, Ina Research Inc, Ina-shi, Japan
| | - Eri Kikkawa
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Research Division, HEALIOS K.K., Kobe, Japan
| | - Hatsue Furukawa
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Integrated & Translational Science, Axcelead Drug Discovery Partners, Inc, Fujisawa, Japan
| | - Masato Nagaoka
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Life Science Research Laboratory, Tosoh Corporation, Ayase-shi, Japan
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| |
Collapse
|
42
|
Koterazawa Y, Koyanagi-Aoi M, Uehara K, Kakeji Y, Aoi T. Retinoic acid receptor γ activation promotes differentiation of human induced pluripotent stem cells into esophageal epithelium. J Gastroenterol 2020; 55:763-774. [PMID: 32556644 PMCID: PMC7376085 DOI: 10.1007/s00535-020-01695-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/02/2020] [Indexed: 02/04/2023]
Abstract
BACKGROUND The esophagus is known to be derived from the foregut. However, the mechanisms regulating this process remain unclear. In particular, the details of the human esophagus itself have been poorly researched. In this decade, studies using human induced pluripotent stem cells (hiPSCs) have proven powerful tools for clarifying the developmental biology of various human organs. Several studies using hiPSCs have demonstrated that retinoic acid (RA) signaling promotes the differentiation of foregut into tissues such as lung and pancreas. However, the effect of RA signaling on the differentiation of foregut into esophagus remains unclear. METHODS We established a novel stepwise protocol with transwell culture and an air-liquid interface system for esophageal epithelial cell (EEC) differentiation from hiPSCs. We then evaluated the effect of all-trans retinoic acid (ATRA), which is a retinoic acid receptor (RAR)α, RARβ and RARγ agonist, on the differentiation from the hiPSC-derived foregut. Finally, to identify which RAR subtype was involved in the differentiation, we used synthetic agonists and antagonists of RARα and RARγ, which are known to be expressed in esophagus. RESULTS We successfully generated stratified layers of cells expressing EEC marker genes that were positive for lugol staining. The enhancing effect of ATRA on EEC differentiation was clearly demonstrated with quantitative reverse transcription polymerase chain reaction, immunohistology, lugol-staining and RNA sequencing analyses. RARγ agonist and antagonist enhanced and suppressed EEC differentiation, respectively. RARα agonist had no effect on the differentiation. CONCLUSION We revealed that RARγ activation promotes the differentiation of hiPSCs-derived foregut into EECs.
Collapse
Affiliation(s)
- Yasufumi Koterazawa
- Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
- Department of iPS Cell Applications, Graduate School of Medicine, Kobe University, Kobe, Japan
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Michiyo Koyanagi-Aoi
- Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
- Department of iPS Cell Applications, Graduate School of Medicine, Kobe University, Kobe, Japan
- Center for Human Resource Development for Regenerative Medicine, Kobe University Hospital, Kobe, Japan
| | - Keiichiro Uehara
- Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
- Department of iPS Cell Applications, Graduate School of Medicine, Kobe University, Kobe, Japan
- Division of Pathology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Yoshihiro Kakeji
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Takashi Aoi
- Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
- Department of iPS Cell Applications, Graduate School of Medicine, Kobe University, Kobe, Japan.
- Center for Human Resource Development for Regenerative Medicine, Kobe University Hospital, Kobe, Japan.
| |
Collapse
|
43
|
Nishimura T, Xu H, Iwasaki M, Karigane D, Saavedra B, Takahashi Y, Suchy FP, Monobe S, Martin RM, Ohtaka M, Nakanishi M, Burrows SR, Cleary ML, Majeti R, Shibuya A, Nakauchi H. Sufficiency for inducible Caspase-9 safety switch in human pluripotent stem cells and disease cells. Gene Ther 2020; 27:525-534. [PMID: 32704085 DOI: 10.1038/s41434-020-0179-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 06/27/2020] [Accepted: 07/14/2020] [Indexed: 12/17/2022]
Abstract
Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have promising potential for opening new avenues in regenerative medicine. However, since the tumorigenic potential of undifferentiated pluripotent stem cells (PSCs) is a major safety concern for clinical transplantation, inducible Caspase-9 (iC9) is under consideration for use as a fail-safe system. Here, we used targeted gene editing to introduce the iC9 system into human iPSCs, and then interrogated the efficiency of inducible apoptosis with normal iPSCs as well as diseased iPSCs derived from patients with acute myeloid leukemia (AML-iPSCs). The iC9 system induced quick and efficient apoptosis to iPSCs in vitro. More importantly, complete eradication of malignant cells without AML recurrence was shown in disease mouse models by using AML-iPSCs. In parallel, it shed light on several limitations of the iC9 system usage. Our results suggest that careful use of the iC9 system will serve as an important countermeasure against posttransplantation adverse events in stem cell transplantation therapies.
Collapse
Affiliation(s)
- Toshinobu Nishimura
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Haojun Xu
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.,Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Masayuki Iwasaki
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Department of Advanced Health Science, Institute of Laboratory Animals, Tokyo Women's Medical University, Tokyo, 162-8666, Japan
| | - Daiki Karigane
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Brandon Saavedra
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Yusuke Takahashi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Fabian P Suchy
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Shinichiro Monobe
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Renata M Martin
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Manami Ohtaka
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, 305-8562, Japan
| | - Mahito Nakanishi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, 305-8562, Japan
| | - Scott R Burrows
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | - Michael L Cleary
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ravindra Majeti
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.,Life Science Center of Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Hiromitsu Nakauchi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA. .,Division of Stem Cell Therapy, Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.
| |
Collapse
|
44
|
Negoro R, Kawai K, Ichikawa M, Deguchi S, Takayama K, Mizuguchi H. Establishment of MDR1-knockout human induced pluripotent stem cell line. Drug Metab Pharmacokinet 2020; 35:288-296. [PMID: 32303458 DOI: 10.1016/j.dmpk.2020.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/28/2020] [Accepted: 01/31/2020] [Indexed: 12/14/2022]
Abstract
Multiple drug resistance 1 (MDR1) is highly expressed in various organs, including the liver, small intestine, and blood-brain barrier (BBB). Because MDR1 plays important roles in the excretion of many drugs, it is necessary to evaluate whether drug candidates are potential substrates of MDR1. Recently, many researchers have shown that human induced pluripotent stem (iPS) cell-derived differentiated cells such as hepatocytes and enterocytes can be applied for pharmacokinetic testing. Here, we attempted to generate MDR1-knockout (KO) iPS cell lines using genome editing technology. The correctly targeted human iPS cell lines were successfully obtained. The expression levels of pluripotent markers in human iPS cells were not changed by MDR1 knockout. The gene expression levels of hepatic markers in MDR1-KO iPS-derived hepatocyte-like cells were higher than those in undifferentiated MDR1-KO iPS cells, suggesting that MDR1-KO iPS cells have hepatic differentiation capacity. In addition, MDR1 expression levels were hardly detected in MDR1-KO iPS cell-derived hepatocyte-like cells. We thus succeeded in establishing MDR1-KO iPS cell lines that could be utilized for pharmacokinetic testing.
Collapse
Affiliation(s)
- Ryosuke Negoro
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Kanae Kawai
- Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Moe Ichikawa
- Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Sayaka Deguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan
| | - Kazuo Takayama
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan; PRESTO, Japan Science and Technology Agency, Saitama, 332-0012, Japan.
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Osaka, 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, 565-0871, Japan.
| |
Collapse
|
45
|
Ishikawa M, Aoyama T, Shibata S, Sone T, Miyoshi H, Watanabe H, Nakamura M, Morota S, Uchino H, Yoo AS, Okano H. miRNA-Based Rapid Differentiation of Purified Neurons from hPSCs Advancestowards Quick Screening for Neuronal Disease Phenotypes In Vitro. Cells 2020; 9:E532. [PMID: 32106535 PMCID: PMC7140514 DOI: 10.3390/cells9030532] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 12/12/2022] Open
Abstract
Obtaining differentiated cells with high physiological functions by an efficient, but simple and rapid differentiation method is crucial for modeling neuronal diseases in vitro using human pluripotent stem cells (hPSCs). Currently, methods involving the transient expression of one or a couple of transcription factors have been established as techniques for inducing neuronal differentiation in a rapid, single step. It has also been reported that microRNAs can function as reprogramming effectors for directly reprogramming human dermal fibroblasts to neurons. In this study, we tested the effect of adding neuronal microRNAs, miRNA-9/9*, and miR-124 (miR-9/9*-124), for the neuronal induction method of hPSCs using Tet-On-driven expression of the Neurogenin2 gene (Ngn2), a proneural factor. While it has been established that Ngn2 can facilitate differentiation from pluripotent stem cells into neurons with high purity due to its neurogenic effect, a long or indefinite time is required for neuronal maturation with Ngn2 misexpression alone. With the present method, the cells maintained a high neuronal differentiation rate while exhibiting increased gene expression of neuronal maturation markers, spontaneous calcium oscillation, and high electrical activity with network bursts as assessed by a multipoint electrode system. Moreover, when applying this method to iPSCs from Alzheimer's disease (AD) patients with presenilin-1 (PS1) or presenilin-2 (PS2) mutations, cellular phenotypes such as increased amount of extracellular secretion of amyloid β42, abnormal oxygen consumption, and increased reactive oxygen species in the cells were observed in a shorter culture period than those previously reported. Therefore, it is strongly anticipated that the induction method combining Ngn2 and miR-9/9*-124 will enable more rapid and simple screening for various types of neuronal disease phenotypes and promote drug discovery.
Collapse
Affiliation(s)
- Mitsuru Ishikawa
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takeshi Aoyama
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shoichiro Shibata
- Department of Anesthesiology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Takefumi Sone
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hiroyuki Miyoshi
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hirotaka Watanabe
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Mari Nakamura
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Saori Morota
- Department of Anesthesiology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Hiroyuki Uchino
- Department of Anesthesiology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Andrew S Yoo
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
46
|
Dayem AA, Won J, Goo HG, Yang GM, Seo DS, Jeon BM, Choi HY, Park SE, Lim KM, Jang SH, Lee SB, Choi SB, Kim K, Kang GH, Yeon GB, Kim DS, Cho SG. The immobilization of fibronectin- and fibroblast growth factor 2-derived peptides on a culture plate supports the attachment and proliferation of human pluripotent stem cells. Stem Cell Res 2020; 43:101700. [PMID: 31981882 DOI: 10.1016/j.scr.2020.101700] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 12/18/2022] Open
Abstract
Pluripotent stem cells (PSCs) offer a promising tool for regenerative medicine. The clinical application of PSCs inevitably requires a large-scale culture in a highly defined environment. The present study aimed to devise defined coating materials for the efficient adhesion and proliferation of human PSCs (hPSCs). We tested the activity of seven fibronectin-derived peptides and three laminin-derived peptides for the attachment and proliferation of hPSCs through their immobilization on the bottom of culture dishes by creating a fusion protein with the mussel adhesion protein. Among the extracellular matrix (ECM) mimetics tested, one fibronectin-derived peptide, PHSRN-GRGDSP, significantly promoted adhesion, enhanced alkaline phosphatase activity, and increased pluripotency-related gene expression in hPSCs compared to Matrigel. Furthermore, co-immobilization of a particular canofin peptide derived from fibroblast growth factor 2 increased pluripotency marker expression, which may offer the possibility of culture without growth factor supplementation. Our findings afford a novel defined condition for the efficient culture of hPSCs and may be utilized in future clinical applications.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jihye Won
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; AMOGREENTECH, 91, Gimpo-daero 1950 beon-gil, Tongjin-eup, Gimpo-si, Gyeonggi-do 10014, Republic of Korea
| | - Hui-Gwan Goo
- AMOGREENTECH, 91, Gimpo-daero 1950 beon-gil, Tongjin-eup, Gimpo-si, Gyeonggi-do 10014, Republic of Korea
| | - Gwang-Mo Yang
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Dong Sik Seo
- AMOGREENTECH, 91, Gimpo-daero 1950 beon-gil, Tongjin-eup, Gimpo-si, Gyeonggi-do 10014, Republic of Korea
| | - Byeong-Min Jeon
- Department of Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Hye Yeon Choi
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sang Eun Park
- AMOGREENTECH, 91, Gimpo-daero 1950 beon-gil, Tongjin-eup, Gimpo-si, Gyeonggi-do 10014, Republic of Korea
| | - Kyung Min Lim
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Seon-Ho Jang
- AMOGREENTECH, 91, Gimpo-daero 1950 beon-gil, Tongjin-eup, Gimpo-si, Gyeonggi-do 10014, Republic of Korea
| | - Soo Bin Lee
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sang Baek Choi
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Kyeongseok Kim
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Geun-Ho Kang
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Gyu-Bum Yeon
- Department of Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Dae-Sung Kim
- Department of Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea; Department of Pediatrics, Korea University College of Medicine, Guro Hospital, 97 Gurodong-gil, Guro-gu, Seoul 08308, Republic of Korea.
| | - Ssang-Goo Cho
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
47
|
Zandrini T, Shan O, Parodi V, Cerullo G, Raimondi MT, Osellame R. Multi-foci laser microfabrication of 3D polymeric scaffolds for stem cell expansion in regenerative medicine. Sci Rep 2019; 9:11761. [PMID: 31409835 PMCID: PMC6692386 DOI: 10.1038/s41598-019-48080-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/22/2019] [Indexed: 11/09/2022] Open
Abstract
High quality large scale fabrication of cellular scaffolds, with three-dimensional resolution comparable to cell size, is an important task to enable regenerative medicine applications with stem cells. We are using two-photon polymerization to produce our stem cell culture substrate called Nichoid, which we already demonstrated capable of stimulating cell proliferation while maintaining their stemness, without the need of dangerous additives. Parallelization of this technique can be achieved with the use of a spatial light modulator: here we show the results obtained combining this device with fast linear stages to produce Nichoid-covered substrates by two-photon polymerization. The well-polymerized structures confirm that this approach is particularly convenient for porous structures, and allows a significant time saving by a factor of almost five, with minor design adjustments. A Live & Dead assay was performed on mesenchymal stem cells cultured into the Nichoid microstructures in order to verify that no difference in cell viability is present, compared to microstructures fabricated by a single focus. This parallel setup opens the possibility to obtain a much larger number of microstructured substrates, that are essential to test new stem cell-based therapies. This approach can be also used for the fast fabrication of other kinds of cell culture devices.
Collapse
Affiliation(s)
- Tommaso Zandrini
- Politecnico di Milano, Department of Chemistry, Materials, and Chemical Engineering "Giulio Natta", Milano, 20133, Italy. .,National Research Council, Institute for Photonics and Nanotechnologies, Milano, 20133, Italy.
| | - Oumin Shan
- Politecnico di Milano, Department of Physics, Milano, 20133, Italy
| | - Valentina Parodi
- Politecnico di Milano, Department of Chemistry, Materials, and Chemical Engineering "Giulio Natta", Milano, 20133, Italy
| | - Giulio Cerullo
- National Research Council, Institute for Photonics and Nanotechnologies, Milano, 20133, Italy.,Politecnico di Milano, Department of Physics, Milano, 20133, Italy
| | - Manuela T Raimondi
- Politecnico di Milano, Department of Chemistry, Materials, and Chemical Engineering "Giulio Natta", Milano, 20133, Italy
| | - Roberto Osellame
- National Research Council, Institute for Photonics and Nanotechnologies, Milano, 20133, Italy.,Politecnico di Milano, Department of Physics, Milano, 20133, Italy
| |
Collapse
|
48
|
Sung TC, Yang JS, Yeh CC, Liu YC, Jiang YP, Lu MW, Ling QD, Kumar SS, Chang Y, Umezawa A, Chen H, Higuchi A. The design of a thermoresponsive surface for the continuous culture of human pluripotent stem cells. Biomaterials 2019; 221:119411. [PMID: 31419657 DOI: 10.1016/j.biomaterials.2019.119411] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/02/2019] [Accepted: 08/03/2019] [Indexed: 01/06/2023]
Abstract
Commonly, stem cell culture is based on batch-type culture, which is laborious and expensive. We continuously cultured human pluripotent stem cells (hPSCs) on thermoresponsive dish surfaces, where hPSCs were partially detached on the same thermoresponsive dish by decreasing the temperature of the thermoresponsive dish to be below the lower critical solution temperature for only 30 min. Then, the remaining cells were continuously cultured in fresh culture medium, and the detached stem cells were harvested in the exchanged culture medium. hPSCs were continuously cultured for ten cycles on the thermoresponsive dish surface, which was prepared by coating the surface with poly(N-isopropylacrylamide-co-styrene) and oligovitronectin-grafted poly(acrylic acid-co-styrene) or recombinant vitronectin for hPSC binding sites to maintain hPSC pluripotency. After ten cycles of continuous culture on the thermoresponsive dish surface, the detached cells expressed pluripotency proteins and had the ability to differentiate into cells derived from the three germ layers in vitro and in vivo. Furthermore, the detached cells differentiated into specific cell lineages, such as cardiomyocytes, with high efficiency.
Collapse
Affiliation(s)
- Tzu-Cheng Sung
- School of Biomedical Engineering, The Eye Hospital of Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China; Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan
| | - Jia-Sin Yang
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan
| | - Chih-Chen Yeh
- Department of Chemical Engineering and R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taoyuan, 320, Taiwan
| | - Ya-Chu Liu
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan
| | - Yi-Peng Jiang
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan
| | - Ming-Wei Lu
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan
| | - Qing-Dong Ling
- Cathay Medical Research Institute, Cathay General Hospital, No. 32, Ln 160, Jian-Cheng Road, Hsi-Chi City, Taipei, 221, Taiwan; Institute of Systems Biology and Bioinformatics, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan
| | - S Suresh Kumar
- Department of Medical Microbiology and Parasitology, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Yung Chang
- Department of Chemical Engineering and R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taoyuan, 320, Taiwan.
| | - Akihiro Umezawa
- Department of Reproduction, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
| | - Hao Chen
- School of Biomedical Engineering, The Eye Hospital of Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China; Wenzhou Institute, University of Chinese Academy of Science, No. 16, Xinsan Road, Hi-tech Industry Park, Wenzhou, Zhejiang, China
| | - Akon Higuchi
- School of Biomedical Engineering, The Eye Hospital of Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China; Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan; Department of Reproduction, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan; Wenzhou Institute, University of Chinese Academy of Science, No. 16, Xinsan Road, Hi-tech Industry Park, Wenzhou, Zhejiang, China; Center for Emergent Matter Science, Riken, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| |
Collapse
|
49
|
Le MNT, Hasegawa K. Expansion Culture of Human Pluripotent Stem Cells and Production of Cardiomyocytes. Bioengineering (Basel) 2019; 6:E48. [PMID: 31137703 PMCID: PMC6632060 DOI: 10.3390/bioengineering6020048] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/15/2019] [Accepted: 05/18/2019] [Indexed: 12/25/2022] Open
Abstract
Transplantation of human pluripotent stem cell (hPSCs)-derived cardiomyocytes for the treatment of heart failure is a promising therapy. In order to implement this therapy requiring numerous cardiomyocytes, substantial production of hPSCs followed by cardiac differentiation seems practical. Conventional methods of culturing hPSCs involve using a 2D culture monolayer that hinders the expansion of hPSCs, thereby limiting their productivity. Advanced culture of hPSCs in 3D aggregates in the suspension overcomes the limitations of 2D culture and attracts immense attention. Although the hPSC production needs to be suitable for subsequent cardiac differentiation, many studies have independently focused on either expansion of hPSCs or cardiac differentiation protocols. In this review, we summarize the recent approaches to expand hPSCs in combination with cardiomyocyte differentiation. A comparison of various suspension culture methods and future prospects for dynamic culture of hPSCs are discussed in this study. Understanding hPSC characteristics in different models of dynamic culture helps to produce numerous cells that are useful for further clinical applications.
Collapse
Affiliation(s)
- Minh Nguyen Tuyet Le
- Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advanced Study, Kyoto University, Kyoto 606-8501, Japan.
| | - Kouichi Hasegawa
- Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advanced Study, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
50
|
Chen LH, Sung TC, Lee HHC, Higuchi A, Su HC, Lin KJ, Huang YR, Ling QD, Kumar SS, Alarfaj AA, Munusamy MA, Nasu M, Chen DC, Hsu ST, Chang Y, Lee KF, Wang HC, Umezawa A. Xeno-free and feeder-free culture and differentiation of human embryonic stem cells on recombinant vitronectin-grafted hydrogels. Biomater Sci 2019; 7:4345-4362. [DOI: 10.1039/c9bm00418a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Xeno-free culture and cardiomyocyte differentiation of human embryonic stem cells on vitronectin-grafted hydrogels by adjusting surface charge and elasticity.
Collapse
|