1
|
Yehya H, Raudins S, Padmanabhan R, Jensen J, Bukys MA. Addressing bioreactor hiPSC aggregate stability, maintenance and scaleup challenges using a design of experiment approach. Stem Cell Res Ther 2024; 15:191. [PMID: 38956608 PMCID: PMC11218057 DOI: 10.1186/s13287-024-03802-4] [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: 10/27/2023] [Accepted: 06/16/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Stem cell-derived therapies hold the potential for treatment of regenerative clinical indications. Static culture has a limited ability to scale up thus restricting its use. Suspension culturing can be used to produce target cells in large quantities, but also presents challenges related to stress and aggregation stability. METHODS Utilizing a design of experiments (DoE) approach in vertical wheel bioreactors, we evaluated media additives that have versatile properties. The additives evaluated are Heparin sodium salt (HS), polyethylene glycol (PEG), poly (vinyl alcohol) (PVA), Pluronic F68 and dextran sulfate (DS). Multiple response variables were chosen to assess cell growth, pluripotency maintenance and aggregate stability in response to the additive inputs, and mathematical models were generated and tuned for maximal predictive power. RESULTS Expansion of iPSCs using 100 ml vertical wheel bioreactor assay for 4 days on 19 different media combinations resulted in models that can optimize pluripotency, stability, and expansion. The expansion optimization resulted in the combination of PA, PVA and PEG with E8. This mixture resulted in an expansion doubling time that was 40% shorter than that of E8 alone. Pluripotency optimizer highlighted the importance of adding 1% PEG to the E8 medium. Aggregate stability optimization that minimizes aggregate fusion in 3D culture indicated that the interaction of both Heparin and PEG can limit aggregation as well as increase the maintenance capacity and expansion of hiPSCs, suggesting that controlling fusion is a critical parameter for expansion and maintenance. Validation of optimized solution on two cell lines in bioreactors with decreased speed of 40 RPM, showed consistency and prolonged control over aggregates that have high frequency of pluripotency markers of OCT4 and SOX2 (> 90%). A doubling time of around 1-1.4 days was maintained after passaging as clumps in the optimized medium. Controlling aggregate fusion allowed for a decrease in bioreactor speed and therefore shear stress exerted on the cells in a large-scale expansion. CONCLUSION This study resulted in a control of aggregate size within suspension cultures, while informing about concomitant state control of the iPSC state. Wider application of this approach can address media optimization complexity and bioreactor scale-up challenges.
Collapse
Affiliation(s)
- Haneen Yehya
- Trailhead Biosystems, 23215 Commerce Park, Beachwood, OH, 44122, USA
- Cleveland State University, 2121 Euclid Ave, Cleveland, OH, 44115, USA
| | - Sofija Raudins
- Trailhead Biosystems, 23215 Commerce Park, Beachwood, OH, 44122, USA
| | | | - Jan Jensen
- Trailhead Biosystems, 23215 Commerce Park, Beachwood, OH, 44122, USA
| | - Michael A Bukys
- Trailhead Biosystems, 23215 Commerce Park, Beachwood, OH, 44122, USA.
| |
Collapse
|
2
|
Mousavi Mirkalaei S, Farivar S. Systematic optimization of culture media for maintenance of human induced pluripotent stem cells using the response surface methodology. Heliyon 2024; 10:e32558. [PMID: 38975108 PMCID: PMC11226774 DOI: 10.1016/j.heliyon.2024.e32558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 07/09/2024] Open
Abstract
The application of human induced pluripotent stem cells (hiPSCs) provides tremendous opportunities in cell therapy. However, culturing these cells faces many practical challenges, including costs associated with cell culture media and the optimization of cell culture conditions. Providing an optimized culture platform for hiPSCs to maintain pluripotency and self-renewal and generate cost-effective and robust therapeutics is an immediate requirement. This study used the design of experiments and the response surface methodology, a powerful statistical tool, to generate empirical models for predicting the optimal culture conditions of the hiPSCs. Pluripotency and cell proliferation were applied as read-outs to determine the optimal concentration of basic fibroblast growth factor (bFGF) and cell density. One model was defined to predict pluripotency and cell proliferation in terms of the predictor variables of the bFGF concentration and cell seeding density. Predicted culture conditions to maximize maintaining cell pluripotency were successfully validated. The present study's findings provide a novel approach that can potentially allow controllable hiPSC culture routine in translational research.
Collapse
Affiliation(s)
- Seyedmilad Mousavi Mirkalaei
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Shirin Farivar
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
3
|
Souralova T, Hulinova D, Jeseta M, Ventruba P, Hampl A, Koutna I. Truncated vitronectin with E-cadherin enables the xeno-free derivation of human embryonic stem cells. Sci Rep 2023; 13:15062. [PMID: 37700192 PMCID: PMC10497536 DOI: 10.1038/s41598-023-42236-5] [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: 06/27/2023] [Accepted: 09/07/2023] [Indexed: 09/14/2023] Open
Abstract
Human embryonic stem cells (hESCs) have unique abilities that enable their use in cell therapy, disease modeling, and drug development. Their derivation is usually performed using a feeder layer, which is undefined and can potentially cause a contamination by xeno components, therefore there is a tendency to replace feeders with xeno-free defined substrates in recent years. Three hESC lines were successfully derived on the vitronectin with a truncated N-terminus (VTN-N) in combination with E-cadherin in xeno-free conditions for the first time, and their undifferentiated state, hESC morphology, and standard karyotypes together with their potential to differentiate into three germ layers were confirmed. These results support the conclusion that the VTN-N/E-cadherin is a suitable substrate for the xeno-free derivation of hESCs and can be used for the derivation of hESCs according to good manufacturing practices.
Collapse
Affiliation(s)
- Tereza Souralova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- International Clinical Research Center, Cell and Tissue Engineering Facility, St. Anne's University Hospital, Pekarska 53, 602 00, Brno, Czech Republic
| | - Daniela Hulinova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- International Clinical Research Center, Cell and Tissue Engineering Facility, St. Anne's University Hospital, Pekarska 53, 602 00, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Michal Jeseta
- Department of Gynecology and Obstetrics, Faculty of Medicine, Center of Assisted Reproduction, Masaryk University Brno and University Hospital, Obilni Trh 11, 602 00, Brno, Czech Republic
| | - Pavel Ventruba
- Department of Gynecology and Obstetrics, Faculty of Medicine, Center of Assisted Reproduction, Masaryk University Brno and University Hospital, Obilni Trh 11, 602 00, Brno, Czech Republic
| | - Ales Hampl
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- International Clinical Research Center, Cell and Tissue Regeneration, St. Anne's University Hospital, Pekarska 53, 602 00, Brno, Czech Republic
| | - Irena Koutna
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic.
- International Clinical Research Center, Cell and Tissue Engineering Facility, St. Anne's University Hospital, Pekarska 53, 602 00, Brno, Czech Republic.
| |
Collapse
|
4
|
Hajizadeh-Tafti F, Golzadeh J, Akyash F, Tahajjodi SS, Farashahi-Yazd E, Heidarian-Meimandi H, Aflatoonian B. Xeno-free generation of new Yazd human embryonic stem cell lines (Yazd4-7) as a prior stage toward good manufacturing practice of clinical-grade raw materials from discarded embryos: A lab resources report. Int J Reprod Biomed 2023; 21:619-628. [PMID: 37885973 PMCID: PMC10598466 DOI: 10.18502/ijrm.v21i8.14017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/22/2023] [Accepted: 07/12/2023] [Indexed: 10/28/2023] Open
Abstract
Background Xeno-free generation of human embryonic stem cells (hESCs) is important to prevent potential animal contaminations in culture for advanced cell-based therapeutic applications. Xeno-free production of hESCs is the first step for manufacturing clinical-grade hESC lines. Objective To produce new hESC lines in xeno-free condition. Materials and Methods This lab resources report was conducted at Stem Cell Biology Research Center, Yazd, Iran from 2019-2022. 4 new hESC lines from 11 (10 fresh and 1 frozen) donated surplus discarded human embryos were established. In this study, we report the xeno-free derivation of new Yazd hESC lines (Yazd4-7), without using immunosurgery, by culturing intact zona-free blastocysts obtained from discarded embryos onto the YhFF#8 cells as a feeder layer in a microdrop culture system. The pluripotency gene expression profile of the cell lines was assessed by reverse transcription polymerase chain reaction and the expression of specific surface markers was detected using immunofluorescent staining. In vitro differentiation was induced using embryoid body formation and gene expression profile of 3 germ layers and germ cells. Reverse transcriptase polymerase chain reaction was investigated to prove their pluripotent capacity. Results In sum, we have been able to generate 4 new hESC lines (Yazd4-7) from 11 discarded embryos in xeno-free culture conditions using a micro drop culture system and YhFF#8 as a human source feeder layer. Conclusion The outcome of this work can be the foundation for the future allogeneic cell-based therapeutic application using clinical grade good manufacturing practice-derived hESC derivatives.
Collapse
Affiliation(s)
- Fatemeh Hajizadeh-Tafti
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Jalal Golzadeh
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Akyash
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Reproductive Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Somayyeh-Sadat Tahajjodi
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Reproductive Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ehsan Farashahi-Yazd
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Reproductive Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hassan Heidarian-Meimandi
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Behrouz Aflatoonian
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Reproductive Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
5
|
Duarte AC, Costa EC, Filipe HAL, Saraiva SM, Jacinto T, Miguel SP, Ribeiro MP, Coutinho P. Animal-derived products in science and current alternatives. BIOMATERIALS ADVANCES 2023; 151:213428. [PMID: 37146527 DOI: 10.1016/j.bioadv.2023.213428] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/08/2023] [Accepted: 04/11/2023] [Indexed: 05/07/2023]
Abstract
More than fifty years after the 3Rs definition and despite the continuous implementation of regulatory measures, animals continue to be widely used in basic research. Their use comprises not only in vivo experiments with animal models, but also the production of a variety of supplements and products of animal origin for cell and tissue culture, cell-based assays, and therapeutics. The animal-derived products most used in basic research are fetal bovine serum (FBS), extracellular matrix proteins such as Matrigel™, and antibodies. However, their production raises several ethical issues regarding animal welfare. Additionally, their biological origin is associated with a high risk of contamination, resulting, frequently, in poor scientific data for clinical translation. These issues support the search for new animal-free products able to replace FBS, Matrigel™, and antibodies in basic research. In addition, in silico methodologies play an important role in the reduction of animal use in research by refining the data previously to in vitro and in vivo experiments. In this review, we depicted the current available animal-free alternatives in in vitro research.
Collapse
Affiliation(s)
- Ana C Duarte
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal; CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Elisabete C Costa
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal
| | - Hugo A L Filipe
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal
| | - Sofia M Saraiva
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal
| | - Telma Jacinto
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal
| | - Sónia P Miguel
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal; CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Maximiano P Ribeiro
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal; CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Paula Coutinho
- CPIRN/IPG - Centro de Potencial e Inovação em Recursos Naturais, Instituto Politécnico da Guarda (CPIRN/IPG), 6300-559 Guarda, Portugal; CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal.
| |
Collapse
|
6
|
Zhou P, Qin L, Ge Z, Xie B, Huang H, He F, Ma S, Ren L, Shi J, Pei S, Dong G, Qi Y, Lan F. Design of chemically defined synthetic substrate surfaces for the in vitro maintenance of human pluripotent stem cells: A review. J Biomed Mater Res B Appl Biomater 2022; 110:1968-1990. [PMID: 35226397 DOI: 10.1002/jbm.b.35034] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 11/11/2022]
Abstract
Human pluripotent stem cells (hPSCs) have the potential of long-term self-renewal and differentiation into nearly all cell types in vitro. Prior to the downstream applications, the design of chemically defined synthetic substrates for the large-scale proliferation of quality-controlled hPSCs is critical. Although great achievements have been made, Matrigel and recombinant proteins are still widely used in the fundamental research and clinical applications. Therefore, much effort is still needed to improve the performance of synthetic substrates in the culture of hPSCs, realizing their commercial applications. In this review, we summarized the design of reported synthetic substrates and especially their limitations in terms of cell culture. Moreover, much attention was paid to the development of promising peptide displaying surfaces. Besides, the biophysical regulation of synthetic substrate surfaces as well as the three-dimensional culture systems were described.
Collapse
Affiliation(s)
- Ping Zhou
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Liying Qin
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Zhangjie Ge
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Biyao Xie
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Hongxin Huang
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Fei He
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Shengqin Ma
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Lina Ren
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Jiamin Shi
- Department of Laboratory Animal Centre, Changzhi Medical College, Changzhi, China
| | - Suying Pei
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Genxi Dong
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Yongmei Qi
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Feng Lan
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen Key Laboratory of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Shenzhen, China
| |
Collapse
|
7
|
Wu Y, Chung YY, Chin YT, Lin CY, Kuo PJ, Chen TY, Lin TY, Chiu HC, Huang HM, Jeng JH, Lee SY. Comparison of 2,3,5,4'-tetrahydroxystilbene-2-O-b-D-glucoside-induced proliferation and differentiation of dental pulp stem cells in 2D and 3D culture systems-gene analysis. J Dent Sci 2022; 17:14-29. [PMID: 35028016 PMCID: PMC8740205 DOI: 10.1016/j.jds.2021.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/10/2021] [Indexed: 12/05/2022] Open
Abstract
Background/purpose Culture environments play a critical role in stem cell expansion. This study aimed to evaluate the effects of 2,3,5,4′-tetrahydroxystilbene-2-O-b-D-glucoside (THSG) on the proliferation and differentiation of human dental pulp stem cells (DPSCs) in 2-dimensional (2D) and 3-dimensional (3D) culture systems. Materials and methods Human DPSCs were seeded in T25 flasks for 2D cultivation. For the 3D culture system, DPSCs were mixed with microcarriers and cultured in spinner flasks. Cells in both culture systems were treated with THSG, and cell proliferation was determined using a cell counter and a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide assay. In THSG-treated DPSCs, the genes associated with proliferation, adipogenesis, neurogenesis, osteogenesis, pluripotency, oncogenesis, and apoptosis were analyzed using real-time polymerase chain reactions. Results The spinner flask time-dependently improved cell numbers, cell viability, and expansion rates in THSG-treated DPSCs. In both the T25 and spinner flasks, the messenger RNA (mRNA) levels of proliferation, osteogenesis, and pluripotent-related genes had a significant maximum expression with 10 μM THSG treatment. However, 0.1 μM of THSG may be the most suitable condition for triggering neurogenesis and adipogenesis gene expression when DPSCs were cultured in spinner flasks. Furthermore, the number of oncogenes and apoptotic genes decreased considerably in the presence of THSG in both the T25 and spinner flasks. Conclusion The spinner flask bioreactor combined with THSG may upregulate proliferation and lineage-specific differentiation in DPSCs. Thus, the combination can be used to mass-produce and cultivate human DPSCs for regenerative dentistry.
Collapse
Affiliation(s)
- Yen Wu
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Dentistry, Wan-Fang Medical Center, Taipei Medical University, Taipei, Taiwan.,Center for Tooth Bank and Dental Stem Cell Technology, Taipei Medical University, Taipei, Taiwan
| | - Yao-Yu Chung
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.,Center for Tooth Bank and Dental Stem Cell Technology, Taipei Medical University, Taipei, Taiwan
| | - Yu-Tang Chin
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.,Center for Tooth Bank and Dental Stem Cell Technology, Taipei Medical University, Taipei, Taiwan
| | - Chi-Yu Lin
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.,Center for Tooth Bank and Dental Stem Cell Technology, Taipei Medical University, Taipei, Taiwan
| | - Po-Jan Kuo
- Department of Periodontology, School of Dentistry, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
| | - Ting-Yi Chen
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Dentistry, Wan-Fang Medical Center, Taipei Medical University, Taipei, Taiwan.,Center for Tooth Bank and Dental Stem Cell Technology, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Yu Lin
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Dentistry, Wan-Fang Medical Center, Taipei Medical University, Taipei, Taiwan.,Center for Tooth Bank and Dental Stem Cell Technology, Taipei Medical University, Taipei, Taiwan
| | - Hsien-Chung Chiu
- Department of Periodontology, School of Dentistry, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
| | - Haw-Ming Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jiiang-Huei Jeng
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Dentistry, Kaohsiung Medical, University Hospital, Kaohsiung, Taiwan
| | - Sheng-Yang Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Dentistry, Wan-Fang Medical Center, Taipei Medical University, Taipei, Taiwan.,Center for Tooth Bank and Dental Stem Cell Technology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
8
|
Nath SC, Harper L, Rancourt DE. Cell-Based Therapy Manufacturing in Stirred Suspension Bioreactor: Thoughts for cGMP Compliance. Front Bioeng Biotechnol 2020; 8:599674. [PMID: 33324625 PMCID: PMC7726241 DOI: 10.3389/fbioe.2020.599674] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/30/2020] [Indexed: 12/23/2022] Open
Abstract
Cell-based therapy (CBT) is attracting much attention to treat incurable diseases. In recent years, several clinical trials have been conducted using human pluripotent stem cells (hPSCs), and other potential therapeutic cells. Various private- and government-funded organizations are investing in finding permanent cures for diseases that are difficult or expensive to treat over a lifespan, such as age-related macular degeneration, Parkinson’s disease, or diabetes, etc. Clinical-grade cell manufacturing requiring current good manufacturing practices (cGMP) has therefore become an important issue to make safe and effective CBT products. Current cell production practices are adopted from conventional antibody or protein production in the pharmaceutical industry, wherein cells are used as a vector to produce the desired products. With CBT, however, the “cells are the final products” and sensitive to physico- chemical parameters and storage conditions anywhere between isolation and patient administration. In addition, the manufacturing of cellular products involves multi-stage processing, including cell isolation, genetic modification, PSC derivation, expansion, differentiation, purification, characterization, cryopreservation, etc. Posing a high risk of product contamination, these can be time- and cost- prohibitive due to maintenance of cGMP. The growing demand of CBT needs integrated manufacturing systems that can provide a more simple and cost-effective platform. Here, we discuss the current methods and limitations of CBT, based upon experience with biologics production. We review current cell manufacturing integration, automation and provide an overview of some important considerations and best cGMP practices. Finally, we propose how multi-stage cell processing can be integrated into a single bioreactor, in order to develop streamlined cGMP-compliant cell processing systems.
Collapse
Affiliation(s)
- Suman C Nath
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Lane Harper
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Derrick E Rancourt
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
9
|
rhBMP-2 Pre-Treated Human Periodontal Ligament Stem Cell Sheets Regenerate a Mineralized Layer Mimicking Dental Cementum. Int J Mol Sci 2020; 21:ijms21113767. [PMID: 32466616 PMCID: PMC7312797 DOI: 10.3390/ijms21113767] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/23/2020] [Accepted: 05/25/2020] [Indexed: 01/01/2023] Open
Abstract
The periodontal complex consisting of alveolar bone, cementum, and periodontal ligaments (PDL) supports human teeth through the systematic orchestration of mineralized tissues and fibrous tissues. Importantly, cementum, the outermost mineralized layer of dental roots, plays an essential role by bridging the inner ligaments from the dental root to the alveolar bone. When the periodontal complex is damaged, the regeneration of each component of the periodontal complex is necessary; however, it is still challenging to achieve complete functional regeneration. In this study, we tried to control the regeneration of cementum and PDL by using a human PDL stem cell (hPDLSC) sheet engineering technology with the pretreatment of recombinant human BMP-2 (rhBMP-2). Isolated hPDLSCs obtained from extracted human teeth were pretreated with rhBMP-2 for in vitro osteogenic differentiation and grafted on the micro/macro-porous biphasic calcium phosphate (MBCP) blocks, which represent dental roots. The MBCPs with hPDLSC sheets were implanted in the subcutaneous layer of immune-compromised mice, and rhBMP-2 pretreated hPDLSC sheets showed higher mineralization and collagen ligament deposition than the no-pretreatment group. Therefore, the rhBMP-2-hPDLSC sheet technique could be an effective strategy for the synchronized regeneration of two different tissues: mineralized tissue and fibrous tissues in periodontal complexes.
Collapse
|
10
|
Cherian DS, Bhuvan T, Meagher L, Heng TSP. Biological Considerations in Scaling Up Therapeutic Cell Manufacturing. Front Pharmacol 2020; 11:654. [PMID: 32528277 PMCID: PMC7247829 DOI: 10.3389/fphar.2020.00654] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
Cell therapeutics - using cells as living drugs - have made advances in many areas of medicine. One of the most clinically studied cell-based therapy products is mesenchymal stromal cells (MSCs), which have shown promising results in promoting tissue regeneration and modulating inflammation. However, MSC therapy requires large numbers of cells, the generation of which is not feasible via conventional planar tissue culture methods. Scale-up manufacturing methods (e.g., propagation on microcarriers in stirred-tank bioreactors), however, are not specifically tailored for MSC expansion. These processes may, in principle, alter the cell secretome, a vital component underlying the immunosuppressive properties and clinical effectiveness of MSCs. This review outlines our current understanding of MSC properties and immunomodulatory function, expansion in commercial manufacturing systems, and gaps in our knowledge that need to be addressed for effective up-scaling commercialization of MSC therapy.
Collapse
Affiliation(s)
- Darshana S Cherian
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Tejasvini Bhuvan
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Laurence Meagher
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, Australia
| | - Tracy S P Heng
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
11
|
Polanco A, Kuang B, Yoon S. Bioprocess Technologies that Preserve the Quality of iPSCs. Trends Biotechnol 2020; 38:1128-1140. [PMID: 32941792 DOI: 10.1016/j.tibtech.2020.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/16/2022]
Abstract
Large-scale production of induced pluripotent stem cells (iPSCs) is essential for the treatment of a variety of clinical indications. However, culturing enough iPSCs for clinical applications is problematic due to their sensitive pluripotent state and dependence on a supporting matrix. Developing stem cell bioprocessing strategies that are scalable and meet clinical needs requires incorporating methods that measure and monitor intrinsic markers of cell differentiation state, developmental status, and viability in real time. In addition, proper cell culture modalities that nurture the growth of high-quality stem cells in suspension are critical for industrial scale-up. In this review, we present an overview of cell culture media, suspension modalities, and monitoring techniques that preserve the quality and pluripotency of iPSCs during initiation, expansion, and manufacturing.
Collapse
Affiliation(s)
- Ashli Polanco
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, USA
| | - Bingyu Kuang
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, USA
| | - Seongkyu Yoon
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, USA.
| |
Collapse
|
12
|
Chan SW, Rizwan M, Yim EKF. Emerging Methods for Enhancing Pluripotent Stem Cell Expansion. Front Cell Dev Biol 2020; 8:70. [PMID: 32117992 PMCID: PMC7033584 DOI: 10.3389/fcell.2020.00070] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
Pluripotent stem cells (PSCs) have great potential to revolutionize the fields of tissue engineering and regenerative medicine as well as stem cell therapeutics. However, the end goal of using PSCs for therapeutic use remains distant due to limitations in current PSC production. Conventional methods for PSC expansion have limited potential to be scaled up to produce the number of cells required for the end-goal of therapeutic use due to xenogenic components, high cost or low efficiency. In this mini review, we explore novel methods and emerging technologies of improving PSC expansion: the use of the two-dimensional mechanobiological strategies of topography and stiffness and the use of three-dimensional (3D) expansion methods including encapsulation, microcarrier-based culture, and suspension culture. Additionally, we discuss the limitations of conventional PSC expansion methods as well as the challenges in implementing non-conventional methods.
Collapse
Affiliation(s)
- Sarah W. Chan
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Muhammad Rizwan
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Evelyn K. F. Yim
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON, Canada
- Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, Canada
- Centre for Biotechnology and Bioengineering, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
13
|
Brault J, Vigne B, Stasia MJ. Ex Vivo Models of Chronic Granulomatous Disease. Methods Mol Biol 2020; 1982:587-622. [PMID: 31172497 DOI: 10.1007/978-1-4939-9424-3_35] [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: 02/11/2023]
Abstract
Induced pluripotent stem cells (iPSCs) are pluripotent stem cells that can be established from dedifferentiation of all somatic cell types by epigenetic phenomena. iPSCs can be differentiated into any mature cells like neurons, hepatocytes, or pancreatic cells that have not been easily available to date. Thus, iPSCs are widely used for disease modeling, drug discovery, and cell therapy development. Here, we describe a protocol to obtain human mature and functional neutrophils and macrophages as ex vivo models of X-linked chronic granulomatous disease (X-CGD). This method can be applied to model the other genetic forms of CGD. We also describe methods for testing the characteristics and functions of neutrophils and macrophages by morphology, phagocytosis assay, release of granule markers or cytokines, cell surface markers, and NADPH oxidase activity.
Collapse
Affiliation(s)
- Julie Brault
- Centre Diagnostic et Recherche CGD (CDiReC), Pôle Biologie, CHU Grenoble Alpes, Grenoble, France
| | - Bénédicte Vigne
- Centre Diagnostic et Recherche CGD (CDiReC), Pôle Biologie, CHU Grenoble Alpes, Grenoble, France
| | - Marie José Stasia
- Centre Diagnostic et Recherche CGD (CDiReC), Pôle Biologie, CHU Grenoble Alpes, Grenoble, France. .,Universite Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France.
| |
Collapse
|
14
|
Kim MH, Kino-Oka M. Bioengineering Considerations for a Nurturing Way to Enhance Scalable Expansion of Human Pluripotent Stem Cells. Biotechnol J 2020; 15:e1900314. [PMID: 31904180 DOI: 10.1002/biot.201900314] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/11/2019] [Indexed: 12/13/2022]
Abstract
Understanding how defects in mechanotransduction affect cell-to-cell variability will add to the fundamental knowledge of human pluripotent stem cell (hPSC) culture, and may suggest new approaches for achieving a robust, reproducible, and scalable process that result in consistent product quality and yields. Here, the current state of the understanding of the fundamental mechanisms that govern the growth kinetics of hPSCs between static and dynamic cultures is reviewed, the factors causing fluctuations are identified, and culture strategies that might eliminate or minimize the occurrence of cell-to-cell variability arising from these fluctuations are discussed. The existing challenges in the development of hPSC expansion methods for enabling the transition from process development to large-scale production are addressed, a mandatory step for industrial and clinical applications of hPSCs.
Collapse
Affiliation(s)
- Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
15
|
Li H, Jiang H, Zhang B, Feng J. Modeling Parkinson's Disease Using Patient-specific Induced Pluripotent Stem Cells. JOURNAL OF PARKINSONS DISEASE 2019; 8:479-493. [PMID: 30149462 PMCID: PMC6218140 DOI: 10.3233/jpd-181353] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disorder. It is characterized by the degeneration of nigral dopaminergic (DA) neurons. While over 90% of cases are idiopathic, without a clear etiology, mutations in many genes have been linked to rare, familial forms of PD. It has been quite challenging to develop effective animal models of PD that capture salient features of PD. The discovery of induced pluripotent stem cells (iPSCs) makes it possible to generate patient-specific DA neurons to study PD. Here, we review the methods for the generation of iPSCs and discuss previous studies using iPSC-derived neurons from monogenic forms of PD. These investigations have revealed several converging pathways that intersect with the unique vulnerabilities of human nigral DA neurons. With the rapid development in stem cell biology, it is possible to generate patient-specific neurons that will be increasingly similar to those in the brain of the patient. Combined with the ability to edit the genome to generate isogenic iPSCs, the generation and analysis of patient-specific midbrain DA neurons will transform PD research by providing a valuable tool for mechanistic study and drug discovery.
Collapse
Affiliation(s)
- Hong Li
- Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA.,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Houbo Jiang
- Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA.,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Boyang Zhang
- Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA.,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Jian Feng
- Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA.,Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
16
|
Engineered biomaterials to mitigate growth factor cost in cell biomanufacturing. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2019. [DOI: 10.1016/j.cobme.2018.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
17
|
Hill CJ, Fleming JR, Mousavinejad M, Nicholson R, Tzokov SB, Bullough PA, Bogomolovas J, Morgan MR, Mayans O, Murray P. Self-Assembling Proteins as High-Performance Substrates for Embryonic Stem Cell Self-Renewal. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1807521. [PMID: 30866118 DOI: 10.1002/adma.201807521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/28/2019] [Indexed: 06/09/2023]
Abstract
The development of extracellular matrix mimetics that imitate niche stem cell microenvironments and support cell growth for technological applications is intensely pursued. Specifically, mimetics are sought that can enact control over the self-renewal and directed differentiation of human pluripotent stem cells (hPSCs) for clinical use. Despite considerable progress in the field, a major impediment to the clinical translation of hPSCs is the difficulty and high cost of large-scale cell production under xeno-free culture conditions using current matrices. Here, a bioactive, recombinant, protein-based polymer, termed ZTFn , is presented that closely mimics human plasma fibronectin and serves as an economical, xeno-free, biodegradable, and functionally adaptable cell substrate. The ZTFn substrate supports with high performance the propagation and long-term self-renewal of human embryonic stem cells while preserving their pluripotency. The ZTFn polymer can, therefore, be proposed as an efficient and affordable replacement for fibronectin in clinical grade cell culturing. Further, it can be postulated that the ZT polymer has significant engineering potential for further orthogonal functionalization in complex cell applications.
Collapse
Affiliation(s)
- Christopher J Hill
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Nuffield Building, Crown Street, Liverpool, L69 3BX, UK
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
| | | | - Masoumeh Mousavinejad
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Nuffield Building, Crown Street, Liverpool, L69 3BX, UK
| | - Rachael Nicholson
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Nuffield Building, Crown Street, Liverpool, L69 3BX, UK
| | - Svetomir B Tzokov
- Department of Molecular Biology and Biotechnology, The Krebs Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Per A Bullough
- Department of Molecular Biology and Biotechnology, The Krebs Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Julius Bogomolovas
- Department of Medicine, UCSD, La Jolla, CA, 92093, USA
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany
| | - Mark R Morgan
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Nuffield Building, Crown Street, Liverpool, L69 3BX, UK
| | - Olga Mayans
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
- Department of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Patricia Murray
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Nuffield Building, Crown Street, Liverpool, L69 3BX, UK
| |
Collapse
|
18
|
Torizal FG, Horiguchi I, Sakai Y. Physiological Microenvironmental Conditions in Different Scalable Culture Systems for Pluripotent Stem Cell Expansion and Differentiation. Open Biomed Eng J 2019. [DOI: 10.2174/1874120701913010041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Human Pluripotent Stem Cells (PSCs) are a valuable cell type that has a wide range of biomedical applications because they can differentiate into many types of adult somatic cell. Numerous studies have examined the clinical applications of PSCs. However, several factors such as bioreactor design, mechanical stress, and the physiological environment have not been optimized. These factors can significantly alter the pluripotency and proliferation properties of the cells, which are important for the mass production of PSCs. Nutritional mass transfer and oxygen transfer must be effectively maintained to obtain a high yield. Various culture systems are currently available for optimum cell propagation by maintaining the physiological conditions necessary for cell cultivation. Each type of culture system using a different configuration with various advantages and disadvantages affecting the mechanical conditions in the bioreactor, such as shear stress. These factors make it difficult to preserve the cellular viability and pluripotency of PSCs. Additional limitations of the culture system for PSCs must also be identified and overcome to maintain the culture conditions and enable large-scale expansion and differentiation of PSCs. This review describes the different physiological conditions in the various culture systems and recent developments in culture technology for PSC expansion and differentiation.
Collapse
|
19
|
Dakhore S, Nayer B, Hasegawa K. Human Pluripotent Stem Cell Culture: Current Status, Challenges, and Advancement. Stem Cells Int 2018; 2018:7396905. [PMID: 30595701 PMCID: PMC6282144 DOI: 10.1155/2018/7396905] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/23/2022] Open
Abstract
Over the past two decades, human embryonic stem cells (hESCs) have gained attention due to their pluripotent and proliferative ability which enables production of almost all cell types in the human body in vitro and makes them an excellent tool to study human embryogenesis and disease, as well as for drug discovery and cell transplantation therapies. Discovery of human-induced pluripotent stem cells (hiPSCs) further expanded therapeutic applications of human pluripotent stem cells (PSCs). hPSCs provide a stable and unlimited original cell source for producing suitable cells and tissues for downstream applications. Therefore, engineering the environment in which these cells are grown, for stable and quality-controlled hPSC maintenance and production, is one of the key factors governing the success of these applications. hPSCs are maintained in a particular niche using specific cell culture components. Ideally, the culture should be free of xenobiotic components to render hPSCs suitable for therapeutic applications. Substantial efforts have been put to identify effective components, and develop culture conditions and protocols, for their large-scale expansion without compromising on quality. In this review, we discuss different media, their components and functions, including specific requirements to maintain the pluripotent and proliferative ability of hPSCs. Understanding the role of culture components would enable the development of appropriate conditions to promote large-scale, quality-controlled expansion of hPSCs thereby increasing their potential applications.
Collapse
Affiliation(s)
- Sushrut Dakhore
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences (NCBS), Bangalore, India
| | - Bhavana Nayer
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences (NCBS), Bangalore, India
| | - Kouichi Hasegawa
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences (NCBS), Bangalore, India
- Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advanced Study, Kyoto University, Japan
| |
Collapse
|
20
|
Hair Follicle Dermal Cells Support Expansion of Murine and Human Embryonic and Induced Pluripotent Stem Cells and Promote Haematopoiesis in Mouse Cultures. Stem Cells Int 2018; 2018:8631432. [PMID: 30154866 PMCID: PMC6098861 DOI: 10.1155/2018/8631432] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 04/26/2018] [Indexed: 12/26/2022] Open
Abstract
In the hair follicle, the dermal papilla (DP) and dermal sheath (DS) support and maintain proliferation and differentiation of the epithelial stem cells that produce the hair fibre. In view of their regulatory properties, in this study, we investigated the interaction between hair follicle dermal cells (DP and DS) and embryonic stem cells (ESCs); induced pluripotent stem cells (iPSCs); and haematopoietic stem cells. We found that coculture of follicular dermal cells with ESCs or iPSCs supported their prolonged maintenance in an apparently undifferentiated state as established by differentiation assays, immunocytochemistry, and RT-PCR for markers of undifferentiated ESCs. We further showed that cytokines that are involved in ESC support are also expressed by cultured follicle dermal cells, providing a possible explanation for maintenance of ES cell stemness in cocultures. The same cytokines were expressed within follicles in situ in a pattern more consistent with a role in follicle growth activities than stem cell maintenance. Finally, we show that cultured mouse follicle dermal cells provide good stromal support for haematopoiesis in an established coculture model. Human follicular dermal cells represent an accessible and readily propagated source of feeder cells for pluripotent and haematopoietic cells and have potential for use in clinical applications.
Collapse
|
21
|
Laminin 521 Stabilizes the Pluripotency Expression Pattern of Human Embryonic Stem Cells Initially Derived on Feeder Cells. Stem Cells Int 2018. [PMID: 29535778 PMCID: PMC5835285 DOI: 10.1155/2018/7127042] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Human embryonic stem (hES) cells represent an important tool to study early cell development. The previously described use of human recombinant laminin (LN) 521 represented a step forward in generating clinically safe culture conditions. To test the short-term effect of LN521 on cultured hES cells, five male hES cell lines were cultured on human foreskin fibroblasts (hFFs), Matrigel, LN521, and LN121 and characterized by qPCR, immunofluorescence analysis, as well as their potential for three-germ layer differentiation. Variations in gene expression related to pluripotency, stemness, and testicular cells at different passages and culture conditions were evaluated by qPCR. All cell lines expressed pluripotency markers at protein and RNA level and were able to differentiate into cell types of the three germ layers after being cultured on LN521 for nine passages. Reduction in variation of pluripotency marker expression could be observed after culturing the cells on LN521 for nine passages. hES cells cultured on LN521 exhibited less differentiation, faster cell growth, and attachment when compared to hES cells cultured on LN121 or Matrigel. Our results indicate a positive effect of LN521 in stabilizing pluripotency gene expression and might be the first step towards more controllable and robust culture conditions for hES cells.
Collapse
|
22
|
Kim MH, Kino-oka M. Bioprocessing Strategies for Pluripotent Stem Cells Based on Waddington’s Epigenetic Landscape. Trends Biotechnol 2018; 36:89-104. [DOI: 10.1016/j.tibtech.2017.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 10/02/2017] [Accepted: 10/10/2017] [Indexed: 12/12/2022]
|
23
|
Suchorska WM, Augustyniak E, Richter M, Trzeciak T. Comparison of Four Protocols to Generate Chondrocyte-Like Cells from Human Induced Pluripotent Stem Cells (hiPSCs). Stem Cell Rev Rep 2017; 13:299-308. [PMID: 27987073 PMCID: PMC5380716 DOI: 10.1007/s12015-016-9708-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Stem cells (SCs) are a promising approach to regenerative medicine, with the potential to treat numerous orthopedic disorders, including osteo-degenerative diseases. The development of human-induced pluripotent stem cells (hiPSCs) has increased the potential of SCs for new treatments. However, current methods of differentiating hiPSCs into chondrocyte-like cells are suboptimal and better methods are needed. The aim of the present study was to assess four different chondrogenic differentiation protocols to identify the most efficient method of generating hiPSC-derived chondrocytes. For this study, hiPSCs were obtained from primary human dermal fibroblasts (PHDFs) and differentiated into chondrocyte-like cells using four different protocols: 1) monolayer culture with defined growth factors (GF); 2) embryoid bodies (EBs) in a chondrogenic medium with TGF-β3 cells; 3) EBs in chondrogenic medium conditioned with human chondrocytes (HC-402-05a cell line) and 4) EBs in chondrogenic medium conditioned with human chondrocytes and supplemented with TGF-β3. The cells obtained through these four protocols were evaluated and compared at the mRNA and protein levels. Although chondrogenic differentiation of hiPSCs was successfully achieved with all of these protocols, the two fastest and most cost-effective methods were the monolayer culture with GFs and the medium conditioned with human chondrocytes. Both of these methods are superior to other available techniques. The main advantage of the conditioned medium is that the technique is relatively simple and inexpensive while the directed method (i.e., monolayer culture with GFs) is faster than any protocol described to date because it is does not require additional steps such as EB formation.
Collapse
Affiliation(s)
- Wiktoria Maria Suchorska
- Radiobiology Lab, Greater Poland Cancer Centre, 61- 866, Poznan, Poland
- Department of Electroradiology, Poznan University of Medical Sciences, 61-866, Poznan, Poland
| | - Ewelina Augustyniak
- Radiobiology Lab, Greater Poland Cancer Centre, 61- 866, Poznan, Poland.
- The Postgraduate School of Molecular Medicine, Medical University of Warsaw, 02-091, Warsaw, Poland.
| | - Magdalena Richter
- Department of Orthopedics and Traumatology, Poznan University of Medical Sciences, 61-545, Poznan, Poland
| | - Tomasz Trzeciak
- Department of Orthopedics and Traumatology, Poznan University of Medical Sciences, 61-545, Poznan, Poland
| |
Collapse
|
24
|
Kim MH, Matsubara Y, Fujinaga Y, Kino-Oka M. A Simple and Robust Method for Culturing Human-Induced Pluripotent Stem Cells in an Undifferentiated State Using Botulinum Hemagglutinin. Biotechnol J 2017; 13. [PMID: 29027750 DOI: 10.1002/biot.201700384] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/24/2017] [Indexed: 01/18/2023]
Abstract
Clinical and industrial applications of human-induced pluripotent stem cells (hiPSCs) is hindered by the lack of robust culture strategies capable of sustaining a culture in an undifferentiated state. Here, a simple and robust hiPSC-culture-propagation strategy incorporating botulinum hemagglutinin (HA)-mediated selective removal of cells deviating from an undifferentiated state is developed. After HA treatment, cell-cell adhesion is disrupted, and deviated cells detached from the central region of the colony to subsequently form tight monolayer colonies following prolonged incubation. The authors find that the temporal and dose-dependent activity of HA regulated deviated-cell removal and recoverability after disruption of cell-cell adhesion in hiPSC colonies. The effects of HA are confirmed under all culture conditions examined, regardless of hiPSC line and feeder-dependent or -free culture conditions. After routine application of our HA-treatment paradigm for serial passages, hiPSCs maintains expression of pluripotent markers and readily forms embryoid bodies expressing markers for all three germ-cell layers. This method enables highly efficient culturing of hiPSCs and use of entire undifferentiated portions without having to pick deviated cells manually. This simple and readily reproducible culture strategy is a potentially useful tool for improving the robust and scalable maintenance of undifferentiated hiPSC cultures.
Collapse
Affiliation(s)
- Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshifumi Matsubara
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yukako Fujinaga
- Laboratory for Infection Cell Biology, International Research Centre for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Bacteriology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara, Kanazawa, Ishikawa, 920-8640, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
25
|
Kropp C, Massai D, Zweigerdt R. Progress and challenges in large-scale expansion of human pluripotent stem cells. Process Biochem 2017. [DOI: 10.1016/j.procbio.2016.09.032] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
26
|
Dias AD, Elicson JM, Murphy WL. Microcarriers with Synthetic Hydrogel Surfaces for Stem Cell Expansion. Adv Healthc Mater 2017; 6:10.1002/adhm.201700072. [PMID: 28509413 PMCID: PMC5607626 DOI: 10.1002/adhm.201700072] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/09/2017] [Indexed: 12/20/2022]
Abstract
Microcarriers are scalable support surfaces for cell growth that enable high levels of expansion, and are particularly relevant for expansion of human mesenchymal stem cells (hMSCs). The goal of this study is to develop a poly(ethylene glycol) (PEG)-based microcarrier coating for hMSC expansion. Commercially available microcarriers do not offer customizability of microcarrier surface properties, including elastic modulus and surface cell adhesion ligands. The lab has previously demonstrated that tuning these material properties on PEG-based hydrogels can modulate important cellular growth characteristics, such as cell attachment and expansion, which are important in microcarrier-based culture. Eosin-Y is adsorbed to polystyrene microcarriers and used as a photoinitiator for thiol-ene polymerization under visible light. Resultant PEG coatings are over 100 µm thick and localized to microcarrier surfaces. This thickness is relevant for cells to react to mechanical properties of the hydrogel coating, and coated microcarriers support hMSC attachment and expansion. hMSC expansion is highly favorable on coated microcarriers in serum-free media, with doubling times under 25 h in the growth phase, and retained osteogenic and adipogenic differentiation capacity after culture on microcarriers. These microcarriers with defined, synthetic coatings enable tailorable surfaces for cell expansion that may be suitable for a variety of biomanufacturing applications.
Collapse
Affiliation(s)
- Andrew D Dias
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Ave., WIMR 5418, Madison, WI, 53705, USA
| | - Jonathan M Elicson
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Ave., WIMR 5418, Madison, WI, 53705, USA
| | - William L Murphy
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Ave., WIMR 5418, Madison, WI, 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Ave., WIMR 5418, Madison, WI, 53705, USA
- Department of Material Science and Engineering, University of Wisconsin-Madison, 1111 Highland Ave., WIMR 5418, Madison, WI, 53705, USA
| |
Collapse
|
27
|
Leino M, Astrand C, Hughes-Brittain N, Robb B, McKean R, Chotteau V. Human embryonic stem cell dispersion in electrospun PCL fiber scaffolds by coating with laminin-521 and E-cadherin-Fc. J Biomed Mater Res B Appl Biomater 2017; 106:1226-1236. [PMID: 28577328 DOI: 10.1002/jbm.b.33928] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 04/21/2017] [Accepted: 05/12/2017] [Indexed: 12/14/2022]
Abstract
Advances in human pluripotent cell cultivation and differentiation protocols have led to production of stem cell-derived progenitors as a promising cell source for replacement therapy. Three-dimensional (3-D) culture is a better mimic of the natural niche for stem cells and is widely used for disease modeling. Here, we describe a nonaggregate culture system of human embryonic stem cells inside electrospun polycaprolactone (PCL) fiber scaffolds combined with defined extracellular proteins naturally occurring in the stem cell niche. PCL fiber scaffolds coated with recombinant human laminin-521 readily supported initial stem cell attachment and growth from a single-cell suspension. The combination of recombinant E-cadherin-Fc and laminin-521 further improved cell dispersion rendering a uniform cell population. Finally, we showed that the cells cultured in E-cadherin-Fc- and laminin-521-coated PCL scaffolds could differentiate into all three germ layers. Importantly, we provided a chemically defined 3-D system in which pluripotent stem cells grown and differentiated avoiding the formation of cell aggregates. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1226-1236, 2018.
Collapse
Affiliation(s)
- Mattias Leino
- School of Biotechnology, Cell Technology Group (CETEG), KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Carolina Astrand
- School of Biotechnology, Cell Technology Group (CETEG), KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Nanayaa Hughes-Brittain
- The Electrospinning Company Ltd, R70 Rutherford Appleton Laboratory, Harwell Oxford, Didcot, Oxfordshire, OX11 0QX, UK
| | - Brendan Robb
- The Electrospinning Company Ltd, R70 Rutherford Appleton Laboratory, Harwell Oxford, Didcot, Oxfordshire, OX11 0QX, UK
| | - Robert McKean
- The Electrospinning Company Ltd, R70 Rutherford Appleton Laboratory, Harwell Oxford, Didcot, Oxfordshire, OX11 0QX, UK
| | - Véronique Chotteau
- School of Biotechnology, Cell Technology Group (CETEG), KTH - Royal Institute of Technology, Stockholm, Sweden.,AdBIOPRO, Competence Centre for Advanced Bioproduction by Continuous Bioprocessing, KTH, Stockholm, Sweden
| |
Collapse
|
28
|
Microcarrier-based platforms for in vitro expansion and differentiation of human pluripotent stem cells in bioreactor culture systems. J Biotechnol 2016; 234:71-82. [PMID: 27480342 DOI: 10.1016/j.jbiotec.2016.07.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 07/26/2016] [Accepted: 07/28/2016] [Indexed: 01/15/2023]
Abstract
Human pluripotent stem cells (hPSC) have attracted a great attention as an unlimited source of cells for cell therapies and other in vitro biomedical applications such as drug screening, toxicology assays and disease modeling. The implementation of scalable culture platforms for the large-scale production of hPSC and their derivatives is mandatory to fulfill the requirement of obtaining large numbers of cells for these applications. Microcarrier technology has been emerging as an effective approach for the large scale ex vivo hPSC expansion and differentiation. This review presents recent achievements in hPSC microcarrier-based culture systems and discusses the crucial aspects that influence the performance of these culture platforms. Recent progress includes addressing chemically-defined culture conditions for manufacturing of hPSC and their derivatives, with the development of xeno-free media and microcarrier coatings to meet good manufacturing practice (GMP) quality requirements. Finally, examples of integrated platforms including hPSC expansion and directed differentiation to specific lineages are also presented in this review.
Collapse
|
29
|
Badenes SM, Fernandes TG, Cordeiro CSM, Boucher S, Kuninger D, Vemuri MC, Diogo MM, Cabral JMS. Defined Essential 8™ Medium and Vitronectin Efficiently Support Scalable Xeno-Free Expansion of Human Induced Pluripotent Stem Cells in Stirred Microcarrier Culture Systems. PLoS One 2016; 11:e0151264. [PMID: 26999816 PMCID: PMC4801338 DOI: 10.1371/journal.pone.0151264] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 02/19/2016] [Indexed: 12/24/2022] Open
Abstract
Human induced pluripotent stem (hiPS) cell culture using Essential 8™ xeno-free medium and the defined xeno-free matrix vitronectin was successfully implemented under adherent conditions. This matrix was able to support hiPS cell expansion either in coated plates or on polystyrene-coated microcarriers, while maintaining hiPS cell functionality and pluripotency. Importantly, scale-up of the microcarrier-based system was accomplished using a 50 mL spinner flask, under dynamic conditions. A three-level factorial design experiment was performed to identify optimal conditions in terms of a) initial cell density b) agitation speed, and c) to maximize cell yield in spinner flask cultures. A maximum cell yield of 3.5 is achieved by inoculating 55,000 cells/cm2 of microcarrier surface area and using 44 rpm, which generates a cell density of 1.4x106 cells/mL after 10 days of culture. After dynamic culture, hiPS cells maintained their typical morphology upon re-plating, exhibited pluripotency-associated marker expression as well as tri-lineage differentiation capability, which was verified by inducing their spontaneous differentiation through embryoid body formation, and subsequent downstream differentiation to specific lineages such as neural and cardiac fates was successfully accomplished. In conclusion, a scalable, robust and cost-effective xeno-free culture system was successfully developed and implemented for the scale-up production of hiPS cells.
Collapse
Affiliation(s)
- Sara M. Badenes
- Department of Bioengineering, and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Tiago G. Fernandes
- Department of Bioengineering, and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- * E-mail:
| | - Cláudia S. M. Cordeiro
- Department of Bioengineering, and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Shayne Boucher
- Thermo Fisher Scientific, Cell Biology, Life Sciences Solutions, Frederick, Maryland, United States of America
| | - David Kuninger
- Thermo Fisher Scientific, Cell Biology, Life Sciences Solutions, Frederick, Maryland, United States of America
| | - Mohan C. Vemuri
- Thermo Fisher Scientific, Cell Biology, Life Sciences Solutions, Frederick, Maryland, United States of America
| | - Maria Margarida Diogo
- Thermo Fisher Scientific, Cell Biology, Life Sciences Solutions, Frederick, Maryland, United States of America
| | - Joaquim M. S. Cabral
- Thermo Fisher Scientific, Cell Biology, Life Sciences Solutions, Frederick, Maryland, United States of America
| |
Collapse
|
30
|
Gelinsky M, Bernhardt A, Milan F. Bioreactors in tissue engineering: Advances in stem cell culture and three-dimensional tissue constructs. Eng Life Sci 2015. [DOI: 10.1002/elsc.201400216] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Michael Gelinsky
- Centre for Translational Bone; Joint and Soft Tissue Research; Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
| | - Anne Bernhardt
- Centre for Translational Bone; Joint and Soft Tissue Research; Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
| | - Falk Milan
- Centre for Translational Bone; Joint and Soft Tissue Research; Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
| |
Collapse
|
31
|
Lam ATL, Li J, Chen AKL, Birch WR, Reuveny S, Oh SKW. Improved Human Pluripotent Stem Cell Attachment and Spreading on Xeno-Free Laminin-521-Coated Microcarriers Results in Efficient Growth in Agitated Cultures. Biores Open Access 2015; 4:242-57. [PMID: 26309800 PMCID: PMC4540119 DOI: 10.1089/biores.2015.0010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Human pluripotent stem cells (hPSC) are self-renewing cells having the potential of differentiation into the three lineages of somatic cells and thus can be medically used in diverse cellular therapies. One of the requirements for achieving these clinical applications is development of completely defined xeno-free systems for large-scale cell expansion and differentiation. Previously, we demonstrated that microcarriers (MCs) coated with mouse laminin-111 (LN111) and positively charged poly-l-lysine (PLL) critically enable the formation and evolution of cells/MC aggregates with high cell yields obtained under agitated conditions. In this article, we further improved the MC system into a defined xeno-free MC one in which the MCs are coated with recombinant human laminin-521 (LN521) alone without additional positive charge. The high binding affinity of the LN521 to cell integrins enables efficient initial HES-3 cell attachment (87%) and spreading (85%), which leads to generation of cells/MC aggregates (400 μm in size) and high cell yields (2.4–3.5×106 cells/mL) within 7 days in agitated plate and scalable spinner cultures. The universality of the system was demonstrated by propagation of an induced pluripotent cells line in this defined MC system. Long-term pluripotent (>90% expression Tra-1-60) cell expansion and maintenance of normal karyotype was demonstrated after 10 cell passages. Moreover, tri-lineage differentiation as well as directed differentiation into cardiomyocytes was achieved. The new LN521-based MC system offers a defined, xeno-free, GMP-compatible, and scalable bioprocessing platform for the production of hPSC with the quantity and quality compliant for clinical applications. Use of LN521 on MCs enabled a 34% savings in matrix and media costs over monolayer cultures to produce 108 cells.
Collapse
Affiliation(s)
- Alan Tin-Lun Lam
- Stem Cell Group, Bioprocessing Technology Institute , Agency for Science, Technology and Research (ASTAR), Singapore , Singapore
| | - Jian Li
- Institute of Materials Research and Engineering , Agency for Science, Technology and Research (ASTAR), Singapore , Singapore
| | - Allen Kuan-Liang Chen
- Stem Cell Group, Bioprocessing Technology Institute , Agency for Science, Technology and Research (ASTAR), Singapore , Singapore
| | - William R Birch
- Institute of Materials Research and Engineering , Agency for Science, Technology and Research (ASTAR), Singapore , Singapore
| | - Shaul Reuveny
- Stem Cell Group, Bioprocessing Technology Institute , Agency for Science, Technology and Research (ASTAR), Singapore , Singapore
| | - Steve Kah-Weng Oh
- Stem Cell Group, Bioprocessing Technology Institute , Agency for Science, Technology and Research (ASTAR), Singapore , Singapore
| |
Collapse
|