51
|
Cell fiber-based three-dimensional culture system for highly efficient expansion of human induced pluripotent stem cells. Sci Rep 2017; 7:2850. [PMID: 28588295 PMCID: PMC5460280 DOI: 10.1038/s41598-017-03246-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/21/2017] [Indexed: 12/12/2022] Open
Abstract
Human pluripotent stem cells are a potentially powerful cellular resource for application in regenerative medicine. Because such applications require large numbers of human pluripotent stem cell-derived cells, a scalable culture system of human pluripotent stem cell needs to be developed. Several suspension culture systems for human pluripotent stem cell expansion exist; however, it is difficult to control the thickness of cell aggregations in these systems, leading to increased cell death likely caused by limited diffusion of gases and nutrients into the aggregations. Here, we describe a scalable culture system using the cell fiber technology for the expansion of human induced pluripotent stem (iPS) cells. The cells were encapsulated and cultured within the core region of core-shell hydrogel microfibers, resulting in the formation of rod-shaped or fiber-shaped cell aggregations with sustained thickness and high viability. By encapsulating the cells with type I collagen, we demonstrated a long-term culture of the cells by serial passaging at a high expansion rate (14-fold in four days) while retaining its pluripotency. Therefore, our culture system could be used for large-scale expansion of human pluripotent stem cells for use in regenerative medicine.
Collapse
|
52
|
Huang H, Yu Y, Hu Y, He X, Usta OB, Yarmush ML. Generation and manipulation of hydrogel microcapsules by droplet-based microfluidics for mammalian cell culture. LAB ON A CHIP 2017; 17:1913-1932. [PMID: 28509918 PMCID: PMC5548188 DOI: 10.1039/c7lc00262a] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Hydrogel microcapsules provide miniaturized and biocompatible niches for three-dimensional (3D) in vitro cell culture. They can be easily generated by droplet-based microfluidics with tunable size, morphology, and biochemical properties. Therefore, microfluidic generation and manipulation of cell-laden microcapsules can be used for 3D cell culture to mimic the in vivo environment towards applications in tissue engineering and high throughput drug screening. In this review of recent advances mainly since 2010, we will first introduce general characteristics of droplet-based microfluidic devices for cell encapsulation with an emphasis on the fluid dynamics of droplet breakup and internal mixing as they directly influence microcapsule's size and structure. We will then discuss two on-chip manipulation strategies: sorting and extraction from oil into aqueous phase, which can be integrated into droplet-based microfluidics and significantly improve the qualities of cell-laden hydrogel microcapsules. Finally, we will review various applications of hydrogel microencapsulation for 3D in vitro culture on cell growth and proliferation, stem cell differentiation, tissue development, and co-culture of different types of cells.
Collapse
Affiliation(s)
- Haishui Huang
- Center for Engineering in Medicine, Massachusetts General Hospital,
Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts
02114, United States
| | - Yin Yu
- Center for Engineering in Medicine, Massachusetts General Hospital,
Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts
02114, United States
| | - Yong Hu
- Center for Engineering in Medicine, Massachusetts General Hospital,
Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts
02114, United States
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University,
Columbus, USA
| | - O. Berk Usta
- Center for Engineering in Medicine, Massachusetts General Hospital,
Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts
02114, United States
| | - Martin L. Yarmush
- Center for Engineering in Medicine, Massachusetts General Hospital,
Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts
02114, United States
- Department of Biomedical Engineering, Rutgers University,
Piscataway, New Jersey 08854, United States
| |
Collapse
|
53
|
Lalwani G, D'agati M, Gopalan A, Patel SC, Talukdar Y, Sitharaman B. Three-dimensional carbon nanotube scaffolds for long-term maintenance and expansion of human mesenchymal stem cells. J Biomed Mater Res A 2017; 105:1927-1939. [DOI: 10.1002/jbm.a.36062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/02/2017] [Accepted: 03/07/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Gaurav Lalwani
- Department of Biomedical Engineering; Stony Brook University; Stony Brook New York 11794-5281
| | - Michael D'agati
- Department of Biomedical Engineering; Stony Brook University; Stony Brook New York 11794-5281
| | - Anu Gopalan
- Department of Biomedical Engineering; Stony Brook University; Stony Brook New York 11794-5281
| | - Sunny C. Patel
- Department of Biomedical Engineering; Stony Brook University; Stony Brook New York 11794-5281
| | - Yahfi Talukdar
- Department of Biomedical Engineering; Stony Brook University; Stony Brook New York 11794-5281
| | - Balaji Sitharaman
- Department of Biomedical Engineering; Stony Brook University; Stony Brook New York 11794-5281
| |
Collapse
|
54
|
Alginate: A Versatile Biomaterial to Encapsulate Isolated Ovarian Follicles. Ann Biomed Eng 2017; 45:1633-1649. [DOI: 10.1007/s10439-017-1816-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 02/22/2017] [Indexed: 12/19/2022]
|
55
|
Liu L, Kamei KI, Yoshioka M, Nakajima M, Li J, Fujimoto N, Terada S, Tokunaga Y, Koyama Y, Sato H, Hasegawa K, Nakatsuji N, Chen Y. Nano-on-micro fibrous extracellular matrices for scalable expansion of human ES/iPS cells. Biomaterials 2017; 124:47-54. [PMID: 28187394 DOI: 10.1016/j.biomaterials.2017.01.039] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 01/06/2017] [Accepted: 01/28/2017] [Indexed: 01/22/2023]
Abstract
Human pluripotent stem cells (hPSCs) hold great potential for industrial and clinical applications. Clinical-grade scaffolds and high-quality hPSCs are required for cell expansion as well as easy handling and manipulation of the products. Current hPSC culture methods do not fulfill these requirements because of a lack of proper extracellular matrices (ECMs) and cell culture wares. We developed a layered nano-on-micro fibrous cellular matrix mimicking ECM, named "fiber-on-fiber (FF)" matrix, which enables easy handling and manipulation of cultured cells. While non-woven sheets of cellulose and polyglycolic acid were used as a microfiber layer facilitating mechanical stability, electrospun gelatin nanofibers were crosslinked on the microfiber layer, generating a mesh structure with connected nanofibers facilitating cell adhesion and growth. Our results showed that the FF matrix supports effective hPSC culture with maintenance of their pluripotency and normal chromosomes over two months, as well as effective scaled-up expansion, with fold increases of 54.1 ± 15.6 and 40.4 ± 8.4 in cell number per week for H1 human embryonic stem cells and 253G1 human induced pluripotent stem cells, respectively. This simple approach to mimick the ECM may have important implications after further optimization to generate lineage-specific products.
Collapse
Affiliation(s)
- Li Liu
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan
| | - Ken-Ichiro Kamei
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan.
| | - Momoko Yoshioka
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan
| | - Minako Nakajima
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan
| | - Junjun Li
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan
| | - Nanae Fujimoto
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan; Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyotodaigaku-katsura, Nishikyo-ku, Kyoto, 615-8540, Japan
| | - Shiho Terada
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan
| | - Yumie Tokunaga
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan
| | - Yoshie Koyama
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan
| | - Hideki Sato
- QOL Research Center, Gunze Limited, Kyoto, 623-8512 Japan
| | - Kouichi Hasegawa
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan; Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences (NCBS), Bangalore, 560065, India
| | - Norio Nakatsuji
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan; Institute for Frontier Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Yong Chen
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan; Ecole Normale Supérieure, CNRS-ENS-UPMC UMR 8640, 24 Rue Lhomond, Paris, 75005, France.
| |
Collapse
|
56
|
Cui X, Hartanto Y, Zhang H. Advances in multicellular spheroids formation. J R Soc Interface 2017; 14:20160877. [PMID: 28202590 PMCID: PMC5332573 DOI: 10.1098/rsif.2016.0877] [Citation(s) in RCA: 306] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/11/2017] [Indexed: 12/20/2022] Open
Abstract
Three-dimensional multicellular spheroids (MCSs) have a complex architectural structure, dynamic cell-cell/cell-matrix interactions and bio-mimicking in vivo microenvironment. As a fundamental building block for tissue reconstruction, MCSs have emerged as a powerful tool to narrow down the gap between the in vitro and in vivo model. In this review paper, we discussed the structure and biology of MCSs and detailed fabricating methods. Among these methods, the approach in microfluidics with hydrogel support for MCS formation is promising because it allows essential cell-cell/cell-matrix interactions in a confined space.
Collapse
Affiliation(s)
- X Cui
- School of Chemical Engineering, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Y Hartanto
- School of Chemical Engineering, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - H Zhang
- School of Chemical Engineering, University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
57
|
|
58
|
Abecasis B, Aguiar T, Arnault É, Costa R, Gomes-Alves P, Aspegren A, Serra M, Alves PM. Expansion of 3D human induced pluripotent stem cell aggregates in bioreactors: Bioprocess intensification and scaling-up approaches. J Biotechnol 2017; 246:81-93. [PMID: 28131858 DOI: 10.1016/j.jbiotec.2017.01.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 01/17/2017] [Accepted: 01/24/2017] [Indexed: 12/15/2022]
Abstract
Human induced pluripotent stem cells (hiPSC) are attractive tools for drug screening and disease modeling and promising candidates for cell therapy applications. However, to achieve the high numbers of cells required for these purposes, scalable and clinical-grade technologies must be established. In this study, we use environmentally controlled stirred-tank bioreactors operating in perfusion as a powerful tool for bioprocess intensification of hiPSC production. We demonstrate the importance of controlling the dissolved oxygen concentration at low levels (4%) and perfusion at 1.3day-1 dilution rate to improve hiPSC growth as aggregates in a xeno-free medium. This strategy allowed for increased cell specific growth rate, maximum volumetric concentrations (4.7×106cell/mL) and expansion factors (approximately 19 in total cells), resulting in a 2.6-fold overall improvement in cell yields. Extensive cell characterization, including whole proteomic analysis, was performed to confirm that cells' pluripotent phenotype was maintained during culture. A scalable protocol for continuous expansion of hiPSC aggregates in bioreactors was implemented using mechanical dissociation for aggregate disruption and cell passaging. A total expansion factor of 1100 in viable cells was obtained in 11days of culture, while cells maintained their proliferation capacity, pluripotent phenotype and potential as well as genomic stability after 3 sequential passages in bioreactors.
Collapse
Affiliation(s)
- Bernardo Abecasis
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| | - Tiago Aguiar
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| | - Émilie Arnault
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| | - Rita Costa
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| | - Patricia Gomes-Alves
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| | - Anders Aspegren
- Takara Bio Europe AB, Arvid Wallgrens Backe 20, SE-413 46 Gothenburg, Sweden.
| | - Margarida Serra
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal.
| | - Paula M Alves
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal
| |
Collapse
|
59
|
Koh E, Jung YC, Woo HM, Kang BJ. Injectable alginate-microencapsulated canine adipose tissue-derived mesenchymal stem cells for enhanced viable cell retention. J Vet Med Sci 2017; 79:492-501. [PMID: 28070061 PMCID: PMC5383167 DOI: 10.1292/jvms.16-0456] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The purpose of this study was to establish an optimized protocol for the production of alginate-encapsulated canine adipose-derived mesenchymal stem cells
(cASCs) and evaluate their suitability for clinical use, including viability, proliferation and in vivo cell retention. Alginate microbeads
were formed by vibrational technology and the production of injectable microbeads was performed using various parameters with standard methodology. Microbead
toxicity was tested in an animal model. Encapsulated cASCs were evaluated for viability and proliferation in vitro. HEK-293 cells, with or
without microencapsulation, were injected into the subcutaneous tissue of mice and were tracked using in vivo bioluminescent imaging to
evaluate the retention of transplanted cells. The optimized injectable microbeads were of uniform size and approximately 250 µm in diameter.
There was no strong evidence of in vivo toxicity for the alginate beads. The cells remained viable after encapsulation, and there was evidence
of in vitro proliferation within the microcapsules. In vivo bioluminescent imaging showed that alginate encapsulation improved
the retention of transplanted cells and the encapsulated cells remained viable in vivo for 7 days. Encapsulation enhances the retention of
viable cells in vivo and might represent a potential strategy to increase the therapeutic potency and efficacy of stem cells.
Collapse
Affiliation(s)
- Eunji Koh
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | | | | | | |
Collapse
|
60
|
Abstract
The goal of this chapter is to provide an overview of the different purposes for which the cell microencapsulation technology can be used. These include immunoisolation of non-autologous cells used for cell therapy; immobilization of cells for localized (targeted) delivery of therapeutic products to ablate, repair, or regenerate tissue; simultaneous delivery of multiple therapeutic agents in cell therapy; spatial compartmentalization of cells in complex tissue engineering; expansion of cells in culture; and production of different probiotics and metabolites for industrial applications. For each of these applications, specific examples are provided to illustrate how the microencapsulation technology can be utilized to achieve the purpose. However, successful use of the cell microencapsulation technology for whatever purpose will ultimately depend upon careful consideration for the choice of the encapsulating polymers, the method of fabrication (cross-linking) of the microbeads, which affects the permselectivity, the biocompatibility and the mechanical strength of the microbeads as well as environmental parameters such as temperature, humidity, osmotic pressure, and storage solutions.The various applications discussed in this chapter are illustrated in the different chapters of this book and where appropriate relevant images of the microencapsulation products are provided. It is hoped that this outline of the different applications of cell microencapsulation would provide a good platform for tissue engineers, scientists, and clinicians to design novel tissue constructs and products for therapeutic and industrial applications.
Collapse
Affiliation(s)
- Emmanuel C Opara
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA. .,Virginia Tech-Wake Forest School of Biomedical Engineering & Sciences (SBES), Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
61
|
Bhowmick A, Pramanik N, Mitra T, Gnanamani A, Das M, Kundu PP. Fabrication of porous magnetic nanocomposites for bone tissue engineering. NEW J CHEM 2017. [DOI: 10.1039/c6nj03358j] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Porous superparamagnetic chitosan/polyethylene glycol/hydroxyapatite–Fe3O4 nanocomposites were developed for bone tissue engineering.
Collapse
Affiliation(s)
- Arundhati Bhowmick
- Department of Polymer Science and Technology
- University of Calcutta
- Kolkata-700009
- India
| | - Nilkamal Pramanik
- Department of Polymer Science and Technology
- University of Calcutta
- Kolkata-700009
- India
| | - Tapas Mitra
- Department of Polymer Science and Technology
- University of Calcutta
- Kolkata-700009
- India
| | - Arumugam Gnanamani
- Microbiology Division
- CSIR-Central Leather Research Institute
- Chennai-600020
- India
| | - Manas Das
- Department of Chemical Engineering
- University of Calcutta
- Kolkata-700009
- India
| | - Patit Paban Kundu
- Department of Polymer Science and Technology
- University of Calcutta
- Kolkata-700009
- India
- Department of Chemical Engineering
| |
Collapse
|
62
|
Abstract
Cryopreservation is the application of low temperatures to preserve the structural and functional integrity of cells and tissues. Conventional cooling protocols allow ice to form and solute concentrations to rise during the cryopreservation process. The damage caused by the rise in solute concentration can be mitigated by the use of compounds known as cryoprotectants. Such compounds protect cells from the consequences of slow cooling injury, allowing them to be cooled at cooling rates which avoid the lethal effects of intracellular ice. An alternative to conventional cooling is vitrification. Vitrification methods incorporate cryoprotectants at sufficiently high concentrations to prevent ice crystallization so that the system forms an amorphous glass thus avoiding the damaging effects caused by conventional slow cooling. However, vitrification too can impose damaging consequences on cells as the cryoprotectant concentrations required to vitrify cells at lower cooling rates are potentially, and often, harmful. While these concentrations can be lowered to nontoxic levels, if the cells are ultra-rapidly cooled, the resulting metastable system can lead to damage through devitrification and growth of ice during subsequent storage and rewarming if not appropriately handled.The commercial and clinical application of stem cells requires robust and reproducible cryopreservation protocols and appropriate long-term, low-temperature storage conditions to provide reliable master and working cell banks. Though current Good Manufacturing Practice (cGMP) compliant methods for the derivation and banking of clinical grade pluripotent stem cells exist and stem cell lines suitable for clinical applications are available, current cryopreservation protocols, whether for vitrification or conventional slow freezing, remain suboptimal. Apart from the resultant loss of valuable product that suboptimal cryopreservation engenders, there is a danger that such processes will impose a selective pressure on the cells selecting out a nonrepresentative, freeze-resistant subpopulation. Optimizing this process requires knowledge of the fundamental processes that occur during the freezing of cellular systems, the mechanisms of damage and methods for avoiding them. This chapter draws together the knowledge of cryopreservation gained in other systems with the current state-of-the-art for embryonic and induced pluripotent stem cell preservation in an attempt to provide the background for future attempts to optimize cryopreservation protocols.
Collapse
Affiliation(s)
- Charles J Hunt
- UK Stem Cell Bank, National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Potters Bar, Hertfordshire, EN6 3QG, UK.
| |
Collapse
|
63
|
Brouwer M, Zhou H, Nadif Kasri N. Choices for Induction of Pluripotency: Recent Developments in Human Induced Pluripotent Stem Cell Reprogramming Strategies. Stem Cell Rev Rep 2016; 12:54-72. [PMID: 26424535 PMCID: PMC4720703 DOI: 10.1007/s12015-015-9622-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The ability to generate human induced pluripotent stem cells (iPSCs) from somatic cells provides tremendous promises for regenerative medicine and its use has widely increased over recent years. However, reprogramming efficiencies remain low and chromosomal instability and tumorigenic potential are concerns in the use of iPSCs, especially in clinical settings. Therefore, reprogramming methods have been under development to generate safer iPSCs with higher efficiency and better quality. Developments have mainly focused on the somatic cell source, the cocktail of reprogramming factors, the delivery method used to introduce reprogramming factors and culture conditions to maintain the generated iPSCs. This review discusses the developments on these topics and briefly discusses pros and cons of iPSCs in comparison with human embryonic stem cells generated from somatic cell nuclear transfer.
Collapse
Affiliation(s)
- Marinka Brouwer
- Department of Cognitive Neuroscience, Radboudumc, Nijmegen, 6500, HB, The Netherlands
| | - Huiqing Zhou
- Department of Human Genetics, Radboudumc, Nijmegen, 6500, HB, The Netherlands. .,Department of Molecular Developmental Biology, Faculty of Science, Radboud University, Nijmegen, 6500, HB, The Netherlands.
| | - Nael Nadif Kasri
- Department of Cognitive Neuroscience, Radboudumc, Nijmegen, 6500, HB, The Netherlands. .,Department of Human Genetics, Radboudumc, Nijmegen, 6500, HB, The Netherlands. .,Donders Institute for Brain, Cognition, and Behaviour , Centre for Neuroscience, Nijmegen, 6525, AJ, The Netherlands.
| |
Collapse
|
64
|
Zheng Y, Ji S, Wu H, Tian S, Zhang Y, Wang L, Fang H, Luo P, Wang X, Hu X, Xiao S, Xia Z. Topical administration of cryopreserved living micronized amnion accelerates wound healing in diabetic mice by modulating local microenvironment. Biomaterials 2016; 113:56-67. [PMID: 27810642 DOI: 10.1016/j.biomaterials.2016.10.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/20/2016] [Accepted: 10/20/2016] [Indexed: 01/09/2023]
Abstract
Approximately 25% of diabetic patients suffer from diabetic lower-extremity ulcer throughout their lives and 7%-20% of patients will eventually need an amputation despite standard care treatment. The development of new therapies to treat diabetic wounds is urgent. In this study, we used cryopreserved living micronized amnion (300-600 μm) to treat wounds in diabetic mice. Post-thaw micronized amnion retained high cell viability, as well as intact cell morphology and membrane structure. When transplanted onto the wounds of db/db mice, the cryopreserved living micronized amnion greatly promoted wound healing in diabetic mice mainly by secreting growth, inflammation, and chemotaxis-related factors that regulated macrophage migration and phenotype switch, recruited CD34+ progenitor cells, and increased neovascularization. In addition, the micronized amnion matrix can exist in the dermis and serve as a long-term dermal scaffold. These results demonstrated the potential of the cryopreserved living micronized amnion as a ready-to-use living dermal substitute that addresses multiple defective physiological processes of impaired wounds to treat diabetic ulcers and other chronic wounds in clinics.
Collapse
Affiliation(s)
- Yongjun Zheng
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Shizhao Ji
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China.
| | - Haibin Wu
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Song Tian
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Yunqing Zhang
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Li Wang
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - He Fang
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Pengfei Luo
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Xingtong Wang
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Xiaoyan Hu
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Shichu Xiao
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China.
| | - Zhaofan Xia
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China.
| |
Collapse
|
65
|
Almela T, Brook IM, Moharamzadeh K. The significance of cell-related challenges in the clinical application of tissue engineering. J Biomed Mater Res A 2016; 104:3157-3163. [DOI: 10.1002/jbm.a.35856] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 06/24/2016] [Accepted: 08/04/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Thafar Almela
- School of Clinical Dentistry; University of Sheffield, Claremont Crescent; Sheffield S10 2TA United Kingdom
| | - Ian M. Brook
- School of Clinical Dentistry; University of Sheffield, Claremont Crescent; Sheffield S10 2TA United Kingdom
| | - Keyvan Moharamzadeh
- School of Clinical Dentistry; University of Sheffield, Claremont Crescent; Sheffield S10 2TA United Kingdom
| |
Collapse
|
66
|
Gomes-Alves P, Serra M, Brito C, Ricardo CP, Cunha R, Sousa MF, Sanchez B, Bernad A, Carrondo MJT, Rodriguez-Borlado L, Alves PM. In vitro expansion of human cardiac progenitor cells: exploring 'omics tools for characterization of cell-based allogeneic products. Transl Res 2016; 171:96-110.e1-3. [PMID: 26924043 DOI: 10.1016/j.trsl.2016.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/27/2016] [Accepted: 02/02/2016] [Indexed: 01/15/2023]
Abstract
Human cardiac stem/progenitor cells (hCPCs) have been shown to be capable to regenerate contractile myocardium. However, because of their relative low abundance in the heart, in vitro expansion of hCPC is mandatory to achieve necessary quantities for allogeneic or autologous cardiac regeneration therapy applications (10(6)-10(9) cells/patient). Up to now, cell number requirements of ongoing phase I/IIa trials have been fulfilled with production in static monolayer cultures. However, this manufacturing process poses critical limitations when moving to the following clinical phases where hundreds of patients will be enrolled. For this, increased process yield is required, while guaranteeing the quality of the cell-based products. In this work, we developed and validated a robust, scalable, and good manufacturing practice (GMP)-compatible bioprocess for the expansion of high-quality hCPC. We applied platforms extensively used by the biopharmaceutical industry, such as microcarrier technology and stirred systems, and assessed culture conditions' impact on hCPC's quality and potency, as required by regulatory agencies. Complementary analytical assays including gene expression microarrays and mass spectrometry-based approaches were explored to compare transcriptome, proteome, surface markers, and secretion profiles of hCPC cultured in static monolayers and in stirred microcarrier-based systems. Our results show that stirred microcarrier-based culture systems enabled achieving more than 3-fold increase in hCPC expansion, when compared with traditional static monolayers, while retaining cell's phenotype and similar "omics" profiles. These findings demonstrate that this change in the production process does not affect cell's identity and quality, with potential to be translated into a transversal production platform for clinical development of stem-cell therapies.
Collapse
Affiliation(s)
- P Gomes-Alves
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - M Serra
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - C Brito
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - C P Ricardo
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - R Cunha
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - M F Sousa
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - B Sanchez
- Coretherapix, Tres Cantos, Madrid, Spain
| | - A Bernad
- Centro Nacional de Biotecnología, Madrid, Spain
| | - M J T Carrondo
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal; Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Monte da Caparica, Portugal
| | | | - P M Alves
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.
| |
Collapse
|
67
|
Richardson T, Barner S, Candiello J, Kumta PN, Banerjee I. Capsule stiffness regulates the efficiency of pancreatic differentiation of human embryonic stem cells. Acta Biomater 2016; 35:153-65. [PMID: 26911881 DOI: 10.1016/j.actbio.2016.02.025] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/24/2015] [Accepted: 02/17/2016] [Indexed: 12/14/2022]
Abstract
Encapsulation of donor islets using a hydrogel material is a well-studied strategy for islet transplantation, which protects donor islets from the host immune response. Replacement of donor islets by human embryonic stem cell (hESC) derived islets will also require a means of immune-isolating hESCs by encapsulation. However, a critical consideration of hESC differentiation is the effect of surrounding biophysical environment, in this case capsule biophysical properties, on differentiation. The objective of this study, thus, was to evaluate the effect of capsule properties on growth, viability, and differentiation of encapsulated hESCs throughout pancreatic induction. It was observed that even in the presence of soluble chemical cues for pancreatic induction, substrate properties can significantly modulate pancreatic differentiation, hence necessitating careful tuning of capsule properties. Capsules in the range of 4-7kPa supported cell growth and viability, whereas capsules of higher stiffness suppressed cell growth. While an increase in capsule stiffness enhanced differentiation at the intermediate definitive endoderm (DE) stage, increased stiffness strongly suppressed pancreatic progenitor (PP) induction. Signaling pathway analysis indicated an increase in pSMAD/pAKT levels with substrate stiffness likely the cause of enhancement of DE differentiation. In contrast, sonic hedgehog inhibition was more efficient under softer gel conditions, which is necessary for successful PP differentiation. STATEMENT OF SIGNIFICANCE Cell replacement therapy for type 1 diabetes (T1D), affecting millions of people worldwide, requires the immunoisolation of insulin-producing islets by encapsulation with a semi-impermeable material. Due to the shortage of donor islets, human pluripotent stem cell (hPSC) derived islets are an attractive alternative. However, properties of the encapsulating substrate are known to influence hPSC cell fate. In this work, we determine the effect of substrate stiffness on growth and pancreatic fate of encapsulated hPSCs. We precisely identify the range of substrate properties conducive for pancreatic cell fate, and also the mechanism by which substrate properties modify the cell signaling pathways and hence cell fate. Such information will be critical in driving regenerative cell therapy for long term treatment of T1D.
Collapse
Affiliation(s)
- Thomas Richardson
- Department of Chemical Engineering, University of Pittsburgh, United States
| | - Sierra Barner
- Department of Chemical Engineering, University of Pittsburgh, United States
| | - Joseph Candiello
- Department of Bioengineering, University of Pittsburgh, United States
| | - Prashant N Kumta
- Department of Chemical Engineering, University of Pittsburgh, United States; McGowan Institute of Regenerative Medicine, University of Pittsburgh, United States; Department of Bioengineering, University of Pittsburgh, United States; Department of Mechanical and Materials Science, University of Pittsburgh, United States; Department of Oral Biology, University of Pittsburgh, United States
| | - Ipsita Banerjee
- Department of Chemical Engineering, University of Pittsburgh, United States; McGowan Institute of Regenerative Medicine, University of Pittsburgh, United States; Department of Bioengineering, University of Pittsburgh, United States.
| |
Collapse
|
68
|
Hara J, Tottori J, Anders M, Dadhwal S, Asuri P, Mobed-Miremadi M. Trehalose effectiveness as a cryoprotectant in 2D and 3D cell cultures of human embryonic kidney cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:609-616. [DOI: 10.3109/21691401.2016.1167698] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Jared Hara
- Department of Bioengineering, Santa Clara University, Santa Clara, CA, USA
| | - Jordan Tottori
- Department of Bioengineering, Santa Clara University, Santa Clara, CA, USA
| | - Megan Anders
- Department of Bioengineering, Santa Clara University, Santa Clara, CA, USA
| | - Smritee Dadhwal
- Department of Bioengineering, Santa Clara University, Santa Clara, CA, USA
| | - Prashanth Asuri
- Department of Bioengineering, Santa Clara University, Santa Clara, CA, USA
| | | |
Collapse
|
69
|
Correia C, Koshkin A, Carido M, Espinha N, Šarić T, Lima PA, Serra M, Alves PM. Effective Hypothermic Storage of Human Pluripotent Stem Cell-Derived Cardiomyocytes Compatible With Global Distribution of Cells for Clinical Applications and Toxicology Testing. Stem Cells Transl Med 2016; 5:658-69. [PMID: 27025693 DOI: 10.5966/sctm.2015-0238] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 01/13/2016] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED To fully explore the potential of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs), efficient methods for storage and shipment of these cells are required. Here, we evaluated the feasibility to cold store monolayers and aggregates of functional CMs obtained from different PSC lines using a fully defined clinical-compatible preservation formulation and investigated the time frame that hPSC-CMs could be subjected to hypothermic storage. We showed that two-dimensional (2D) monolayers of hPSC-CMs can be efficiently stored at 4°C for 3 days without compromising cell viability. However, cell viability decreased when the cold storage interval was extended to 7 days. We demonstrated that hPSC-CMs are more resistant to prolonged hypothermic storage-induced cell injury in three-dimensional aggregates than in 2D monolayers, showing high cell recoveries (>70%) after 7 days of storage. Importantly, hPSC-CMs maintained their typical (ultra)structure, gene and protein expression profile, electrophysiological profiles, and drug responsiveness. SIGNIFICANCE The applicability of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) in the clinic/industry is highly dependent on the development of efficient methods for worldwide shipment of these cells. This study established effective clinically compatible strategies for cold (4°C) storage of hPSC-CMs cultured as two-dimensional (2D) monolayers and three-dimensional (3D) aggregates. Cell recovery of 2D monolayers of hPSC-CMs was found to be dependent on the time of storage, and 3D cell aggregates were more resistant to prolonged cold storage than 2D monolayers. Of note, it was demonstrated that 7 days of cold storage did not affect hPSC-CM ultrastructure, phenotype, or function. This study provides important insights into the cold preservation of PSC-CMs that could be valuable in improving global commercial distribution of hPSC-CMs.
Collapse
Affiliation(s)
- Cláudia Correia
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Alexey Koshkin
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Madalena Carido
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Nuno Espinha
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Tomo Šarić
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Pedro A Lima
- Nova Medical School, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Margarida Serra
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Paula M Alves
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| |
Collapse
|
70
|
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
|
71
|
Dumbleton J, Agarwal P, Huang H, Hogrebe N, Han R, Gooch KJ, He X. The effect of RGD peptide on 2D and miniaturized 3D culture of HEPM cells, MSCs, and ADSCs with alginate hydrogel. Cell Mol Bioeng 2016; 9:277-288. [PMID: 27990180 DOI: 10.1007/s12195-016-0428-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Advancements in tissue engineering require the development of new technologies to study cell behavior in vitro. This study focuses on stem cell behavior within various miniaturized three-dimensional (3D) culture conditions of alginate biomaterials modified with the Arg-Gly-Asp (RGD) peptide known for its role in cell adhesion/attachment. Human embryonic palatal mesenchyme (HEPM) cells, bone marrow derived mesenchymal stem cells (MSCs), and human adipose derived stem cells (ADSCs) were cultured on a flat hydrogel of different concentrations of alginate-RGD, and in the miniaturized 3D core of microcapsules with either a 2% alginate or 2% alginate-RGD shell. The core was made of 0%, 0.5%, or 2% alginate-RGD. Cell spreading was observed in all systems containing the RGD peptide, and the cell morphology was quantified by measuring the cell surface area and circularity. In all types of stem cells, there was a significant increase in the cell surface area (p < 0.05) and a significant decrease in cell circularity (p < 0.01) in alginate-RGD conditions, indicating that cells spread much more readily in environments containing the peptide. This control over the cell spreading within a 3D microenvironment can help to create the ideal biomimetic condition in which to conduct further studies on cell behavior.
Collapse
Affiliation(s)
- Jenna Dumbleton
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210 (USA); Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210 (USA)
| | - Pranay Agarwal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210 (USA); Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210 (USA)
| | - Haishui Huang
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210 (USA); Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210 (USA); Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH 43210 (USA)
| | - Nathaniel Hogrebe
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210 (USA); Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210 (USA)
| | - Renzhi Han
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210 (USA); Department of Surgery, The Ohio State University, Columbus, OH 43210 (USA)
| | - Keith J Gooch
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210 (USA); Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210 (USA)
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210 (USA); Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210 (USA); Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210 (USA)
| |
Collapse
|
72
|
Santo VE, Estrada MF, Rebelo SP, Abreu S, Silva I, Pinto C, Veloso SC, Serra AT, Boghaert E, Alves PM, Brito C. Adaptable stirred-tank culture strategies for large scale production of multicellular spheroid-based tumor cell models. J Biotechnol 2016; 221:118-29. [PMID: 26815388 DOI: 10.1016/j.jbiotec.2016.01.031] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 01/15/2016] [Accepted: 01/21/2016] [Indexed: 12/29/2022]
Abstract
Currently there is an effort toward the development of in vitro cancer models more predictive of clinical efficacy. The onset of advanced analytical tools and imaging technologies has increased the utilization of spheroids in the implementation of high throughput approaches in drug discovery. Agitation-based culture systems are commonly proposed as an alternative method for the production of tumor spheroids, despite the scarce experimental evidence found in the literature. In this study, we demonstrate the robustness and reliability of stirred-tank cultures for the scalable generation of 3D cancer models. We developed standardized protocols to a panel of tumor cell lines from different pathologies and attained efficient tumor cell aggregation by tuning hydrodynamic parameters. Large numbers of spheroids were obtained (typically 1000-1500 spheroids/mL) presenting features of native tumors, namely morphology, proliferation and hypoxia gradients, in a cell line-dependent mode. Heterotypic 3D cancer models, based on co-cultures of tumor cells and fibroblasts, were also established in the absence or presence of additional physical support from an alginate matrix, with maintenance of high cell viability. Altogether, we demonstrate that 3D tumor cell model production in stirred-tank culture systems is a robust and versatile approach, providing reproducible tools for drug screening and target verification in pre-clinical oncology research.
Collapse
Affiliation(s)
- Vítor E Santo
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Marta F Estrada
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Sofia P Rebelo
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Sofia Abreu
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Inês Silva
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Catarina Pinto
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Susana C Veloso
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Ana Teresa Serra
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | | | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal.
| |
Collapse
|
73
|
Demont A, Cole H, Marison IW. An understanding of potential and limitations of alginate/PLL microcapsules as a cell retention system for perfusion cultures. J Microencapsul 2016; 33:80-8. [PMID: 26754597 DOI: 10.3109/02652048.2015.1134686] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Microcapsules for high cell density culture of mammalian cells have found an increasing interest, however, the poor stability of the microcapsules and the lack of characterisation methods led to few quantitative results. Alginate-poly-L-lysine (PLL) microcapsules have been studied in detail in order to form a basis for comparison of capsules made from different polymers. Since the microcapsules can be easily retained in the bioreactor without the need for a cell separation device, high cell densities were achieved with a maximum of 4 × 10(7) cell/ml(microcapsules), corresponding to a colonisation of 5% of the internal capsule volume. Measurement of microcapsule integrity and mechanical resistance showed that alginate-PLL microcapsules are not suitable for perfusion cultures since they are very sensitive to media composition, mainly the presence of non-gelling ions that have a higher affinity for alginate than PLL and Ca(2+), leading to the leakage of PLL and Ca(2+), and to microcapsule rupture.
Collapse
Affiliation(s)
- Aurelie Demont
- a Laboratory of Integrated Bioprocessing, School of Biotechnology , Dublin City University , Dublin , Ireland
| | - Harriet Cole
- a Laboratory of Integrated Bioprocessing, School of Biotechnology , Dublin City University , Dublin , Ireland
| | - Ian W Marison
- a Laboratory of Integrated Bioprocessing, School of Biotechnology , Dublin City University , Dublin , Ireland
| |
Collapse
|
74
|
Production of human pluripotent stem cell therapeutics under defined xeno-free conditions: progress and challenges. Stem Cell Rev Rep 2015; 11:96-109. [PMID: 25077810 DOI: 10.1007/s12015-014-9544-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Recent advances on human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) have brought us closer to the realization of their clinical potential. Nonetheless, tissue engineering and regenerative medicine applications will require the generation of hPSC products well beyond the laboratory scale. This also mandates the production of hPSC therapeutics in fully-defined, xeno-free systems and in a reproducible manner. Toward this goal, we summarize current developments in defined media free of animal-derived components for hPSC culture. Bioinspired and synthetic extracellular matrices for the attachment, growth and differentiation of hPSCs are also reviewed. Given that most progress in xeno-free medium and substrate development has been demonstrated in two-dimensional rather than three dimensional culture systems, translation from the former to the latter poses unique difficulties. These challenges are discussed in the context of cultivation platforms of hPSCs as aggregates, on microcarriers or after encapsulation in biocompatible scaffolds.
Collapse
|
75
|
Formo K, Cho CHH, Vallier L, Strand BL. Culture of hESC-derived pancreatic progenitors in alginate-based scaffolds. J Biomed Mater Res A 2015; 103:3717-26. [PMID: 26014279 DOI: 10.1002/jbm.a.35507] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 04/22/2015] [Accepted: 05/13/2015] [Indexed: 12/15/2022]
Abstract
The effect of alginate-based scaffolds with added basement membrane proteins on the in vitro development of hESC-derived pancreatic progenitors was investigated. Cell clusters were encapsulated in scaffolds containing the basement membrane proteins collagen IV, laminin, fibronectin, or extracellular matrix-derived peptides, and maintained in culture for up to 46 days. The cells remained viable throughout the experiment with no signs of central necrosis. Whereas nonencapsulated cells aggregated into larger clusters, some of which showed signs of morphological changes and tissue organization, the alginate matrix stabilized the cluster size and displayed more homogeneous cell morphologies, allowing culture for long periods of time. For all conditions tested, a stable or declining expression of insulin and PDX1 and an increase in glucagon and somatostatin over time indicated a progressive reduction in beta cell-related gene expression. Alginate scaffolds can provide a chemically defined, xeno-free and easily scalable alternative for culture of pancreatic progenitors. Although no increase in insulin and PDX1 gene expression after alginate-immobilized cell culture was seen in this study, further optimization of the matrix physicochemical and biological properties and of the medium composition may still be a relevant strategy to promote the stabilization or maturation of stem cell-derived beta cells.
Collapse
Affiliation(s)
- Kjetil Formo
- Department of Biotechnology, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Candy H-H Cho
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ludovic Vallier
- Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Berit L Strand
- Department of Biotechnology, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Central Norwegian Regional Health Authority, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
76
|
Huang H, Choi JK, Rao W, Zhao S, Agarwal P, Zhao G, He X. Alginate Hydrogel Microencapsulation Inhibits Devitrification and Enables Large-Volume Low-CPA Cell Vitrification. ADVANCED FUNCTIONAL MATERIALS 2015; 25:6939-6850. [PMID: 26640426 PMCID: PMC4667367 DOI: 10.1002/adfm.201503047] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Cryopreservation of stem cells is important to meet their ever-increasing demand by the burgeoning cell-based medicine. The conventional slow freezing for stem cell cryopreservation suffers from inevitable cell injury associated with ice formation and the vitrification (i.e., no visible ice formation) approach is emerging as a new strategy for cell cryopreservation. A major challenge to cell vitrification is intracellular ice formation (IIF, a lethal event to cells) induced by devitrification (i.e., formation of visible ice in previously vitrified solution) during warming the vitrified cells at cryogenic temperature back to super-zero temperatures. Consequently, high and toxic concentrations of penetrating cryoprotectants (i.e., high CPAs, up to ~8 M) and/or limited sample volumes (up to ~2.5 μl) have been used to minimize IIF during vitrification. We reveal that alginate hydrogel microencapsulation can effectively inhibit devitrification during warming. Our data show that if ice formation were minimized during cooling, IIF is negligible in alginate hydrogel-microencapsulated cells during the entire cooling and warming procedure of vitrification. This enables vitrification of pluripotent and multipotent stem cells with up to ~4 times lower concentration of penetrating CPAs (up to 2 M, low CPA) in up to ~100 times larger sample volume (up to ~250 μl, large volume).
Collapse
Affiliation(s)
- Haishui Huang
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA. Department of Mechanical Engineering, The Ohio State University, Columbus, OH 43210, USA. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jung Kyu Choi
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Wei Rao
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Shuting Zhao
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Pranay Agarwal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Gang Zhao
- Center for Biomedical Engineering, Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA. Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
77
|
Kumar A, Starly B. Large scale industrialized cell expansion: producing the critical raw material for biofabrication processes. Biofabrication 2015; 7:044103. [DOI: 10.1088/1758-5090/7/4/044103] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
78
|
Cunha B, Aguiar T, Silva MM, Silva RJ, Sousa MF, Pineda E, Peixoto C, Carrondo MJ, Serra M, Alves PM. Exploring continuous and integrated strategies for the up- and downstream processing of human mesenchymal stem cells. J Biotechnol 2015; 213:97-108. [DOI: 10.1016/j.jbiotec.2015.02.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 02/08/2015] [Accepted: 02/16/2015] [Indexed: 01/08/2023]
|
79
|
Huang H, Sun M, Heisler-Taylor T, Kiourti A, Volakis J, Lafyatis G, He X. Stiffness-Independent Highly Efficient On-Chip Extraction of Cell-Laden Hydrogel Microcapsules from Oil Emulsion into Aqueous Solution by Dielectrophoresis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:5369-74. [PMID: 26297051 PMCID: PMC4690616 DOI: 10.1002/smll.201501388] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 07/23/2015] [Indexed: 05/21/2023]
Abstract
A dielectrophoresis (DEP)-based method achieves highly efficient on-chip extraction of cell-laden microcapsules of any stiffness from oil into aqueous solution. The hydrogel microcapsules can be extracted into the aqueous solution by DEP and interfacial tension forces with no trapped oil, while the encapsulated cells are free from electrical damage due to the Faraday cage effect.
Collapse
Affiliation(s)
- Haishui Huang
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Mingrui Sun
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Tyler Heisler-Taylor
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Asimina Kiourti
- Department of Electrical and Computer Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - John Volakis
- Department of Electrical and Computer Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Gregory Lafyatis
- Department of Physics, The Ohio State University, Columbus, Ohio 43210, USA
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| |
Collapse
|
80
|
Brouwer M, Zhou H, Nadif Kasri N. Choices for Induction of Pluripotency: Recent Developments in Human Induced Pluripotent Stem Cell Reprogramming Strategies. Stem Cell Rev Rep 2015. [PMID: 26424535 DOI: 10.1007/s12015‐015‐9622‐8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The ability to generate human induced pluripotent stem cells (iPSCs) from somatic cells provides tremendous promises for regenerative medicine and its use has widely increased over recent years. However, reprogramming efficiencies remain low and chromosomal instability and tumorigenic potential are concerns in the use of iPSCs, especially in clinical settings. Therefore, reprogramming methods have been under development to generate safer iPSCs with higher efficiency and better quality. Developments have mainly focused on the somatic cell source, the cocktail of reprogramming factors, the delivery method used to introduce reprogramming factors and culture conditions to maintain the generated iPSCs. This review discusses the developments on these topics and briefly discusses pros and cons of iPSCs in comparison with human embryonic stem cells generated from somatic cell nuclear transfer.
Collapse
Affiliation(s)
- Marinka Brouwer
- Department of Cognitive Neuroscience, Radboudumc, Nijmegen, 6500, HB, The Netherlands
| | - Huiqing Zhou
- Department of Human Genetics, Radboudumc, Nijmegen, 6500, HB, The Netherlands. .,Department of Molecular Developmental Biology, Faculty of Science, Radboud University, Nijmegen, 6500, HB, The Netherlands.
| | - Nael Nadif Kasri
- Department of Cognitive Neuroscience, Radboudumc, Nijmegen, 6500, HB, The Netherlands. .,Department of Human Genetics, Radboudumc, Nijmegen, 6500, HB, The Netherlands. .,Donders Institute for Brain, Cognition, and Behaviour , Centre for Neuroscience, Nijmegen, 6525, AJ, The Netherlands.
| |
Collapse
|
81
|
Fan R, Naqvi K, Patel K, Sun J, Wan J. Evaporation-based microfluidic production of oil-free cell-containing hydrogel particles. BIOMICROFLUIDICS 2015; 9:052602. [PMID: 25825624 PMCID: PMC4376759 DOI: 10.1063/1.4916508] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/27/2015] [Indexed: 05/16/2023]
Abstract
We demonstrate an evaporation-based microfluidic strategy to produce oil-free cell containing hydrogel particles. Perfluoro-n-pentane, which is used as the continuous oil phase to generate cell-containing hydrogel (Extracel) particles, is removed at an elevated temperature. Human colon cancer cells (HCT116) encapsulated in the hydrogel particles show higher viability than cells encapsulated in particles that are produced via a non-evaporative oil phase. In addition, single HCT116 cells can be cultured for a week in such particles and respond to inflammatory stimuli, highlighting the potential applications of the developed strategy for 3D cell culture, drug testing, and cell-based drug delivery.
Collapse
Affiliation(s)
- Rong Fan
- Microsystems Engineering, Rochester Institute of Technology , Rochester, New York 14623, USA
| | - Kubra Naqvi
- College of Science, Rochester Institute of Technology , Rochester, New York 14623, USA
| | - Krishna Patel
- Webster Schroeder High School , Webster, New York 14580, USA
| | - Jun Sun
- Department of Biochemistry, Rush University , Chicago, Illinois 60612, USA
| | - Jiandi Wan
- Microsystems Engineering, Rochester Institute of Technology , Rochester, New York 14623, USA
| |
Collapse
|
82
|
Simão D, Pinto C, Fernandes P, Peddie CJ, Piersanti S, Collinson LM, Salinas S, Saggio I, Schiavo G, Kremer EJ, Brito C, Alves PM. Evaluation of helper-dependent canine adenovirus vectors in a 3D human CNS model. Gene Ther 2015; 23:86-94. [PMID: 26181626 DOI: 10.1038/gt.2015.75] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/23/2015] [Accepted: 07/08/2015] [Indexed: 01/24/2023]
Abstract
Gene therapy is a promising approach with enormous potential for treatment of neurodegenerative disorders. Viral vectors derived from canine adenovirus type 2 (CAV-2) present attractive features for gene delivery strategies in the human brain, by preferentially transducing neurons, are capable of efficient axonal transport to afferent brain structures, have a 30-kb cloning capacity and have low innate and induced immunogenicity in preclinical tests. For clinical translation, in-depth preclinical evaluation of efficacy and safety in a human setting is primordial. Stem cell-derived human neural cells have a great potential as complementary tools by bridging the gap between animal models, which often diverge considerably from human phenotype, and clinical trials. Herein, we explore helper-dependent CAV-2 (hd-CAV-2) efficacy and safety for gene delivery in a human stem cell-derived 3D neural in vitro model. Assessment of hd-CAV-2 vector efficacy was performed at different multiplicities of infection, by evaluating transgene expression and impact on cell viability, ultrastructural cellular organization and neuronal gene expression. Under optimized conditions, hd-CAV-2 transduction led to stable long-term transgene expression with minimal toxicity. hd-CAV-2 preferentially transduced neurons, whereas human adenovirus type 5 (HAdV5) showed increased tropism toward glial cells. This work demonstrates, in a physiologically relevant 3D model, that hd-CAV-2 vectors are efficient tools for gene delivery to human neurons, with stable long-term transgene expression and minimal cytotoxicity.
Collapse
Affiliation(s)
- D Simão
- iBET-Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - C Pinto
- iBET-Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - P Fernandes
- iBET-Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - C J Peddie
- The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London, UK
| | - S Piersanti
- Dipartimento di Biologia e Biotecnologie 'Charles Darwin', Università di Roma La Sapienza, Rome, Italy
| | - L M Collinson
- The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London, UK
| | - S Salinas
- Institut de Génétique Moléculaire de Montpellier, Montpellier, France.,Université Montpellier, Montpellier, France
| | - I Saggio
- Dipartimento di Biologia e Biotecnologie 'Charles Darwin', Università di Roma La Sapienza, Rome, Italy.,Istituto Pasteur Fondazione Cenci Bolognetti, Università di Roma La Sapienza, Rome, Italy.,Istituto di Biologia e Patologia Molecolari del CNR, Università di Roma La Sapienza, Rome, Italy
| | - G Schiavo
- Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, University College London, London, UK
| | - E J Kremer
- Institut de Génétique Moléculaire de Montpellier, Montpellier, France.,Université Montpellier, Montpellier, France
| | - C Brito
- iBET-Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - P M Alves
- iBET-Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
83
|
Elanzew A, Sommer A, Pusch-Klein A, Brüstle O, Haupt S. A reproducible and versatile system for the dynamic expansion of human pluripotent stem cells in suspension. Biotechnol J 2015; 10:1589-99. [PMID: 26110829 DOI: 10.1002/biot.201400757] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 03/13/2015] [Accepted: 06/23/2015] [Indexed: 11/11/2022]
Abstract
Reprogramming of patient cells to human induced pluripotent stem cells (hiPSC) has facilitated in vitro disease modeling studies aiming at deciphering the molecular and cellular mechanisms that contribute to disease pathogenesis and progression. To fully exploit the potential of hiPSC for biomedical applications, technologies that enable the standardized generation and expansion of hiPSC from large numbers of donors are required. Paralleled automated processes for the expansion of hiPSC could provide an opportunity to maximize the generation of hiPSC collections from patient cohorts while minimizing hands-on time and costs. In order to develop a simple method for the parallel expansion of human pluripotent stem cells (hPSC) we established a protocol for their cultivation as undifferentiated aggregates in a bench-top bioreactor system (BioLevitator™). We show that long-term expansion (10 passages) of hPSCs either in mTeSR or E8 medium preserved a normal karyotype, three-germ-layer differentiation potential and high expression of pluripotency-associated markers. The system enables the expansion from low inoculation densities (0.3 × 10(5) cells/mL) and provides a simplified, cost-efficient and time-saving method for the provision of hiPSC at midi-scale. Implementation of this protocol in cell production schemes has the potential to advance cell manufacturing in many areas of hiPSC-based medical research.
Collapse
Affiliation(s)
- Andreas Elanzew
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, University of Bonn, Bonn, Germany.,LIFE&BRAIN GmbH, Bonn, Germany
| | | | | | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, University of Bonn, Bonn, Germany. .,LIFE&BRAIN GmbH, Bonn, Germany.
| | - Simone Haupt
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, University of Bonn, Bonn, Germany. .,LIFE&BRAIN GmbH, Bonn, Germany.
| |
Collapse
|
84
|
Wang Y, Wang J. Mixed hydrogel bead-based tumor spheroid formation and anticancer drug testing. Analyst 2015; 139:2449-58. [PMID: 24699505 DOI: 10.1039/c4an00015c] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Three-dimensional multicellular tumor spheroids have become critical for anticancer study since they may provide a better model than conventional monolayer cultures of cancer cells. Various methods for tumor spheroid formation have been explored. However, only one kind of hydrogel was used in these methods, which has an influence on the size and morphology of the obtained tumor spheroids. Herein, we present a microfluidic droplet-based method for the formation of multicellular tumor spheroids using alginate and matrigel mixed hydrogel beads. By on-chip changing the flow rate of the two hydrogel solutions, mixed hydrogel beads with different volume ratios between alginate and matrigel are obtained. Meanwhile, human cervical carcinoma (HeLa) cells are encapsulated in the mixed hydrogel beads. Acridine orange and propidium iodide double-staining assay shows that the viability of cells encapsulated in the mixed hydrogel beads was more than 90%. After 4 day culture, the multicellular tumor spheroids were successfully formed with spherical shape and uniform size distribution compared with spheroids formed in pure alginate beads. Cytoskeletal analysis by TRITC-phalloidin staining show that HeLa cells in the mixed hydrogel beads closely link to each other. The dose-dependent response assay of HeLa cell spheroids to vincristine show that multicellular spheroids have more powerful resistance to vincristine compared to conventional monolayer culture cells. Taken together, this novel technology may be of importance to facilitate in vitro culture of tumor spheroids for their ever-increasing utilization in modern cell-based medicine.
Collapse
Affiliation(s)
- Yaolei Wang
- College of Science, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | | |
Collapse
|
85
|
Sambu S. A Bayesian approach to optimizing cryopreservation protocols. PeerJ 2015; 3:e1039. [PMID: 26131379 PMCID: PMC4485240 DOI: 10.7717/peerj.1039] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 05/30/2015] [Indexed: 11/20/2022] Open
Abstract
Cryopreservation is beset with the challenge of protocol alignment across a wide range of cell types and process variables. By taking a cross-sectional assessment of previously published cryopreservation data (sample means and standard errors) as preliminary meta-data, a decision tree learning analysis (DTLA) was performed to develop an understanding of target survival using optimized pruning methods based on different approaches. Briefly, a clear direction on the decision process for selection of methods was developed with key choices being the cooling rate, plunge temperature on the one hand and biomaterial choice, use of composites (sugars and proteins as additional constituents), loading procedure and cell location in 3D scaffolding on the other. Secondly, using machine learning and generalized approaches via the Naïve Bayes Classification (NBC) method, these metadata were used to develop posterior probabilities for combinatorial approaches that were implicitly recorded in the metadata. These latter results showed that newer protocol choices developed using probability elicitation techniques can unearth improved protocols consistent with multiple unidimensionally-optimized physical protocols. In conclusion, this article proposes the use of DTLA models and subsequently NBC for the improvement of modern cryopreservation techniques through an integrative approach.
Collapse
|
86
|
Silva MM, Rodrigues AF, Correia C, Sousa MFQ, Brito C, Coroadinha AS, Serra M, Alves PM. Robust Expansion of Human Pluripotent Stem Cells: Integration of Bioprocess Design With Transcriptomic and Metabolomic Characterization. Stem Cells Transl Med 2015; 4:731-42. [PMID: 25979863 DOI: 10.5966/sctm.2014-0270] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 03/09/2015] [Indexed: 12/30/2022] Open
Abstract
UNLABELLED : Human embryonic stem cells (hESCs) have an enormous potential as a source for cell replacement therapies, tissue engineering, and in vitro toxicology applications. The lack of standardized and robust bioprocesses for hESC expansion has hindered the application of hESCs and their derivatives in clinical settings. We developed a robust and well-characterized bioprocess for hESC expansion under fully defined conditions and explored the potential of transcriptomic and metabolomic tools for a more comprehensive assessment of culture system impact on cell proliferation, metabolism, and phenotype. Two different hESC lines (feeder-dependent and feeder-free lines) were efficiently expanded on xeno-free microcarriers in stirred culture systems. Both hESC lines maintained the expression of stemness markers such as Oct-4, Nanog, SSEA-4, and TRA1-60 and the ability to spontaneously differentiate into the three germ layers. Whole-genome transcriptome profiling revealed a phenotypic convergence between both hESC lines along the expansion process in stirred-tank bioreactor cultures, providing strong evidence of the robustness of the cultivation process to homogenize cellular phenotype. Under low-oxygen tension, results showed metabolic rearrangement with upregulation of the glycolytic machinery favoring an anaerobic glycolysis Warburg-effect-like phenotype, with no evidence of hypoxic stress response, in contrast to two-dimensional culture. Overall, we report a standardized expansion bioprocess that can guarantee maximal product quality. Furthermore, the "omics" tools used provided relevant findings on the physiological and metabolic changes during hESC expansion in environmentally controlled stirred-tank bioreactors, which can contribute to improved scale-up production systems. SIGNIFICANCE The clinical application of human pluripotent stem cells (hPSCs) has been hindered by the lack of robust protocols able to sustain production of high cell numbers, as required for regenerative medicine. In this study, a strategy was developed for the expansion of human embryonic stem cells in well-defined culture conditions using microcarrier technology and stirred-tank bioreactors. The use of transcriptomic and metabolic tools allowed detailed characterization of the cell-based product and showed a phenotypic convergence between both hESC lines along the expansion process. This study provided valuable insights into the metabolic hallmarks of hPSC expansion and new information to guide bioprocess design and media optimization for the production of cells with higher quantity and improved quality, which are requisite for translation to the clinic.
Collapse
Affiliation(s)
- Marta M Silva
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ana F Rodrigues
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Cláudia Correia
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Marcos F Q Sousa
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ana S Coroadinha
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Margarida Serra
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
87
|
Multipotent stromal cells derived from common marmoset Callithrix jacchus within alginate 3D environment: Effect of cryopreservation procedures. Cryobiology 2015; 71:103-11. [PMID: 25980899 DOI: 10.1016/j.cryobiol.2015.05.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 05/05/2015] [Accepted: 05/06/2015] [Indexed: 11/23/2022]
Abstract
Multipotent stromal cells derived from the common marmoset monkey Callithrix jacchus (cjMSCs) possess high phylogenetic similarity to humans, with a great potential for preclinical studies in the field of regenerative medicine. Safe and effective long-term storage of cells is of great significance to clinical and research applications. Encapsulation of such cell types within alginate beads that can mimic an extra-cellular matrix and provide a supportive environment for cells during cryopreservation, has several advantages over freezing of cells in suspension. In this study we have analysed the effect of dimethyl sulfoxide (Me2SO, 2.5-10%, v/v) and pre-freeze loading time of alginate encapsulated cjMSCs in Me2SO (0-45 min) on the viability and metabolic activity of the cells after freezing using a slow cooling rate (-1°C/min). It was found that these parameters affect the stability and homogeneity of alginate beads after thawing. Moreover, the cjMSCs can be frozen in alginate beads with lower Me2SO concentration of 7.5% after 30 min of loading, while retaining high cryopreservation outcome. We demonstrated the maximum viability, membrane integrity and metabolic activity of the cells under optimized, less cytotoxic conditions. The results of this study are another step forward towards the application of cryopreservation for the long-term storage and subsequent applications of transplants in cell-based therapies.
Collapse
|
88
|
Hickman JA, Graeser R, de Hoogt R, Vidic S, Brito C, Gutekunst M, van der Kuip H. Three-dimensional models of cancer for pharmacology and cancer cell biology: capturing tumor complexity in vitro/ex vivo. Biotechnol J 2015; 9:1115-28. [PMID: 25174503 DOI: 10.1002/biot.201300492] [Citation(s) in RCA: 262] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 07/11/2014] [Accepted: 08/05/2014] [Indexed: 12/12/2022]
Abstract
Cancers are complex and heterogeneous pathological "organs" in a dynamic interplay with their host. Models of human cancer in vitro, used in cancer biology and drug discovery, are generally highly reductionist. These cancer models do not incorporate complexity or heterogeneity. This raises the question as to whether the cancer models' biochemical circuitry (not their genome) represents, with sufficient fidelity, a tumor in situ. Around 95% of new anticancer drugs eventually fail in clinical trial, despite robust indications of activity in existing in vitro pre-clinical models. Innovative models are required that better capture tumor biology. An important feature of all tissues, and tumors, is that cells grow in three dimensions. Advances in generating and characterizing simple and complex (with added stromal components) three-dimensional in vitro models (3D models) are reviewed in this article. The application of stirred bioreactors to permit both scale-up/scale-down of these cancer models and, importantly, methods to permit controlled changes in environment (pH, nutrients, and oxygen) are also described. The challenges of generating thin tumor slices, their utility, and potential advantages and disadvantages are discussed. These in vitro/ex vivo models represent a distinct move to capture the realities of tumor biology in situ, but significant characterization work still remains to be done in order to show that their biochemical circuitry accurately reflects that of a tumor.
Collapse
|
89
|
Desai N, Rambhia P, Gishto A. Human embryonic stem cell cultivation: historical perspective and evolution of xeno-free culture systems. Reprod Biol Endocrinol 2015; 13:9. [PMID: 25890180 PMCID: PMC4351689 DOI: 10.1186/s12958-015-0005-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 02/09/2015] [Indexed: 01/23/2023] Open
Abstract
Human embryonic stem cells (hESC) have emerged as attractive candidates for cell-based therapies that are capable of restoring lost cell and tissue function. These unique cells are able to self-renew indefinitely and have the capacity to differentiate in to all three germ layers (ectoderm, endoderm and mesoderm). Harnessing the power of these pluripotent stem cells could potentially offer new therapeutic treatment options for a variety of medical conditions. Since the initial derivation of hESC lines in 1998, tremendous headway has been made in better understanding stem cell biology and culture requirements for maintenance of pluripotency. The approval of the first clinical trials of hESC cells for treatment of spinal cord injury and macular degeneration in 2010 marked the beginning of a new era in regenerative medicine. Yet it was clearly recognized that the clinical utility of hESC transplantation was still limited by several challenges. One of the most immediate issues has been the exposure of stem cells to animal pathogens, during hESC derivation and during in vitro propagation. Initial culture protocols used co-culture with inactivated mouse fibroblast feeder (MEF) or human feeder layers with fetal bovine serum or alternatively serum replacement proteins to support stem cell proliferation. Most hESC lines currently in use have been exposed to animal products, thus carrying the risk of xeno-transmitted infections and immune reaction. This mini review provides a historic perspective on human embryonic stem cell culture and the evolution of new culture models. We highlight the challenges and advances being made towards the development of xeno-free culture systems suitable for therapeutic applications.
Collapse
Affiliation(s)
- Nina Desai
- Department of Obstetrics and Gynecology, Cleveland Clinic, Beachwood, OH, USA.
| | - Pooja Rambhia
- Department of Obstetrics and Gynecology, Cleveland Clinic, Beachwood, OH, USA.
| | - Arsela Gishto
- Department of Obstetrics and Gynecology, Cleveland Clinic, Beachwood, OH, USA.
| |
Collapse
|
90
|
Abstract
Anchorage-dependent cells are of great interest for various biotechnological applications. (i) They represent a formidable production means of viruses for vaccination purposes at very large scales (in 1000-6000 l reactors) using microcarriers, and in the last decade many more novel viral vaccines have been developed using this production technology. (ii) With the advent of stem cells and their use/potential use in clinics for cell therapy and regenerative medicine purposes, the development of novel culture devices and technologies for adherent cells has accelerated greatly with a view to the large-scale expansion of these cells. Presently, the really scalable systems--microcarrier/microcarrier-clump cultures using stirred-tank reactors--for the expansion of stem cells are still in their infancy. Only laboratory scale reactors of maximally 2.5 l working volume have been evaluated because thorough knowledge and basic understanding of critical issues with respect to cell expansion while retaining pluripotency and differentiation potential, and the impact of the culture environment on stem cell fate, etc., are still lacking and require further studies. This article gives an overview on critical issues common to all cell culture systems for adherent cells as well as specifics for different types of stem cells in view of small- and large-scale cell expansion and production processes.
Collapse
|
91
|
Simão D, Pinto C, Piersanti S, Weston A, Peddie CJ, Bastos AE, Licursi V, Schwarz SC, Collinson LM, Salinas S, Serra M, Teixeira AP, Saggio I, Lima PA, Kremer EJ, Schiavo G, Brito C, Alves PM. Modeling Human Neural Functionality In Vitro: Three-Dimensional Culture for Dopaminergic Differentiation. Tissue Eng Part A 2015; 21:654-68. [DOI: 10.1089/ten.tea.2014.0079] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Daniel Simão
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Catarina Pinto
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Stefania Piersanti
- Dipartimento di Biologia e Biotecnologie “Charles Darwin,” Università di Roma La Sapienza, Rome, Italy
| | - Anne Weston
- Lincoln's Inn Fields Laboratories, Cancer Research UK London Research Institute, London, United Kingdom
| | - Christopher J. Peddie
- Lincoln's Inn Fields Laboratories, Cancer Research UK London Research Institute, London, United Kingdom
| | - André E.P. Bastos
- NOVA Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisboa, Portugal
- Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade de Lisboa, Lisboa, Portugal
| | - Valerio Licursi
- Dipartimento di Biologia e Biotecnologie “Charles Darwin,” Università di Roma La Sapienza, Rome, Italy
| | | | - Lucy M. Collinson
- Lincoln's Inn Fields Laboratories, Cancer Research UK London Research Institute, London, United Kingdom
| | - Sara Salinas
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Université Montpellier I and II, Montpellier, France
| | - Margarida Serra
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ana P. Teixeira
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Isabella Saggio
- Dipartimento di Biologia e Biotecnologie “Charles Darwin,” Università di Roma La Sapienza, Rome, Italy
- Istituto Pasteur Fondazione Cenci Bolognetti, Università di Roma La Sapienza, Rome, Italy
- Istituto di Biologia e Patologia Molecolari del CNR, Università di Roma La Sapienza, Rome, Italy
| | - Pedro A. Lima
- NOVA Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisboa, Portugal
| | - Eric J. Kremer
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Université Montpellier I and II, Montpellier, France
| | - Giampietro Schiavo
- Lincoln's Inn Fields Laboratories, Cancer Research UK London Research Institute, London, United Kingdom
- Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, University College London, London, United Kingdom
| | - Catarina Brito
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paula M. Alves
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
92
|
|
93
|
Higuchi A, Ling QD, Kumar SS, Munusamy M, Alarfajj AA, Umezawa A, Wu GJ. Design of polymeric materials for culturing human pluripotent stem cells: Progress toward feeder-free and xeno-free culturing. Prog Polym Sci 2014. [DOI: 10.1016/j.progpolymsci.2014.01.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
94
|
Lei Y, Jeong D, Xiao J, Schaffer DV. Developing Defined and Scalable 3D Culture Systems for Culturing Human Pluripotent Stem Cells at High Densities. Cell Mol Bioeng 2014; 7:172-183. [PMID: 25419247 DOI: 10.1007/s12195-014-0333-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) - including embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) - are very promising candidates for cell therapies, tissue engineering, high throughput pharmacology screens, and toxicity testing. These applications require large numbers of high quality cells; however, scalable production of human pluripotent stem cells and their derivatives at a high density and under well-defined conditions has been a challenge. We recently reported a simple, efficient, fully defined, scalable, and good manufacturing practice (GMP) compatible 3D culture system based on a thermoreversible hydrogel for hPSC expansion and differentiation. Here, we describe additional design rationale and characterization of this system. For instance, we have determined that culturing hPSCs as a suspension in a liquid medium can exhibit lower volumetric yields due to cell agglomeration and possible shear force-induced cell loss. By contrast, using hydrogels as 3D scaffolds for culturing hPSCs reduces aggregation and may insulate from shear forces. Additionally, hydrogel-based 3D culture systems can support efficient hPSC expansion and differentiation at a high density if compatible with hPSC biology. Finally, there are considerable opportunities for future development to further enhance hydrogel-based 3D culture systems for producing hPSCs and their progeny.
Collapse
Affiliation(s)
- Yuguo Lei
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California, 94720, USA ; Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, USA ; California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, California, 94720, USA ; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Daeun Jeong
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Jifang Xiao
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California, 94720, USA ; Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, USA ; California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, California, 94720, USA ; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, 94720, USA
| |
Collapse
|
95
|
Wilson JL, Najia MA, Saeed R, McDevitt TC. Alginate encapsulation parameters influence the differentiation of microencapsulated embryonic stem cell aggregates. Biotechnol Bioeng 2014; 111:618-31. [PMID: 24166004 PMCID: PMC4163549 DOI: 10.1002/bit.25121] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 08/26/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023]
Abstract
Pluripotent embryonic stem cells (ESCs) have tremendous potential as tools for regenerative medicine and drug discovery, yet the lack of processes to manufacture viable and homogenous cell populations of sufficient numbers limits the clinical translation of current and future cell therapies. Microencapsulation of ESCs within microbeads can shield cells from hydrodynamic shear forces found in bioreactor environments while allowing for sufficient diffusion of nutrients and oxygen through the encapsulation material. Despite initial studies examining alginate microbeads as a platform for stem cell expansion and directed differentiation, the impact of alginate encapsulation parameters on stem cell phenotype has not been thoroughly investigated. Therefore, the objective of this study was to systematically examine the effects of varying alginate compositions on microencapsulated ESC expansion and phenotype. Pre-formed aggregates of murine ESCs were encapsulated in alginate microbeads composed of a high or low ratio of guluronic to mannuronic acid residues (High G and High M, respectively), with and without a poly-L-lysine (PLL) coating, thereby providing four distinct alginate bead compositions for analysis. Encapsulation in all alginate compositions was found to delay differentiation, with encapsulation within High G alginate yielding the least differentiated cell population. The addition of a PLL coating to the High G alginate prevented cell escape from beads for up to 14 days. Furthermore, encapsulation within High M alginate promoted differentiation toward a primitive endoderm phenotype. Taken together, the findings of this study suggest that distinct ESC expansion capacities and differentiation trajectories emerge depending on the alginate composition employed, indicating that encapsulation material physical properties can be used to control stem cell fate.
Collapse
Affiliation(s)
- Jenna L Wilson
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | | | | | | |
Collapse
|
96
|
Meng X, Leslie P, Zhang Y, Dong J. Stem cells in a three-dimensional scaffold environment. SPRINGERPLUS 2014; 3:80. [PMID: 24570851 PMCID: PMC3931863 DOI: 10.1186/2193-1801-3-80] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 01/31/2014] [Indexed: 02/08/2023]
Abstract
Stem cells have emerged as important players in the generation and maintenance of many tissues. However, the accurate in vitro simulation of the native stem cell niche remains difficult due at least in part to the lack of a comprehensive definition of the critical factors of the stem cell niche based on in vivo models. Three-dimensional (3D) cell culture systems have allowed the development of useful models for investigating stem cell physiology particularly with respect to their ability to sense and generate mechanical force in response to their surrounding environment. We review the use of 3D culture systems for stem cell culture and discuss the relationship between stem cells and 3D growth matrices including the roles of the extracellular matrix, scaffolds, soluble factors, cell-cell interactions and shear stress effects within this environment. We also discuss the potential for novel methods that mimic the native stem cell niche in vitro as well as the current associated challenges.
Collapse
Affiliation(s)
- Xuan Meng
- Hospital & Institute of Hepatobiliary Surgery, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853 China ; Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599-7512 USA ; Hospital & Institute of Hepatobiliary Surgery, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853 China
| | - Patrick Leslie
- Hospital & Institute of Hepatobiliary Surgery, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853 China ; Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599-7512 USA ; Hospital & Institute of Hepatobiliary Surgery, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853 China
| | - Yanping Zhang
- Hospital & Institute of Hepatobiliary Surgery, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853 China ; Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599-7512 USA ; Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599-7512 USA
| | - Jiahong Dong
- Hospital & Institute of Hepatobiliary Surgery, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853 China
| |
Collapse
|
97
|
Hepatic differentiation of human embryonic stem cells on microcarriers. J Biotechnol 2014; 174:39-48. [PMID: 24480567 DOI: 10.1016/j.jbiotec.2014.01.025] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 12/23/2013] [Accepted: 01/14/2014] [Indexed: 01/31/2023]
Abstract
Translation of stem cell research to industrial and clinical settings mostly requires large quantities of cells, especially those involving large organs such as the liver. A scalable reactor system is desirable to ensure a reliable supply of sufficient quantities of differentiated cells. To increase the culture efficiency in bioreactor system, high surface to volume ratio needs to be achieved. We employed a microcarrier culture system for the expansion of undifferentiated human embryonic stem cells (hESCs) as well as for directed differentiation of these cells to hepatocyte-like cells. Cells in single cell suspension were attached to the bead surface in even distribution and were expanded to 1×10(6)cells/ml within 2 days of hESC culture with maintenance of the level of pluripotency markers. Directed differentiation into hepatocyte-like cells on microcarriers, both in static culture and stirred bioreactors, induced similar levels of hepatocyte-like cell differentiation as observed with cells cultured in conventional tissue culture plates. The cells expressed both immature and mature hepatocyte-lineage genes and proteins such as asialoglycoprotein receptor-1 (ASGPR-1) and albumin. Differentiated cells exhibited functional characteristics such as secretion of albumin and urea, and CYP3A4 activity could be detected. Microcarriers thus offer the potential for large-scale expansion and differentiation of hESCs induced hepatocyte-like cells in a more controllable bioreactor environment.
Collapse
|
98
|
Chen KG, Mallon BS, McKay RDG, Robey PG. Human pluripotent stem cell culture: considerations for maintenance, expansion, and therapeutics. Cell Stem Cell 2014; 14:13-26. [PMID: 24388173 PMCID: PMC3915741 DOI: 10.1016/j.stem.2013.12.005] [Citation(s) in RCA: 247] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human pluripotent stem cells (hPSCs) provide powerful resources for application in regenerative medicine and pharmaceutical development. In the past decade, various methods have been developed for large-scale hPSC culture that rely on combined use of multiple growth components, including media containing various growth factors, extracellular matrices, 3D environmental cues, and modes of multicellular association. In this Protocol Review, we dissect these growth components by comparing cell culture methods and identifying the benefits and pitfalls associated with each one. We further provide criteria, considerations, and suggestions to achieve optimal cell growth for hPSC expansion, differentiation, and use in future therapeutic applications.
Collapse
Affiliation(s)
- Kevin G Chen
- NIH Stem Cell Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Barbara S Mallon
- NIH Stem Cell Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ronald D G McKay
- The Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Pamela G Robey
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
99
|
Agarwal P, Zhao S, Bielecki P, Rao W, Choi JK, Zhao Y, Yu J, Zhang W, He X. One-step microfluidic generation of pre-hatching embryo-like core-shell microcapsules for miniaturized 3D culture of pluripotent stem cells. LAB ON A CHIP 2013; 13:4525-33. [PMID: 24113543 PMCID: PMC3848340 DOI: 10.1039/c3lc50678a] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
A novel core-shell microcapsule system is developed in this study to mimic the miniaturized 3D architecture of pre-hatching embryos with an aqueous liquid-like core of embryonic cells and a hydrogel-shell of zona pellucida. This is done by microfabricating a non-planar microfluidic flow-focusing device that enables one-step generation of microcapsules with an alginate hydrogel shell and an aqueous liquid core of cells from two aqueous fluids. Mouse embryonic stem (ES) cells encapsulated in the liquid core are found to survive well (>92%). Moreover, ~20 ES cells in the core can proliferate to form a single ES cell aggregate in each microcapsule within 7 days while at least a few hundred cells are usually needed by the commonly used hanging-drop method to form an embryoid body (EB) in each hanging drop. Quantitative RT-PCR analyses show significantly higher expression of pluripotency marker genes in the 3D aggregated ES cells compared to the cells under 2D culture. The aggregated ES cells can be efficiently differentiated into beating cardiomyocytes using a small molecule (cardiogenol C) without complex combination of multiple growth factors. Taken together, the novel 3D microfluidic and pre-hatching embryo-like microcapsule systems are of importance to facilitate in vitro culture of pluripotent stem cells for their ever-increasing use in modern cell-based medicine.
Collapse
Affiliation(s)
- Pranay Agarwal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
100
|
A fully defined and scalable 3D culture system for human pluripotent stem cell expansion and differentiation. Proc Natl Acad Sci U S A 2013; 110:E5039-48. [PMID: 24248365 DOI: 10.1073/pnas.1309408110] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Human pluripotent stem cells (hPSCs), including human embryonic stem cells and induced pluripotent stem cells, are promising for numerous biomedical applications, such as cell replacement therapies, tissue and whole-organ engineering, and high-throughput pharmacology and toxicology screening. Each of these applications requires large numbers of cells of high quality; however, the scalable expansion and differentiation of hPSCs, especially for clinical utilization, remains a challenge. We report a simple, defined, efficient, scalable, and good manufacturing practice-compatible 3D culture system for hPSC expansion and differentiation. It employs a thermoresponsive hydrogel that combines easy manipulation and completely defined conditions, free of any human- or animal-derived factors, and entailing only recombinant protein factors. Under an optimized protocol, the 3D system enables long-term, serial expansion of multiple hPSCs lines with a high expansion rate (~20-fold per 5-d passage, for a 10(72)-fold expansion over 280 d), yield (~2.0 × 10(7) cells per mL of hydrogel), and purity (~95% Oct4+), even with single-cell inoculation, all of which offer considerable advantages relative to current approaches. Moreover, the system enabled 3D directed differentiation of hPSCs into multiple lineages, including dopaminergic neuron progenitors with a yield of ~8 × 10(7) dopaminergic progenitors per mL of hydrogel and ~80-fold expansion by the end of a 15-d derivation. This versatile system may be useful at numerous scales, from basic biological investigation to clinical development.
Collapse
|