1
|
Ye S, Marsee A, van Tienderen GS, Rezaeimoghaddam M, Sheikh H, Samsom RA, de Koning EJP, Fuchs S, Verstegen MMA, van der Laan LJW, van de Vosse F, Malda J, Ito K, Spee B, Schneeberger K. Accelerated production of human epithelial organoids in a miniaturized spinning bioreactor. CELL REPORTS METHODS 2024; 4:100903. [PMID: 39561715 DOI: 10.1016/j.crmeth.2024.100903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 08/01/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024]
Abstract
Conventional static culture of organoids necessitates weekly manual passaging and results in nonhomogeneous exposure of organoids to nutrients, oxygen, and toxic metabolites. Here, we developed a miniaturized spinning bioreactor, RPMotion, specifically optimized for accelerated and cost-effective culture of epithelial organoids under homogeneous conditions. We established tissue-specific RPMotion settings and standard operating protocols for the expansion of human epithelial organoids derived from the liver, intestine, and pancreas. All organoid types proliferated faster in the bioreactor (5.2-fold, 3-fold, and 4-fold, respectively) compared to static culture while keeping their organ-specific phenotypes. We confirmed that the bioreactor is suitable for organoid establishment directly from biopsies and for long-term expansion of liver organoids. Furthermore, we showed that after accelerated expansion, liver organoids can be differentiated into hepatocyte-like cells in the RPMotion bioreactor. In conclusion, this miniaturized bioreactor enables work-, time-, and cost-efficient organoid culture, holding great promise for organoid-based fundamental and translational research and development.
Collapse
Affiliation(s)
- Shicheng Ye
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Uppsalalaan 8, Utrecht 3584 CT, the Netherlands
| | - Ary Marsee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Uppsalalaan 8, Utrecht 3584 CT, the Netherlands
| | - Gilles S van Tienderen
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Uppsalalaan 8, Utrecht 3584 CT, the Netherlands
| | - Mohammad Rezaeimoghaddam
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven 5600 MB, the Netherlands
| | - Hafsah Sheikh
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Uppsalalaan 8, Utrecht 3584 CT, the Netherlands
| | - Roos-Anne Samsom
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Uppsalalaan 8, Utrecht 3584 CT, the Netherlands
| | - Eelco J P de Koning
- Department of Internal Medicine, Leiden University Medical Center, P.O. Box 9600, Leiden 2300 RC, the Netherlands; Hubrecht Institute, KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht 3584 CT, the Netherlands
| | - Sabine Fuchs
- Department of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, Utrecht 3584 EA, the Netherlands
| | - Monique M A Verstegen
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, P.O. Box 2040, Rotterdam 3000 CA, the Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, P.O. Box 2040, Rotterdam 3000 CA, the Netherlands
| | - Frans van de Vosse
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven 5600 MB, the Netherlands
| | - Jos Malda
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Uppsalalaan 8, Utrecht 3584 CT, the Netherlands; Department of Orthopedics, University Medical Center Utrecht, Utrecht University, Utrecht 3584 CX, the Netherlands
| | - Keita Ito
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven 5600 MB, the Netherlands
| | - Bart Spee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Uppsalalaan 8, Utrecht 3584 CT, the Netherlands
| | - Kerstin Schneeberger
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Uppsalalaan 8, Utrecht 3584 CT, the Netherlands.
| |
Collapse
|
2
|
Ewere EE, White S, Mauleon R, Benkendorff K. Soil microbial communities and degradation of pesticides in greenhouse effluent through a woodchip bioreactor. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 359:124561. [PMID: 39019308 DOI: 10.1016/j.envpol.2024.124561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 07/05/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024]
Abstract
Pesticides, including insecticides and fungicides, are major contaminants in the effluent from intensive agricultural systems, such as greenhouses. Because of their constant use and persistence, some pesticides can accumulate in soil and/or run off into adjacent waterways. Microbial communities in soil can degrade some pesticides, and bioreactors with enhanced microbial communities have the potential to facilitate decontamination before the effluent is released into the environment. In this study, we sampled the soil along a gradient from immediately below greenhouses, into, through and below a bioreactor. Multi-analyte pesticide screening was undertaken along with shotgun metagenomic sequencing, to assess microbial community taxonomic profiles and metabolic pathway responses for functional analysis. Two insecticides (imidacloprid and fipronil) and nine fungicides were identified in the soil samples, with a general decrease in most pesticides with increasing distance from the greenhouses. Diversity indexes of taxonomic profiles show changes in the microbial community along the gradient. In particular, microbial communities were significantly different in the bioreactor, with lower Shannon diversity compared to immediately below the greenhouses, in the channels leading into the bioreactor and further downstream. Metabolic pathway analysis revealed significant changes in a wide range of core housekeeping genes such as protein/amino acid synthesis and lipid/fatty acid biosynthesis among the sampling sites. The result demonstrates that the composition and potential functional pathways of the microbial community shifted towards an increased tendency for phytol and contaminant degradation in the bioreactor, facilitated by high organic matter content. This highlights the potential to use enhanced microbial communities within bioreactors to reduce contamination by some pesticides in sediment receiving run-off from greenhouses.
Collapse
Affiliation(s)
- Endurance E Ewere
- National Marine Science Centre, Southern Cross University, Coffs Harbour, NSW, 2450, Australia
| | - Shane White
- National Marine Science Centre, Southern Cross University, Coffs Harbour, NSW, 2450, Australia
| | - Ramil Mauleon
- Faculty of Science and Engineering, Southern Cross University, East Lismore, NSW, 2480, Australia
| | - Kirsten Benkendorff
- National Marine Science Centre, Southern Cross University, Coffs Harbour, NSW, 2450, Australia.
| |
Collapse
|
3
|
Bandarra-Tavares H, Franchi-Mendes T, Ulpiano C, Morini S, Kaur N, Harris-Becker A, Vemuri MC, Cabral JMS, Fernandes-Platzgummer A, da Silva CL. Dual production of human mesenchymal stromal cells and derived extracellular vesicles in a dissolvable microcarrier-based stirred culture system. Cytotherapy 2024; 26:749-756. [PMID: 38506771 DOI: 10.1016/j.jcyt.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 01/29/2024] [Accepted: 03/02/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND & AIMS Cell therapies based on mesenchymal stromal cells (MSCs) have gained an increasing therapeutic interest in the context of multiple disorders. Nonetheless, this field still faces important challenges, particularly concerning suitable manufacturing platforms. Here, we aimed at establishing a scalable culture system to expand umbilical cord-derived Wharton's jelly MSC (MSC(WJ)) and their derived extracellular vesicles (EVs) by using dissolvable microcarriers combined with xeno(geneic)-free culture medium. METHODS MSC(WJ) isolated from three donors were cultured at a starting density of 1 × 106 cells per spinner flask, i.e., 2.8 × 103 cells per cm2 of dissolvable microcarrier surface area. After a 6-day expansion period of MSC(WJ), extracellular vesicles (EVs) were produced for 24 h. RESULTS Taking advantage of an intermittent agitation regimen, we observed high adhesion rates to the microcarriers (over 90% at 24 h) and achieved 15.8 ± 0.7-fold expansion after 6 days of culture. Notably, dissolution of the microcarriers was achieved through a pectinase-based solution to recover the cell product, reducing the hurdles of downstream processing. MSC identity was validated by detecting the characteristic MSC immunophenotype and by multilineage differentiation assays. Considering the growing interest in MSC-derived EVs, which are known to be mediators of the therapeutic features of MSC, this platform also was evaluated for EV production. Upon a 24-h period of conditioning, secreted EVs were isolated by ultrafiltration followed by anion-exchange chromatography and exhibited the typical cup-shaped morphology, small size distribution (162.6 ± 30.2 nm) and expressed EV markers (CD63, CD9 and syntenin-1). CONCLUSIONS Taken together, we established a time-effective and robust scalable platform that complies with clinical-grade standards for the dual production of MSC(WJ) and their derived EV.
Collapse
Affiliation(s)
- Hélder Bandarra-Tavares
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Teresa Franchi-Mendes
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Cristiana Ulpiano
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Sara Morini
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Navjot Kaur
- Cell and Gene Therapy, Thermo Fisher Scientific, Cell Biology, Frederick, Maryland, USA
| | - Abigail Harris-Becker
- Cell and Gene Therapy, Thermo Fisher Scientific, Cell Biology, Frederick, Maryland, USA
| | - Mohan C Vemuri
- Cell and Gene Therapy, Thermo Fisher Scientific, Cell Biology, Frederick, Maryland, USA
| | - Joaquim M S Cabral
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Fernandes-Platzgummer
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
4
|
Tsai CW, Chen TY, Wang JH, Young TH. Effect of Chitosan on Synovial Membrane Derived Cells and Anterior Cruciate Ligament Fibroblasts. Tissue Eng Part A 2024. [PMID: 38695112 DOI: 10.1089/ten.tea.2024.0077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024] Open
Abstract
Previously, chitosan reduces the senescence-related phenotypes in human foreskin fibroblasts through the transforming growth factor beta (TGF-β) pathway, and enhances the proliferation and migration capabilities of these cells are demonstrated. In this study, we examined whether the senescence-delaying effect of chitosan could be applied to primary knee-related fibroblasts, such as human synovial membrane derived cells (SCs) and anterior cruciate ligament fibroblasts (ACLs). These two types of cells were obtained from donors who needed ACL reconstruction or knee replacement. We found that chitosan treatment effectively reduced aging-associated β-galactosidase (SA-β-gal)-positive cells, downregulated the expression of senescence-related proteins pRB and p53, and enhanced the 5-bromo-2'-deoxyuridine (BrdU) incorporation ability of SCs and ACLs. Moreover, chitosan could make SCs secret more glycosaminoglycans (GAGs) and produce type I collagen. The ability of ACLs to close the wound was also enhanced, and the TGF-β and alpha smooth muscle actin (αSMA) protein expression decreased after chitosan treatment. In summary, chitosan not only delayed the senescence but also enhanced the functions of SCs and ACLs, which is beneficial to the application of chitosan in cell expansion in vitro and cell therapy.
Collapse
Affiliation(s)
- Ching-Wen Tsai
- Department of Biomedical Engineering , National Taiwan University, Taipei, Taiwan
- Taiwan Instrument Research Institute, National Applied Research Laboratories, Hsinchu, Taiwan
| | - Tzung-Yu Chen
- Department of Biomedical Engineering , National Taiwan University, Taipei, Taiwan
| | - Jyh-Horng Wang
- Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Tai-Horng Young
- Department of Biomedical Engineering , National Taiwan University, Taipei, Taiwan
| |
Collapse
|
5
|
Lee J, Kim H, Lim HR, Kim YS, Hoang TTT, Choi J, Jeong GJ, Kim H, Herbert R, Soltis I, Kim KR, Lee SH, Kwon Y, Lee Y, Jang YC, Yeo WH. Large-scale smart bioreactor with fully integrated wireless multivariate sensors and electronics for long-term in situ monitoring of stem cell culture. SCIENCE ADVANCES 2024; 10:eadk6714. [PMID: 38354246 PMCID: PMC10866562 DOI: 10.1126/sciadv.adk6714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 01/17/2024] [Indexed: 02/16/2024]
Abstract
Achieving large-scale, cost-effective, and reproducible manufacturing of stem cells with the existing devices is challenging. Traditional single-use cell-bag bioreactors, limited by their rigid and single-point sensors, struggle with accuracy and scalability for high-quality cell manufacturing. Here, we introduce a smart bioreactor system that enables multi-spatial sensing for real-time, wireless culture monitoring. This scalable system includes a low-profile, label-free thin-film sensor array and electronics integrated with a flexible cell bag, allowing for simultaneous assessment of culture properties such as pH, dissolved oxygen, glucose, and temperature, to receive real-time feedback for up to 30 days. The experimental results show the accurate monitoring of time-dynamic and spatial variations of stem cells and myoblast cells with adjustable carriers from a plastic dish to a 2-liter cell bag. These advances open up the broad applicability of the smart sensing system for large-scale, lower-cost, reproducible, and high-quality engineered cell manufacturing for broad clinical use.
Collapse
Affiliation(s)
- Jimin Lee
- George W. Woodruff School of Mechanical Engineering, College of Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- IEN Center for Wearable Intelligent Systems and Healthcare at the Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Hojoong Kim
- George W. Woodruff School of Mechanical Engineering, College of Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- IEN Center for Wearable Intelligent Systems and Healthcare at the Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Hyo-Ryoung Lim
- Major of Human Biocovergence, Division of Smart Healthcare, College of Information Technology and Convergence, Pukyong National University, Busan 48513, Republic of Korea
| | - Yun Soung Kim
- Biomedical Engineering and Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Thi Thai Thanh Hoang
- Department of Orthopaedics, Musculoskeletal Institute, Emory University, Atlanta, GA 30329, USA
- Atlanta VA Medical Center, Decatur, GA 30033, USA
| | - Jeongmoon Choi
- Department of Orthopaedics, Musculoskeletal Institute, Emory University, Atlanta, GA 30329, USA
- Altos Labs-San Diego Institute of Science, San Diego, CA 92121, USA
| | - Gun-Jae Jeong
- Department of Orthopaedics, Musculoskeletal Institute, Emory University, Atlanta, GA 30329, USA
- Institute of Cell and Tissue Engineering, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hodam Kim
- George W. Woodruff School of Mechanical Engineering, College of Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- IEN Center for Wearable Intelligent Systems and Healthcare at the Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Robert Herbert
- George W. Woodruff School of Mechanical Engineering, College of Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- IEN Center for Wearable Intelligent Systems and Healthcare at the Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213 USA
| | - Ira Soltis
- George W. Woodruff School of Mechanical Engineering, College of Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- IEN Center for Wearable Intelligent Systems and Healthcare at the Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Ka Ram Kim
- George W. Woodruff School of Mechanical Engineering, College of Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- IEN Center for Wearable Intelligent Systems and Healthcare at the Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Sung Hoon Lee
- IEN Center for Wearable Intelligent Systems and Healthcare at the Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA
- School of Electrical and Computer Engineering, College of Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Youngjin Kwon
- George W. Woodruff School of Mechanical Engineering, College of Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- IEN Center for Wearable Intelligent Systems and Healthcare at the Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Yunki Lee
- Department of Orthopaedics, Musculoskeletal Institute, Emory University, Atlanta, GA 30329, USA
- Atlanta VA Medical Center, Decatur, GA 30033, USA
| | - Young Charles Jang
- Department of Orthopaedics, Musculoskeletal Institute, Emory University, Atlanta, GA 30329, USA
- Atlanta VA Medical Center, Decatur, GA 30033, USA
| | - Woon-Hong Yeo
- George W. Woodruff School of Mechanical Engineering, College of Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- IEN Center for Wearable Intelligent Systems and Healthcare at the Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Institute for Materials, Institute for Robotics and Intelligent Machines, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
6
|
Terada M, Kogawa Y, Shibata Y, Kitagawa M, Kato S, Iida T, Yorimitsu T, Kato A, Matsukuma K, Maeda T, Takahashi M, Kanda GN. Robotic cell processing facility for clinical research of retinal cell therapy. SLAS Technol 2023; 28:449-459. [PMID: 39470449 DOI: 10.1016/j.slast.2023.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/14/2023] [Accepted: 10/24/2023] [Indexed: 10/30/2024]
Abstract
The consistent production of high-quality cells in cell therapy highlights the potential of automated manufacturing. Humanoid robots are a useful option for transferring technology to automate human cell cultures. This study evaluated a robotic cell-processing facility (R-CPF) for clinical research on retinal cell therapy, incorporating the versatile humanoid robot Maholo LabDroid and an All-in-One CP unit. The R-CPF platform consists of a robot area for handling cells and an operator area for the maintenance of the robot, designed with a clean airflow to ensure sterility. Monitoring the falling, floating, and adhering bacteria demonstrated that the required cleanliness and aseptic environment for cell manufacturing were satisfied. We then conducted cell manufacturing equivalent to the transplantation therapy of induced pluripotent stem cell (iPSC)-derived retinal pigment epithelial cells that met the clinical quality standards for transplantation. These results indicate that R-CPF is suitable for cell manufacturing purposes and suggest that utilizing the same robotic system in basic and clinical research can accelerate the translation of basic research findings into clinical applications.
Collapse
Affiliation(s)
- Motoki Terada
- VCCT Inc. Kobe Eye Center Building 5F, 2-1-8 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047 Japan; Kobe City Eye Hospital. 2-1-8 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047 Japan
| | - Yu Kogawa
- Technical Research Laboratory Innovation Division, DAI-DAN Co., Ltd. 390 Kitanagai, Miyoshi-cho, Iruma-gun, Saitama 354-0044 Japan
| | - Yumiko Shibata
- VCCT Inc. Kobe Eye Center Building 5F, 2-1-8 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047 Japan; Kobe City Eye Hospital. 2-1-8 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047 Japan
| | - Michinori Kitagawa
- VCCT Inc. Kobe Eye Center Building 5F, 2-1-8 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047 Japan
| | - Shinya Kato
- VCCT Inc. Kobe Eye Center Building 5F, 2-1-8 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047 Japan
| | - Tomomitsu Iida
- VCCT Inc. Kobe Eye Center Building 5F, 2-1-8 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047 Japan; Kobe City Eye Hospital. 2-1-8 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047 Japan
| | - Tsuyoshi Yorimitsu
- Technical Research Laboratory Innovation Division, DAI-DAN Co., Ltd. 390 Kitanagai, Miyoshi-cho, Iruma-gun, Saitama 354-0044 Japan
| | - Akari Kato
- Laboratory for Biologically Inspired Computing, RIKEN Center for Biosystems Dynamics Research. 6-2-3 Furuedai, Suita, Osaka 565-0874 Japan
| | - Kenji Matsukuma
- Robotic Biology Institute Inc. Telecom Center Building East Wing 1F, 2-5-10 Aomi, Koto-ku, Tokyo 135-0064 Japan
| | - Tadao Maeda
- VCCT Inc. Kobe Eye Center Building 5F, 2-1-8 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047 Japan; Kobe City Eye Hospital. 2-1-8 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047 Japan
| | - Masayo Takahashi
- VCCT Inc. Kobe Eye Center Building 5F, 2-1-8 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047 Japan; Kobe City Eye Hospital. 2-1-8 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047 Japan; Vision Care Inc. Kobe Eye Center Building 5F, 2-1-8 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047 Japan.
| | - Genki N Kanda
- Kobe City Eye Hospital. 2-1-8 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047 Japan; Robotic Biology Institute Inc. Telecom Center Building East Wing 1F, 2-5-10 Aomi, Koto-ku, Tokyo 135-0064 Japan; Laboratory for Biologically Inspired Computing, RIKEN Center for Biosystems Dynamics Research. 6-2-3 Furuedai, Suita, Osaka 565-0874 Japan; Vision Care Inc. Kobe Eye Center Building 5F, 2-1-8 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047 Japan.
| |
Collapse
|
7
|
Ando Y, Chang FC, James M, Zhou Y, Zhang M. Chitosan Scaffolds as Microcarriers for Dynamic Culture of Human Neural Stem Cells. Pharmaceutics 2023; 15:1957. [PMID: 37514142 PMCID: PMC10384976 DOI: 10.3390/pharmaceutics15071957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/03/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Human neural stem cells (hNSCs) possess remarkable potential for regenerative medicine in the treatment of presently incurable diseases. However, a key challenge lies in producing sufficient quantities of hNSCs, which is necessary for effective treatment. Dynamic culture systems are recognized as a powerful approach to producing large quantities of hNSCs required, where microcarriers play a critical role in supporting cell expansion. Nevertheless, the currently available microcarriers have limitations, including a lack of appropriate surface chemistry to promote cell adhesion, inadequate mechanical properties to protect cells from dynamic forces, and poor suitability for mass production. Here, we present the development of three-dimensional (3D) chitosan scaffolds as microcarriers for hNSC expansion under defined conditions in bioreactors. We demonstrate that chitosan scaffolds with a concentration of 4 wt% (4CS scaffolds) exhibit desirable microstructural characteristics and mechanical properties suited for hNSC expansion. Furthermore, they could also withstand degradation in dynamic conditions. The 4CS scaffold condition yields optimal metabolic activity, cell adhesion, and protein expression, enabling sustained hNSC expansion for up to three weeks in a dynamic culture. Our study introduces an effective microcarrier approach for prolonged expansion of hNSCs, which has the potential for mass production in a three-dimensional setting.
Collapse
Affiliation(s)
- Yoshiki Ando
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
- Materials Department, Medical R&D Center, Corporate R&D Group, KYOCERA Corporation, Yasu 520-2362, Shiga, Japan
| | - Fei-Chien Chang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Matthew James
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Yang Zhou
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
8
|
Peng L, Nadal C, Gautrot JE. Growth of mesenchymal stem cells at the surface of silicone, mineral and plant-based oils. Biomed Mater 2023; 18. [PMID: 36808917 DOI: 10.1088/1748-605x/acbdda] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/21/2023] [Indexed: 02/23/2023]
Abstract
Bioemulsions are attractive platforms for the expansion of adherent cells in bioreactors. Their design relies on the self-assembly of protein nanosheets at liquid-liquid interfaces, displaying strong interfacial mechanical properties and promoting integrin-mediated cell adhesion. However, most systems developed to date have focused on fluorinated oils, which are unlikely to be accepted for direct implantation of resulting cell products for regenerative medicine, and protein nanosheets self-assembly at other interfaces has not been investigated. In this report, the composition of aliphatic pro-surfactants palmitoyl chloride and sebacoyl chloride, on the assembly kinetics of poly(L-lysine) at silicone oil interfaces and characterisation of ultimate interfacial shear mechanics and viscoelasticity is presented. The impact of the resulting nanosheets on the adhesion of mesenchymal stem cells (MSCs) is investigated via immunostaining and fluorescence microscopy, demonstrating the engagement of the classic focal adhesion-actin cytoskeleton machinery. The ability of MSCs to proliferate at the corresponding interfaces is quantified. In addition, expansion of MSCs at other non-fluorinated oil interfaces, based on mineral and plant-based oils is investigated. Finally, the proof-of-concept of such non-fluorinated oil systems for the formulation of bioemulsions supporting stem cell adhesion and expansion is demonstrated.
Collapse
Affiliation(s)
- Lihui Peng
- Institute of Bioengineering, University of London, Mile End Road, London E1 4NS, United Kingdom
- School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London E1 4NS, United Kingdom
| | - Clémence Nadal
- Institute of Bioengineering, University of London, Mile End Road, London E1 4NS, United Kingdom
- School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London E1 4NS, United Kingdom
| | - Julien E Gautrot
- Institute of Bioengineering, University of London, Mile End Road, London E1 4NS, United Kingdom
- School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London E1 4NS, United Kingdom
| |
Collapse
|
9
|
Baudequin T, Wee H, Cui Z, Ye H. Towards Ready-to-Use Iron-Crosslinked Alginate Beads as Mesenchymal Stem Cell Carriers. Bioengineering (Basel) 2023; 10:bioengineering10020163. [PMID: 36829657 PMCID: PMC9951883 DOI: 10.3390/bioengineering10020163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/12/2023] [Accepted: 01/19/2023] [Indexed: 01/28/2023] Open
Abstract
Micro-carriers, thanks to high surface/volume ratio, are widely studied as mesenchymal stem cell (MSCs) in vitro substrate for proliferation at clinical rate. In particular, Ca-alginate-based biomaterials (sodium alginate crosslinked with CaCl2) are commonly investigated. However, Ca-alginate shows low bioactivity and requires functionalization, increasing labor work and costs. In contrast, films of sodium alginate crosslinked with iron chloride (Fe-alginate) have shown good bioactivity with fibroblasts, but MSCs studies are lacking. We propose a first proof-of-concept study of Fe-alginate beads supporting MSCs proliferation without functionalization. Macro- and micro-carriers were prepared (extrusion and electrospray) and we report for the first time Fe-alginate electrospraying optimization. FTIR spectra, stability with various mannuronic acids/guluronic acids (M/G) ratios and size distribution were analyzed before performing cell culture. After confirming literature results on films with human MSCs, we showed that Macro-Fe-alginate beads offered a better environment for MSCs adhesion than Ca-alginate. We concluded that Fe-alginate beads showed great potential as ready-to-use carriers.
Collapse
Affiliation(s)
- Timothée Baudequin
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DQ, UK
- Biomechanics and Bioengineering, CNRS, Centre de Recherche Royallieu, Université de Technologie de Compiègne, CS 60 319, 60203 Compiègne, France
| | - Hazel Wee
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DQ, UK
| | - Zhanfeng Cui
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DQ, UK
| | - Hua Ye
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DQ, UK
- Correspondence:
| |
Collapse
|
10
|
Chrysanthou A, Kanso H, Zhong W, Shang L, Gautrot JE. Supercharged Protein Nanosheets for Cell Expansion on Bioemulsions. ACS APPLIED MATERIALS & INTERFACES 2023; 15:2760-2770. [PMID: 36598358 PMCID: PMC9869332 DOI: 10.1021/acsami.2c20188] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/21/2022] [Indexed: 05/27/2023]
Abstract
Cell culture at liquid-liquid interfaces, for example, at the surface of oil microdroplets, is an attractive strategy to scale up adherent cell manufacturing while replacing the use of microplastics. Such a process requires the adhesion of cells at interfaces stabilized and reinforced by protein nanosheets displaying not only high elasticity but also presenting cell adhesive ligands able to bind integrin receptors. In this report, supercharged albumins are found to form strong elastic protein nanosheets when co-assembling with the co-surfactant pentafluorobenzoyl chloride (PFBC) and mediate extracellular matrix (ECM) protein adsorption and cell adhesion. The interfacial mechanical properties and elasticity of supercharged nanosheets are characterized by interfacial rheology, and behaviors are compared to those of native bovine serum albumin, human serum albumin, and α-lactalbumin. The impact of PFBC on such assembly is investigated. ECM protein adsorption to resulting supercharged nanosheets is then quantified via surface plasmon resonance and fluorescence microscopy, demonstrating that the dual role supercharged albumins are proposed to play as scaffold protein structuring liquid-liquid interfaces and substrates for the capture of ECM molecules. Finally, the adhesion and proliferation of primary human epidermal stem cells are investigated, at pinned droplets, as well as on bioemulsions stabilized by corresponding supercharged nanosheets. This study demonstrates the potential of supercharged proteins for the engineering of biointerfaces for stem cell manufacturing and draws structure-property relationships that will guide further engineering of associated systems.
Collapse
Affiliation(s)
- Alexandra Chrysanthou
- Institute
of Bioengineering, Queen Mary, University
of London, Mile End Road, London E1 4NS, U.K.
- School
of Engineering and Materials Science, Queen
Mary, University of London, Mile End Road, London E1 4NS, U.K.
| | - Hassan Kanso
- Institute
of Bioengineering, Queen Mary, University
of London, Mile End Road, London E1 4NS, U.K.
- School
of Engineering and Materials Science, Queen
Mary, University of London, Mile End Road, London E1 4NS, U.K.
| | - Wencheng Zhong
- State
Key Laboratory of Solidification Processing, School of Materials Science
and Engineering, Northwestern Polytechnical
University and Shaanxi Joint Laboratory of Graphene (NPU), Xi’an 710072, China
| | - Li Shang
- State
Key Laboratory of Solidification Processing, School of Materials Science
and Engineering, Northwestern Polytechnical
University and Shaanxi Joint Laboratory of Graphene (NPU), Xi’an 710072, China
- NPU-QMUL
Joint Research Institute of Advanced Materials and Structures (JRI-AMAS), Northwestern Polytechnical University, Xi’an 710072, China
| | - Julien E. Gautrot
- Institute
of Bioengineering, Queen Mary, University
of London, Mile End Road, London E1 4NS, U.K.
- School
of Engineering and Materials Science, Queen
Mary, University of London, Mile End Road, London E1 4NS, U.K.
| |
Collapse
|
11
|
Albayrak E, Kocabaş F. Therapeutic targeting and HSC proliferation by small molecules and biologicals. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 135:425-496. [PMID: 37061339 DOI: 10.1016/bs.apcsb.2022.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Hematopoietic stem cells (HSCs) have considerably therapeutic value on autologous and allogeneic transplantation for many malignant/non-malignant hematological diseases, especially with improvement of gene therapy. However, acquirement of limited cell dose from HSC sources is the main handicap for successful transplantation. Therefore, many strategies based on the utilization of various cytokines, interaction of stromal cells, modulation of several extrinsic and intrinsic factors have been developed to promote ex vivo functional HSC expansion with high reconstitution ability until today. Besides all these strategies, small molecules become prominent with their ease of use and various advantages when they are translated to the clinic. In the last two decades, several small molecule compounds have been investigated in pre-clinical studies and, some of them were evaluated in different stages of clinical trials for their safety and efficiencies. In this chapter, we will present an overview of HSC biology, function, regulation and also, pharmacological HSC modulation with small molecules from pre-clinical and clinical perspectives.
Collapse
|
12
|
Ganeeva I, Zmievskaya E, Valiullina A, Kudriaeva A, Miftakhova R, Rybalov A, Bulatov E. Recent Advances in the Development of Bioreactors for Manufacturing of Adoptive Cell Immunotherapies. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120808. [PMID: 36551014 PMCID: PMC9774716 DOI: 10.3390/bioengineering9120808] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Harnessing the human immune system as a foundation for therapeutic technologies capable of recognizing and killing tumor cells has been the central objective of anti-cancer immunotherapy. In recent years, there has been an increasing interest in improving the effectiveness and accessibility of this technology to make it widely applicable for adoptive cell therapies (ACTs) such as chimeric antigen receptor T (CAR-T) cells, tumor infiltrating lymphocytes (TILs), dendritic cells (DCs), natural killer (NK) cells, and many other. Automated, scalable, cost-effective, and GMP-compliant bioreactors for production of ACTs are urgently needed. The primary efforts in the field of GMP bioreactors development are focused on closed and fully automated point-of-care (POC) systems. However, their clinical and industrial application has not yet reached full potential, as there are numerous obstacles associated with delicate balancing of the complex and often unpredictable cell biology with the need for precision and full process control. Here we provide a brief overview of the existing and most advanced systems for ACT manufacturing, including cell culture bags, G-Rex flasks, and bioreactors (rocking motion, stirred-flask, stirred-tank, hollow-fiber), as well as semi- and fully-automated closed bioreactor systems.
Collapse
Affiliation(s)
- Irina Ganeeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Ekaterina Zmievskaya
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Aygul Valiullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Anna Kudriaeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Regina Miftakhova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | | | - Emil Bulatov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
- Correspondence:
| |
Collapse
|
13
|
Facile suspension culture protocol of the liver biliary organoids. Biodes Manuf 2022. [DOI: 10.1007/s42242-022-00213-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
14
|
Fuentes P, Torres MJ, Arancibia R, Aulestia F, Vergara M, Carrión F, Osses N, Altamirano C. Dynamic Culture of Mesenchymal Stromal/Stem Cell Spheroids and Secretion of Paracrine Factors. Front Bioeng Biotechnol 2022; 10:916229. [PMID: 36046670 PMCID: PMC9421039 DOI: 10.3389/fbioe.2022.916229] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years, conditioned medium (CM) obtained from the culture of mesenchymal stromal/stem cells (MSCs) has been shown to effectively promote tissue repair and modulate the immune response in vitro and in different animal models, with potential for application in regenerative medicine. Using CM offers multiple advantages over the implantation of MSCs themselves: 1) simpler storage, transport, and preservation requirements, 2) avoidance of the inherent risks of cell transplantation, and 3) potential application as a ready-to-go biologic product. For these reasons, a large amount of MSCs research has focused on the characterization of the obtained CM, including soluble trophic factors and vesicles, preconditioning strategies for enhancing paracrine secretion, such as hypoxia, a three-dimensional (3D) environment, and biochemical stimuli, and potential clinical applications. In vitro preconditioning strategies can increase the viability, proliferation, and paracrine properties of MSCs and therefore improve the therapeutic potential of the cells and their derived products. Specifically, dynamic cultivation conditions, such as fluid flow and 3D aggregate culture, substantially impact cellular behaviour. Increased levels of growth factors and cytokines were observed in 3D cultures of MSC grown on orbital or rotatory shaking platforms, in stirred systems, such as spinner flasks or stirred tank reactors, and in microgravity bioreactors. However, only a few studies have established dynamic culture conditions and protocols for 3D aggregate cultivation of MSCs as a scalable and reproducible strategy for CM production. This review summarizes significant advances into the upstream processing, mainly the dynamic generation and cultivation of MSC aggregates, for de CM manufacture and focuses on the standardization of the soluble factor production.
Collapse
Affiliation(s)
- Paloma Fuentes
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - María José Torres
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Rodrigo Arancibia
- Cellus Medicina Regenerativa S.A., Santiago, Chile
- Cellus Biomédica, Parque Tecnológico de León, León, Spain
| | - Francisco Aulestia
- Cellus Medicina Regenerativa S.A., Santiago, Chile
- Cellus Biomédica, Parque Tecnológico de León, León, Spain
| | - Mauricio Vergara
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Flavio Carrión
- Cellus Medicina Regenerativa S.A., Santiago, Chile
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile
| | - Nelson Osses
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Claudia Altamirano
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
- CREAS, Centro Regional de Estudios en Alimentos Saludables, Valparaíso, Chile
- *Correspondence: Claudia Altamirano,
| |
Collapse
|
15
|
Kanda GN, Tsuzuki T, Terada M, Sakai N, Motozawa N, Masuda T, Nishida M, Watanabe CT, Higashi T, Horiguchi SA, Kudo T, Kamei M, Sunagawa GA, Matsukuma K, Sakurada T, Ozawa Y, Takahashi M, Takahashi K, Natsume T. Robotic search for optimal cell culture in regenerative medicine. eLife 2022; 11:77007. [PMID: 35762203 PMCID: PMC9239686 DOI: 10.7554/elife.77007] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/17/2022] [Indexed: 12/25/2022] Open
Abstract
Induced differentiation is one of the most experience- and skill-dependent experimental processes in regenerative medicine, and establishing optimal conditions often takes years. We developed a robotic AI system with a batch Bayesian optimization algorithm that autonomously induces the differentiation of induced pluripotent stem cell-derived retinal pigment epithelial (iPSC-RPE) cells. From 200 million possible parameter combinations, the system performed cell culture in 143 different conditions in 111 days, resulting in 88% better iPSC-RPE production than that obtained by the pre-optimized culture in terms of the pigmentation scores. Our work demonstrates that the use of autonomous robotic AI systems drastically accelerates systematic and unbiased exploration of experimental search space, suggesting immense use in medicine and research.
Collapse
Affiliation(s)
- Genki N Kanda
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Laboratory for Biologically Inspired Computing, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan.,Robotic Biology Institute Inc., Tokyo, Japan
| | | | - Motoki Terada
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,VCCT Inc., Kobe, Japan
| | - Noriko Sakai
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,VCCT Inc., Kobe, Japan
| | - Naohiro Motozawa
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Tomohiro Masuda
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,VCCT Inc., Kobe, Japan
| | - Mitsuhiro Nishida
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,VCCT Inc., Kobe, Japan
| | | | | | | | - Taku Kudo
- Robotic Biology Institute Inc., Tokyo, Japan
| | | | - Genshiro A Sunagawa
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Laboratory for Molecular Biology of Aging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | | | | | | | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,VCCT Inc., Kobe, Japan.,Vision Care Inc., Kobe, Japan
| | - Koichi Takahashi
- Laboratory for Biologically Inspired Computing, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan.,Graduate School of Media and Governance, Keio University, Fujisawa, Japan.,Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Tohru Natsume
- Robotic Biology Institute Inc., Tokyo, Japan.,Department of Life Science and Biotechnology, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| |
Collapse
|
16
|
Kong D, Peng L, Bosch-Fortea M, Chrysanthou A, Alexis CVM, Matellan C, Zarbakhsh A, Mastroianni G, del Rio Hernandez A, Gautrot JE. Impact of the multiscale viscoelasticity of quasi-2D self-assembled protein networks on stem cell expansion at liquid interfaces. Biomaterials 2022; 284:121494. [DOI: 10.1016/j.biomaterials.2022.121494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 11/02/2022]
|
17
|
Itoh TD, Horinouchi T, Uchida H, Takahashi K, Ozaki H. Optimal Scheduling for Laboratory Automation of Life Science Experiments with Time Constraints. SLAS Technol 2021; 26:650-659. [PMID: 34167357 PMCID: PMC8641030 DOI: 10.1177/24726303211021790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/05/2021] [Accepted: 05/11/2021] [Indexed: 11/15/2022]
Abstract
In automated laboratories consisting of multiple different types of instruments, scheduling algorithms are useful for determining the optimal allocations of instruments to minimize the time required to complete experimental procedures. However, previous studies on scheduling algorithms for laboratory automation have not emphasized the time constraints by mutual boundaries (TCMBs) among operations, which is important in procedures involving live cells or unstable biomolecules. Here, we define the "scheduling for laboratory automation in biology" (S-LAB) problem as a scheduling problem for automated laboratories in which operations with TCMBs are performed by multiple different instruments. We formulate an S-LAB problem as a mixed-integer programming (MIP) problem and propose a scheduling method using the branch-and-bound algorithm. Simulations show that our method can find the optimal schedules of S-LAB problems that minimize overall execution time while satisfying the TCMBs. Furthermore, we propose the use of our scheduling method for the simulation-based design of job definitions and laboratory configurations.
Collapse
Affiliation(s)
- Takeshi D. Itoh
- Mathematical Informatics Laboratory, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara, Japan
- Anchorage Works, Inc., Kobe, Hyogo, Japan
| | - Takaaki Horinouchi
- Laboratory for Biologically Inspired Computing, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Laboratory for Multiscale Biosystem Dynamics, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | | | - Koichi Takahashi
- Laboratory for Biologically Inspired Computing, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Haruka Ozaki
- Bioinformatics Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Center for Artificial Intelligence Research, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
18
|
Peng L, Gautrot JE. Long term expansion profile of mesenchymal stromal cells at protein nanosheet-stabilised bioemulsions for next generation cell culture microcarriers. Mater Today Bio 2021; 12:100159. [PMID: 34841241 PMCID: PMC8605361 DOI: 10.1016/j.mtbio.2021.100159] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/10/2021] [Accepted: 11/13/2021] [Indexed: 12/27/2022] Open
Abstract
Tremendous progress in the identification, isolation and expansion of stem cells has allowed their application in regenerative medicine and tissue engineering, and their use as advanced in vitro models. As a result, stem cell manufacturing increasingly requires scale up, parallelisation and automation. However, solid substrates currently used for the culture of adherent cells are poorly adapted for such applications, owing to their difficult processing from cell products, relatively high costs and their typical reliance on difficult to recycle plastics and microplastics. In this work, we show that bioemulsions formed of microdroplets stabilised by protein nanosheets displaying strong interfacial mechanics are well-suited for the scale up of adherent stem cells such as mesenchymal stromal cells (MSCs). We demonstrate that, over multiple passages (up to passage 10), MSCs retain comparable phenotypes when cultured on such bioemulsions, solid microcarriers (Synthemax II) and classic 2D tissue culture polystyrene. Phenotyping (cell proliferation, morphometry, flow cytometry and differentiation assays) of MSCs cultured for multiple passages on these systems indicate that, although stemness is lost at late passages when cultured on these different substrates, stem cell phenotypes remained comparable between different culture conditions, at any given passage. Hence our study validates the use of bioemulsions for the long term expansion of adherent stem cells and paves the way to the design of novel 3D bioreactors based on microdroplet microcarriers.
Collapse
Affiliation(s)
- Lihui Peng
- Institute of Bioengineering and, UK.,School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| | - Julien E Gautrot
- Institute of Bioengineering and, UK.,School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| |
Collapse
|
19
|
Atkinson SP. A preview of selected articles-September 2021. Stem Cells Transl Med 2021; 10:1249-1252. [PMID: 34420262 PMCID: PMC8380439 DOI: 10.1002/sctm.21-0253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 07/03/2021] [Indexed: 01/02/2023] Open
|
20
|
Van Beylen K, Papantoniou I, Aerts JM. Microcarrier Screening and Evaluation for Dynamic Expansion of Human Periosteum-Derived Progenitor Cells in a Xenogeneic Free Medium. Front Bioeng Biotechnol 2021; 9:624890. [PMID: 34109163 PMCID: PMC8181150 DOI: 10.3389/fbioe.2021.624890] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 04/27/2021] [Indexed: 11/13/2022] Open
Abstract
An increasing need toward a more efficient expansion of adherent progenitor cell types arises with the advancements of cell therapy. The use of a dynamic expansion instead of a static planar expansion could be one way to tackle the challenges of expanding adherent cells at a large scale. Microcarriers are often reported as a biomaterial for culturing cells in suspension. However, the type of microcarrier has an effect on the cell expansion. In order to find an efficient expansion process for a specific adherent progenitor cell type, it is important to investigate the effect of the type of microcarrier on the cell expansion. Human periosteum-derived progenitor cells are extensively used in skeletal tissue engineering for the regeneration of bone defects. Therefore, we evaluated the use of different microcarriers on human periosteum-derived progenitor cells. In order to assess the potency, identity and viability of these cells after being cultured in the spinner flasks, this study performed several in vitro and in vivo analyses. The novelty of this work lies in the combination of screening different microcarriers for human periosteum-derived progenitor cells with in vivo assessments of the cells’ potency using the microcarrier that was selected as the most promising one. The results showed that expanding human periosteum-derived progenitor cells in spinner flasks using xeno-free medium and Star-Plus microcarriers, does not affect the potency, identity or viability of the cells. The potency of the cells was assured with an in vivo evaluation, where bone formation was achieved. In summary, this expansion method has the potential to be used for large scale cell expansion with clinical relevance.
Collapse
Affiliation(s)
- Kathleen Van Beylen
- M3-BIORES: Measure, Model, and Manage Bioresponses, Division Animal and Human Health Engineering, Department of Biosystems, KU Leuven, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Ioannis Papantoniou
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Centre, Leuven, Belgium.,Foundation for Research and Technology - Hellas (FORTH), Institute of Chemical Engineering Sciences, Patras, Greece
| | - Jean-Marie Aerts
- M3-BIORES: Measure, Model, and Manage Bioresponses, Division Animal and Human Health Engineering, Department of Biosystems, KU Leuven, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
21
|
Chen S, Sato Y, Tada Y, Suzuki Y, Takahashi R, Okanojo M, Nakashima K. Facile bead-to-bead cell-transfer method for serial subculture and large-scale expansion of human mesenchymal stem cells in bioreactors. Stem Cells Transl Med 2021; 10:1329-1342. [PMID: 34008349 PMCID: PMC8380445 DOI: 10.1002/sctm.20-0501] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/29/2021] [Accepted: 04/21/2021] [Indexed: 12/13/2022] Open
Abstract
The conventional planar culture of adherent cells is inefficient for large‐scale manufacturing of cell and gene therapy products. We developed a facile and efficient bead‐to‐bead cell‐transfer method for serial subculture and large‐scale expansion of human mesenchymal stem cells (hMSCs) with microcarriers in bioreactors. We first compared culture medium with and without nucleosides and found the former maintained the expression of surface markers of hMSCs during their prolonged culture and enabled faster cell proliferation. Subsequently, we developed our bead‐to‐bead cell transfer method to subculture hMSCs and found that intermittent agitation after adding fresh microcarriers to cell‐populated microcarriers could promote spontaneous cell migration to fresh microcarriers, reduce microcarrier aggregation, and improve cell yield. This method enabled serial subculture of hMSCs in spinner flasks from passage 4 to passage 9 without using proteolytic enzymes, which showed faster cell proliferation than the serial planar cultures undergoing multiple enzyme treatment. Finally, we used the medium containing nucleosides and our bead‐to‐bead cell transfer method for cell culture scale‐up from 4‐ to 50‐L cultures in single‐use bioreactors. We achieved a 242‐fold increase in the number of cells to 1.45 × 1010 after 27‐day culture and found that the cells harvested from the bioreactors maintained proliferation ability, expression of their surface markers, tri‐lineage differentiation potential and immunomodulatory property. This study shows the promotive effect of nucleosides on hMSC expansion and the potential of using our bead‐to‐bead transfer method for larger‐scale manufacturing of hMSCs for cell therapy.
Collapse
Affiliation(s)
- Shangwu Chen
- Regenerative Medicine Business Sector, Showa Denko Materials Co, Ltd, Yokohama-shi, Kanagawa, Japan
| | - Yushi Sato
- Regenerative Medicine Business Sector, Showa Denko Materials Co, Ltd, Yokohama-shi, Kanagawa, Japan
| | - Yasuhiko Tada
- Regenerative Medicine Business Sector, Showa Denko Materials Co, Ltd, Yokohama-shi, Kanagawa, Japan
| | - Yuma Suzuki
- Regenerative Medicine Business Sector, Showa Denko Materials Co, Ltd, Yokohama-shi, Kanagawa, Japan
| | - Ryosuke Takahashi
- Regenerative Medicine Business Sector, Showa Denko Materials Co, Ltd, Yokohama-shi, Kanagawa, Japan
| | - Masahiro Okanojo
- Regenerative Medicine Business Sector, Showa Denko Materials Co, Ltd, Yokohama-shi, Kanagawa, Japan
| | - Katsuhiko Nakashima
- Regenerative Medicine Business Sector, Showa Denko Materials Co, Ltd, Yokohama-shi, Kanagawa, Japan
| |
Collapse
|
22
|
Ornelas-González A, González-González M, Rito-Palomares M. Microcarrier-based stem cell bioprocessing: GMP-grade culture challenges and future trends for regenerative medicine. Crit Rev Biotechnol 2021; 41:1081-1095. [PMID: 33730936 DOI: 10.1080/07388551.2021.1898328] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Recently, stem cell-based therapies have been proposed as an alternative for the treatment of many diseases. Stem cells (SCs) are well known for their capacity to preserve themselves, proliferate, and differentiate into multiple lineages. These characteristics allow stem cells to be a viable option for the treatment of diverse diseases. Traditional methodologies based on 2-dimensional culture techniques (T-flasks and Petri dishes) are simple and well standardized; however, they present disadvantages that limit the production of the cell yield required for regenerative medicine applications. Lately, microcarrier (MC)-based culture techniques have emerged as an attractive platform for expanding stem cells in suspension systems. Although the use of stem cell expansion on MCs has recently shown significant increase, their implementation for medical purposes is been hampered by bottlenecks in upstream and downstream processing. Therefore, there is an urgent need in the development of bioprocesses that simplify stem cell cultures under xeno-free conditions and detachment from MCs without diminishing their pluripotency and viability. A critical analysis of the factors that impact the up and downstream bioprocessing on MC-based stem cell cultures is presented in this review. This analysis aims to raise the awareness of the current drawbacks that limit MC-based stem cell bioprocessing in regenerative medicine and propose alternatives to overcome them.
Collapse
Affiliation(s)
| | | | - Marco Rito-Palomares
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Mexico
| |
Collapse
|
23
|
Ochiai K, Motozawa N, Terada M, Horinouchi T, Masuda T, Kudo T, Kamei M, Tsujikawa A, Matsukuma K, Natsume T, Kanda GN, Takahashi M, Takahashi K. A Variable Scheduling Maintenance Culture Platform for Mammalian Cells. SLAS Technol 2020; 26:209-217. [PMID: 33269985 PMCID: PMC7985857 DOI: 10.1177/2472630320972109] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Cell culturing is a basic experimental technique in cell biology and medical science. However, culturing high-quality cells with a high degree of reproducibility relies heavily on expert skills and tacit knowledge, and it is not straightforward to scale the production process due to the education bottleneck. Although many automated culture systems have been developed and a few have succeeded in mass production environments, very few robots are permissive of frequent protocol changes, which are often required in basic research environments. LabDroid is a general-purpose humanoid robot with two arms that performs experiments using the same tools as humans. Combining our newly developed AI software with LabDroid, we developed a variable scheduling system that continuously produces subcultures of cell lines without human intervention. The system periodically observes the cells on plates with a microscope, predicts the cell growth curve by processing cell images, and decides the best times for passage. We have succeeded in developing a system that maintains the cultures of two HEK293A cell plates with no human intervention for 192 h.
Collapse
Affiliation(s)
- Koji Ochiai
- Laboratory for Biologically Inspired Computing, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Naohiro Motozawa
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe, Hyogo, Japan.,Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Motoki Terada
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe, Hyogo, Japan
| | - Takaaki Horinouchi
- Laboratory for Biologically Inspired Computing, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan.,Laboratory for Multiscale Biosystem Dynamics, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Tomohiro Masuda
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe, Hyogo, Japan
| | - Taku Kudo
- Robotic Biology Institute Inc., Koto-ku, Tokyo, Japan
| | | | - Akitaka Tsujikawa
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | | | - Tohru Natsume
- Robotic Biology Institute Inc., Koto-ku, Tokyo, Japan.,Department of Life Science and Biotechnology, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Koto-ku, Tokyo, Japan
| | - Genki N Kanda
- Laboratory for Biologically Inspired Computing, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan.,Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe, Hyogo, Japan.,Robotic Biology Institute Inc., Koto-ku, Tokyo, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe, Hyogo, Japan.,VisionCare Inc., Chuo-ku, Kobe, Hyogo, Japan
| | - Koichi Takahashi
- Laboratory for Biologically Inspired Computing, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| |
Collapse
|
24
|
Silva Couto P, Rotondi M, Bersenev A, Hewitt C, Nienow A, Verter F, Rafiq Q. Expansion of human mesenchymal stem/stromal cells (hMSCs) in bioreactors using microcarriers: lessons learnt and what the future holds. Biotechnol Adv 2020; 45:107636. [DOI: 10.1016/j.biotechadv.2020.107636] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 08/01/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
|
25
|
Hernigou P. Bone marrow in orthopaedics (part II): a three hundred and seventy million-year saga from the Devonian to the coronavirus disease 2019 pandemic-osteonecrosis; transplantation; "human chimera"; stem cells, bioreactors, and coronavirus disease. INTERNATIONAL ORTHOPAEDICS 2020; 44:2787-2805. [PMID: 33037446 PMCID: PMC7546933 DOI: 10.1007/s00264-020-04843-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 09/29/2020] [Indexed: 01/06/2023]
Abstract
PURPOSE Three hundred seventy million years ago, bone marrow appeared in skeleton of a fish. More than one hundred years ago, the concept of bone marrow transplantation was proposed to treat human diseases. During the last five decades, this concept became a reality first in hematology and later for orthopaedic diseases. MATERIAL AND METHODS These advances were possible due to the comprehension of the three major components of bone marrow: the fat part, the haematologic part, and the stroma part. Each part has a different history, but the three parts are linked in physiology as in history. RESULTS During many centuries, bone marrow was considered just as food; however, one hundred years ago, the concept of bone marrow transplantation to treat humans was proposed by the French physician Brown-Séquard. During the last five decades, this concept became a reality first in haematology and later for orthopaedic diseases. Transferring what was known from experimental animal models to humans was met with many challenges, the atomic bomb research, and many deaths. Yet through the recognition and subsequent understanding of fundamental processes, medical resiliency, and the determination of a few pioneers, local bone marrow transplantation in orthopaedic surgery became a therapeutic option first for a limited number of diseases and patients. Over the last two decades, mesenchymal stromal cells (MSCs) have been the focus of intense research by acadaemia and industry due to their unique features. MSCs can be easily isolated and expanded through in vitro culture by taking full advantage of their self-renewing capacity. In addition, MSCs exert immunomodulatory effects and can be differentiated into various lineages, which makes them highly attractive for clinical applications in cell-based therapies. CONCLUSION In this review, we attempted to provide a historical overview of bone marrow history, MSC discovery, characterization, and the first clinical studies conducted.
Collapse
Affiliation(s)
- Philippe Hernigou
- Orthopedic Department Henri Mondor Hospital, University Paris East, Paris, France.
| |
Collapse
|
26
|
Pigeot S, Bourgine PE, Claude J, Scotti C, Papadimitropoulos A, Todorov A, Epple C, Peretti GM, Martin I. Orthotopic Bone Formation by Streamlined Engineering and Devitalization of Human Hypertrophic Cartilage. Int J Mol Sci 2020; 21:ijms21197233. [PMID: 33008121 PMCID: PMC7582540 DOI: 10.3390/ijms21197233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/22/2020] [Accepted: 09/26/2020] [Indexed: 12/25/2022] Open
Abstract
Most bones of the human body form and heal through endochondral ossification, whereby hypertrophic cartilage (HyC) is formed and subsequently remodeled into bone. We previously demonstrated that HyC can be engineered from human mesenchymal stromal cells (hMSC), and subsequently devitalized by apoptosis induction. The resulting extracellular matrix (ECM) tissue retained osteoinductive properties, leading to ectopic bone formation. In this study, we aimed at engineering and devitalizing upscaled quantities of HyC ECM within a perfusion bioreactor, followed by in vivo assessment in an orthotopic bone repair model. We hypothesized that the devitalized HyC ECM would outperform a clinical product currently used for bone reconstructive surgery. Human MSC were genetically engineered with a gene cassette enabling apoptosis induction upon addition of an adjuvant. Engineered hMSC were seeded, differentiated, and devitalized within a perfusion bioreactor. The resulting HyC ECM was subsequently implanted in a 10-mm rabbit calvarial defect model, with processed human bone (Maxgraft®) as control. Human MSC cultured in the perfusion bioreactor generated a homogenous HyC ECM and were efficiently induced towards apoptosis. Following six weeks of in vivo implantation, microcomputed tomography and histological analyses of the defects revealed an increased bone formation in the defects filled with HyC ECM as compared to Maxgraft®. This work demonstrates the suitability of engineered devitalized HyC ECM as a bone substitute material, with a performance superior to a state-of-the-art commercial graft. Streamlined generation of the devitalized tissue transplant within a perfusion bioreactor is relevant towards standardized and automated manufacturing of a clinical product.
Collapse
Affiliation(s)
- Sébastien Pigeot
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (S.P.); (P.E.B.); (A.P.); (A.T.)
| | - Paul Emile Bourgine
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (S.P.); (P.E.B.); (A.P.); (A.T.)
| | - Jaquiery Claude
- Department of Surgery, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (J.C.); (C.E.)
| | - Celeste Scotti
- Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland;
- IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy;
| | - Adam Papadimitropoulos
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (S.P.); (P.E.B.); (A.P.); (A.T.)
| | - Atanas Todorov
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (S.P.); (P.E.B.); (A.P.); (A.T.)
| | - Christian Epple
- Department of Surgery, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (J.C.); (C.E.)
| | - Giuseppe M. Peretti
- IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy;
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (S.P.); (P.E.B.); (A.P.); (A.T.)
- Department of Surgery, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (J.C.); (C.E.)
- Correspondence:
| |
Collapse
|
27
|
Rohani L, Borys BS, Razian G, Naghsh P, Liu S, Johnson AA, Machiraju P, Holland H, Lewis IA, Groves RA, Toms D, Gordon PMK, Li JW, So T, Dang T, Kallos MS, Rancourt DE. Stirred suspension bioreactors maintain naïve pluripotency of human pluripotent stem cells. Commun Biol 2020; 3:492. [PMID: 32895477 PMCID: PMC7476926 DOI: 10.1038/s42003-020-01218-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 08/03/2020] [Indexed: 11/11/2022] Open
Abstract
Due to their ability to standardize key physiological parameters, stirred suspension bioreactors can potentially scale the production of quality-controlled pluripotent stem cells (PSCs) for cell therapy application. Because of differences in bioreactor expansion efficiency between mouse (m) and human (h) PSCs, we investigated if conversion of hPSCs, from the conventional "primed" pluripotent state towards the "naïve" state prevalent in mPSCs, could be used to enhance hPSC production. Through transcriptomic enrichment of mechano-sensing signaling, the expression of epigenetic regulators, metabolomics, and cell-surface protein marker analyses, we show that the stirred suspension bioreactor environment helps maintain a naïve-like pluripotent state. Our research corroborates that converting hPSCs towards a naïve state enhances hPSC manufacturing and indicates a potentially important role for the stirred suspension bioreactor's mechanical environment in maintaining naïve-like pluripotency.
Collapse
Affiliation(s)
- Leili Rohani
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Breanna S Borys
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Golsa Razian
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Pooyan Naghsh
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Shiying Liu
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Pranav Machiraju
- Department of Paediatrics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Heidrun Holland
- Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, Leipzig, Germany
| | - Ian A Lewis
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Ryan A Groves
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Derek Toms
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul M K Gordon
- CSM Center for Health Genomic and Informatics, University of Calgary, Calgary, AB, Canada
| | - Joyce W Li
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Tania So
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Tiffany Dang
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Michael S Kallos
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Derrick E Rancourt
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
28
|
de Sá da Silva J, Severino P, Wodewotzky TI, Covas DT, Swiech K, Cavalheiro Marti L, Torres Suazo CA. Mesenchymal stromal cells maintain the major quality attributes when expanded in different bioreactor systems. Biochem Eng J 2020. [DOI: 10.1016/j.bej.2020.107693] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
29
|
García-Fernández C, López-Fernández A, Borrós S, Lecina M, Vives J. Strategies for large-scale expansion of clinical-grade human multipotent mesenchymal stromal cells. Biochem Eng J 2020. [DOI: 10.1016/j.bej.2020.107601] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
30
|
Schneeberger K, Sánchez‐Romero N, Ye S, van Steenbeek FG, Oosterhoff LA, Pla Palacin I, Chen C, van Wolferen ME, van Tienderen G, Lieshout R, Colemonts‐Vroninks H, Schene I, Hoekstra R, Verstegen MM, van der Laan LJ, Penning LC, Fuchs SA, Clevers H, De Kock J, Baptista PM, Spee B. Large-Scale Production of LGR5-Positive Bipotential Human Liver Stem Cells. Hepatology 2020; 72:257-270. [PMID: 31715015 PMCID: PMC7496924 DOI: 10.1002/hep.31037] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 11/07/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIMS The gap between patients on transplant waiting lists and available donor organs is steadily increasing. Human organoids derived from leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5)-positive adult stem cells represent an exciting new cell source for liver regeneration; however, culturing large numbers of organoids with current protocols is tedious and the level of hepatic differentiation is limited. APPROACH AND RESULTS Here, we established a method for the expansion of large quantities of human liver organoids in spinner flasks. Due to improved oxygenation in the spinner flasks, organoids rapidly proliferated and reached an average 40-fold cell expansion after 2 weeks, compared with 6-fold expansion in static cultures. The organoids repopulated decellularized liver discs and formed liver-like tissue. After differentiation in spinner flasks, mature hepatocyte markers were highly up-regulated compared with static organoid cultures, and cytochrome p450 activity reached levels equivalent to hepatocytes. CONCLUSIONS We established a highly efficient method for culturing large numbers of LGR5-positive stem cells in the form of organoids, which paves the way for the application of organoids for tissue engineering and liver transplantation.
Collapse
Affiliation(s)
- Kerstin Schneeberger
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | | | - Shicheng Ye
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Frank G. van Steenbeek
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Loes A. Oosterhoff
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Iris Pla Palacin
- Instituto de Investigación Sanitaria Aragón (IIS Aragón)ZaragozaSpain
| | - Chen Chen
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands,Hubrecht InstituteRoyal Netherlands Academy of Arts and Sciences and University Medical Center UtrechtUtrechtthe Netherlands
| | - Monique E. van Wolferen
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Gilles van Tienderen
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Ruby Lieshout
- Department of SurgeryErasmus MC‐University Medical CenterRotterdamthe Netherlands
| | - Haaike Colemonts‐Vroninks
- Department of In Vitro Toxicology and Dermato‐cosmetologyFaculty of Medicine and PharmacyVrije Universiteit BrusselBrusselsBelgium
| | - Imre Schene
- Division of Pediatric GastroenterologyWilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtthe Netherlands
| | - Ruurdtje Hoekstra
- Tytgat Institute for Liver and Intestinal ResearchGastroenterology and MetabolismAcademic Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands,Surgical LaboratoryDepartment of SurgeryAcademic Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands
| | | | | | - Louis C. Penning
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Sabine A. Fuchs
- Division of Pediatric GastroenterologyWilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtthe Netherlands
| | - Hans Clevers
- Hubrecht InstituteRoyal Netherlands Academy of Arts and Sciences and University Medical Center UtrechtUtrechtthe Netherlands,Cancer Genomics NetherlandsUniversity Medical Center UtrechtUtrechtthe Netherlands,Princess Máxima CenterUtrechtthe Netherlands
| | - Joery De Kock
- Department of In Vitro Toxicology and Dermato‐cosmetologyFaculty of Medicine and PharmacyVrije Universiteit BrusselBrusselsBelgium
| | - Pedro M. Baptista
- Instituto de Investigación Sanitaria Aragón (IIS Aragón)ZaragozaSpain,Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas (CIBERehd)MadridSpain,Fundación ARAIDZaragozaSpain,Instituto de Investigación Sanitaria de la Fundación Jiménez DíazMadridSpain,Department of Biomedical and Aerospace EngineeringUniversidad Carlos III de MadridMadridSpain
| | - Bart Spee
- Department of Clinical Sciences of Companion AnimalsFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| |
Collapse
|
31
|
Tsai AC, Jeske R, Chen X, Yuan X, Li Y. Influence of Microenvironment on Mesenchymal Stem Cell Therapeutic Potency: From Planar Culture to Microcarriers. Front Bioeng Biotechnol 2020; 8:640. [PMID: 32671039 PMCID: PMC7327111 DOI: 10.3389/fbioe.2020.00640] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) are a promising candidate in cell therapy as they exhibit multilineage differentiation, homing to the site of injury, and secretion of trophic factors that facilitate tissue healing and/or modulate immune response. As a result, hMSC-derived products have attracted growing interests in preclinical and clinical studies. The development of hMSC culture platforms for large-scale biomanufacturing is necessary to meet the requirements for late-phase clinical trials and future commercialization. Microcarriers in stirred-tank bioreactors have been widely utilized in large-scale expansion of hMSCs for translational applications because of a high surface-to-volume ratio compared to conventional 2D planar culture. However, recent studies have demonstrated that microcarrier-expanded hMSCs differ from dish- or flask-expanded cells in size, morphology, proliferation, viability, surface markers, gene expression, differentiation potential, and secretome profile which may lead to altered therapeutic potency. Therefore, understanding the bioprocessing parameters that influence hMSC therapeutic efficacy is essential for the optimization of microcarrier-based bioreactor system to maximize hMSC quantity without sacrificing quality. In this review, biomanufacturing parameters encountered in planar culture and microcarrier-based bioreactor culture of hMSCs are compared and discussed with specific focus on cell-adhesion surface (e.g., discontinuous surface, underlying curvature, microcarrier stiffness, porosity, surface roughness, coating, and charge) and the dynamic microenvironment in bioreactor culture (e.g., oxygen and nutrients, shear stress, particle collision, and aggregation). The influence of dynamic culture in bioreactors on hMSC properties is also reviewed in order to establish connection between bioprocessing and stem cell function. This review addresses fundamental principles and concepts for future design of biomanufacturing systems for hMSC-based therapy.
Collapse
Affiliation(s)
- Ang-Chen Tsai
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Richard Jeske
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Xingchi Chen
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Xuegang Yuan
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
32
|
Mehrian M, Lambrechts T, Papantoniou I, Geris L. Computational Modeling of Human Mesenchymal Stromal Cell Proliferation and Extra-Cellular Matrix Production in 3D Porous Scaffolds in a Perfusion Bioreactor: The Effect of Growth Factors. Front Bioeng Biotechnol 2020; 8:376. [PMID: 32411692 PMCID: PMC7201129 DOI: 10.3389/fbioe.2020.00376] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 04/06/2020] [Indexed: 01/20/2023] Open
Abstract
Stem cell expansion on 3D porous scaffolds cultured in bioreactor systems has been shown to be beneficial for maintenance of the original cell functionality in tissue engineering strategies (TE). However, the production of extracellular matrix (ECM) makes harvesting the progenitor cell population from 3D scaffolds a challenge. Medium composition plays a role in stimulating cell proliferation over extracellular matrix (ECM) production. In this regard, a computational model describing tissue growth inside 3D scaffolds can be a great tool in designing optimal experimental conditions. In this study, a computational model describing cell and ECM growth in a perfusion bioreactor is developed, including a description of the effect of a (generic) growth factor on the biological processes taking place inside the 3D scaffold. In the model, the speed of cell and ECM growth depends on the flow-induced shear stress, curvature and the concentrations of oxygen, glucose, lactate, and growth factor. The effect of the simulated growth factor is to differentially enhance cell proliferation over ECM production. After model calibration with historic in-house data, a multi-objective optimization procedure is executed aiming to minimize the total experimental cost whilst maximizing cell growth during culture. The obtained results indicate there are multiple optimum points for the medium refreshment regime and the initial growth factor concentration where a trade-off is made between the final amount of cells and the culture cost. Finally, the model is applied to experiments reported in the literature studying the effects of perfusion-based cell culture and/or growth factor supplementation on cell expansion. The qualitative similarities between the simulation and experimental results, even in the absence of proper model calibration, reinforces the generic character of the proposed modeling framework. The model proposed in this study can contribute to the cost efficient production of cell-based TE products, ultimately contributing to their affordability and accessibility.
Collapse
Affiliation(s)
- Mohammad Mehrian
- Biomechanics Research Unit, GIGA In silico Medicine, University of Liège, Liège, Belgium.,Prometheus, The Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Toon Lambrechts
- Prometheus, The Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,M3-BIORES, KU Leuven, Leuven, Belgium
| | - Ioannis Papantoniou
- Prometheus, The Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Institute of Chemical Engineering Sciences (ICEHT), Foundation for Research and Technology - Hellas (FORTH), Patras, Greece
| | - Liesbet Geris
- Biomechanics Research Unit, GIGA In silico Medicine, University of Liège, Liège, Belgium.,Prometheus, The Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Biomechanics Section, KU Leuven, Leuven, Belgium
| |
Collapse
|
33
|
Moreira F, Mizukami A, de Souza LEB, Cabral JMS, da Silva CL, Covas DT, Swiech K. Successful Use of Human AB Serum to Support the Expansion of Adipose Tissue-Derived Mesenchymal Stem/Stromal Cell in a Microcarrier-Based Platform. Front Bioeng Biotechnol 2020; 8:307. [PMID: 32373600 PMCID: PMC7184110 DOI: 10.3389/fbioe.2020.00307] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 03/20/2020] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSC) are promising candidates for cell-based therapies and for the promotion of tissue repair, hence the increase of clinical trials in a worldwide scale. In particular, adipose tissue-derived stem/stromal cells (AT MSC) present easy accessibility and a rather straightforward process of isolation, providing a clear advantage over other sources. The high demand of cell doses (millions of cells/kg), needed for infusion in clinical settings, requires a scalable and efficient manufacturing of AT MSC under xenogeneic(xeno)-free culture conditions. Here we describe the successful use of human AB serum [10%(v/v)] as a culture supplement, as well as coating substrate for the expansion of these cells in microcarriers using (i) a spinner flask and (ii) a 500-mL mini-bioreactor (ApplikonTM Biotechnology). Cells were characterized by immunophenotype and multilineage differentiation potential. Upon an initial cell adhesion in the spinner flask of 35 ± 2.5%, culture reached a maximal cell density of 2.6 ± 0.1 × 105 at day 7, obtaining a 15 ± 1-fold increase. The implementation of the culture in the 500-mL mini-bioreactor presented an initial cell adhesion of 22 ± 5%, but it reached maximal cell density of 2.7 ± 0.4 × 105 at day 7, obtaining a 27 ± 8-fold increase. Importantly, in both stirred systems, cells retained their immunophenotype and multilineage differentiation potential (osteo-, chondro- and adipogenic lineages). Overall, the scalability of this microcarrier-based system presented herein is of major importance for the purpose of achieving clinically meaningful cell numbers.
Collapse
Affiliation(s)
- Francisco Moreira
- Department of Bioengineering, Instituto Superior Técnico, iBB-Institute for Bioengineering and Biosciences, Universidade de Lisboa, Lisbon, Portugal
| | - Amanda Mizukami
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | | | - Joaquim M S Cabral
- Department of Bioengineering, Instituto Superior Técnico, iBB-Institute for Bioengineering and Biosciences, Universidade de Lisboa, Lisbon, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering, Instituto Superior Técnico, iBB-Institute for Bioengineering and Biosciences, Universidade de Lisboa, Lisbon, Portugal
| | - Dimas T Covas
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Kamilla Swiech
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil.,Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
34
|
Lam AT, Reuveny S, Oh SKW. Human mesenchymal stem cell therapy for cartilage repair: Review on isolation, expansion, and constructs. Stem Cell Res 2020; 44:101738. [DOI: 10.1016/j.scr.2020.101738] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/31/2020] [Accepted: 02/07/2020] [Indexed: 12/29/2022] Open
|
35
|
Sion C, Loubière C, Wlodarczyk-Biegun M, Davoudi N, Müller-Renno C, Guedon E, Chevalot I, Olmos E. Effects of microcarriers addition and mixing on WJ-MSC culture in bioreactors. Biochem Eng J 2020. [DOI: 10.1016/j.bej.2020.107521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
36
|
Borys BS, So T, Roberts EL, Ferrie L, Larijani L, Abraham B, Krawetz R, Rancourt DE, Kallos MS. Large-scale expansion of feeder-free mouse embryonic stem cells serially passaged in stirred suspension bioreactors at low inoculation densities directly from cryopreservation. Biotechnol Bioeng 2020; 117:1316-1328. [PMID: 31960947 DOI: 10.1002/bit.27279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/06/2020] [Accepted: 01/15/2020] [Indexed: 12/23/2022]
Abstract
Embryonic stem cells (ESCs) have almost unlimited proliferation capacity in vitro and can retain the ability to contribute to all cell lineages, making them an ideal platform material for cell-based therapies. ESCs are traditionally cultured in static flasks on a feeder layer of murine embryonic fibroblast cells. Although sufficient to generate cells for research purposes, this approach is impractical to achieve large quantities for clinical applications. In this study, we have developed protocols that address a variety of challenges that currently bottleneck clinical translation of ESCs expanded in stirred suspension bioreactors. We demonstrated that mouse ESCs (mESCs) cryopreserved in the absence of feeder cells could be thawed directly into stirred suspension bioreactors at extremely low inoculation densities (100 cells/ml). These cells sustained proliferative capacity through multiple passages and various reactor sizes and geometries, producing clinically relevant numbers (109 cells) and maintaining pluripotency phenotypic and functional properties. Passages were completed in stirred suspension bioreactors of increasing scale, under defined batch conditions which greatly improved resource efficiency. Output mESCs were analyzed for pluripotency marker expression (SSEA-1, SOX-2, and Nanog) through flow cytometry, and spontaneous differentiation and teratoma analysis was used to demonstrate functional maintenance of pluripotency.
Collapse
Affiliation(s)
- Breanna S Borys
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada
| | - Tania So
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - Erin L Roberts
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada
| | - Leah Ferrie
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada
| | - Leila Larijani
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Brett Abraham
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - Roman Krawetz
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Derrick E Rancourt
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michael S Kallos
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada.,Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
37
|
Godoy JAP, Paiva RMA, Souza AM, Kondo AT, Kutner JM, Okamoto OK. Clinical Translation of Mesenchymal Stromal Cell Therapy for Graft Versus Host Disease. Front Cell Dev Biol 2019; 7:255. [PMID: 31824942 PMCID: PMC6881464 DOI: 10.3389/fcell.2019.00255] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/15/2019] [Indexed: 12/15/2022] Open
Abstract
Graft versus host disease (GVHD) is a common condition in patients subjected to allogeneic hematopoietic stem cell transplantation (HSCT). The immune cells derived from the grafted stem cells attack recipient's tissues, including those from the skin, liver, eyes, mouth, lungs, gastrointestinal tract, neuromuscular system, and genitourinary tract, may lead to severe morbidity and mortality. Acute GVHD can occur within few weeks after the allogeneic cells have engrafted in the recipient while chronic GVHD may occur any time after transplant, typically within months. Although treatable by systemic corticosteroid administration, effective responses are not achieved for a significant proportion of patients, a condition associated with poor prognosis. The use of multipotent mesenchymal stromal cells (MSCs) as an alternative to treat steroid-refractory GVHD had improved last decade, but the results are still controversial. Some studies have shown improvement in the life quality of patients after MSCs treatment, while others have found no significant benefits. In addition to variations in trial design, discrepancies in protocols for MSCs isolation, characterization, and ex vivo manipulation, account for inconsistent clinical results. In this review, we discuss the immunomodulatory properties supporting the therapeutic use of MSCs in GVHD and contextualize the main clinical findings of recent trials using these cells. Critical parameters for the clinical translation of MSCs, including consistent production of MSCs according to Good Manufacturing Practices (GMPs) and informative potency assays for product quality control (QC), are addressed.
Collapse
Affiliation(s)
- Juliana A. P. Godoy
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Raquel M. A. Paiva
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Aline M. Souza
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Andrea T. Kondo
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Jose M. Kutner
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Oswaldo K. Okamoto
- Departamento de Hemoterapia e Terapia Celular, Hospital Israelita Albert Einstein, São Paulo, Brazil
- Centro de Pesquisa sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
38
|
Chen R, Li L, Feng L, Luo Y, Xu M, Leong KW, Yao R. Biomaterial-assisted scalable cell production for cell therapy. Biomaterials 2019; 230:119627. [PMID: 31767445 DOI: 10.1016/j.biomaterials.2019.119627] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 11/01/2019] [Accepted: 11/11/2019] [Indexed: 12/24/2022]
Abstract
Cell therapy, the treatment of diseases using living cells, offers a promising clinical approach to treating refractory diseases. The global market for cell therapy is growing rapidly, and there is an increasing demand for automated methods that can produce large quantities of high quality therapeutic cells. Biomaterials can be used during cell production to establish a biomimetic microenvironment that promotes cell adhesion and proliferation while maintaining target cell genotype and phenotype. Here we review recent progress and emerging techniques in biomaterial-assisted cell production. The increasing use of auxiliary biomaterials and automated production methods provides an opportunity to improve quality control and increase production efficiency using standardized GMP-compliant procedures.
Collapse
Affiliation(s)
- Ruoyu Chen
- Key Laboratory for Advanced Materials Processing Technology of Ministry of Education, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Ling Li
- Key Laboratory for Advanced Materials Processing Technology of Ministry of Education, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Lu Feng
- Key Laboratory for Advanced Materials Processing Technology of Ministry of Education, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Yixue Luo
- Key Laboratory for Advanced Materials Processing Technology of Ministry of Education, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Mingen Xu
- Key Laboratory of Medical Information and 3D Bioprinting of Zhejiang Province, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA.
| | - Rui Yao
- Key Laboratory for Advanced Materials Processing Technology of Ministry of Education, Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
39
|
A fully automated bioreactor system for precise control of stem cell proliferation and differentiation. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2019.107258] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
40
|
Isu G, Morbiducci U, De Nisco G, Kropp C, Marsano A, Deriu MA, Zweigerdt R, Audenino A, Massai D. Modeling methodology for defining a priori the hydrodynamics of a dynamic suspension bioreactor. Application to human induced pluripotent stem cell culture. J Biomech 2019; 94:99-106. [PMID: 31376980 DOI: 10.1016/j.jbiomech.2019.07.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 11/16/2022]
Abstract
Three-dimensional dynamic suspension is becoming an effective cell culture method for a wide range of bioprocesses, with an increasing number of bioreactors proposed for this purpose. The complex hydrodynamics establishing within these devices affects bioprocess outcomes and efficiency, and usually expensive in vitro trial-and-error experiments are needed to properly set the working parameters. Here we propose a methodology to define a priori the hydrodynamic working parameters of a dynamic suspension bioreactor, selected as a test case because of the complex hydrodynamics characterizing its operating condition. A combination of computational and analytical approaches was applied to generate operational guideline graphs for defining a priori specific working parameters. In detail, 43 simulations were performed under pulsed flow regime to characterize advective transport within the device depending on different operative conditions, i.e., culture medium flow rate and its duty cycle, cultured particle diameter, and initial particle suspension volume. The operational guideline graphs were then used to set specific hydrodynamic working parameters for an in vitro proof-of-principle test, where human induced pluripotent stem cell (hiPSC) aggregates were cultured for 24 h within the bioreactor. The in vitro findings showed that, under the selected pulsed flow regime, sedimentation was avoided, hiPSC aggregate circularity and viability were preserved, and culture heterogeneity was reduced, thus confirming the appropriateness of the a priori method. This methodology has the potential to be adaptable to other dynamic suspension devices to support experimental studies by providing in silico-based a priori knowledge, useful to limit costs and to optimize culture bioprocesses.
Collapse
Affiliation(s)
- Giuseppe Isu
- Polito(BIO)Med Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Department of Surgery and Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Umberto Morbiducci
- Polito(BIO)Med Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Italy
| | - Giuseppe De Nisco
- Polito(BIO)Med Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Italy
| | - Christina Kropp
- Leibniz Research Laboratories for Biotechnology and Artificial Organ, Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Anna Marsano
- Department of Surgery and Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Marco A Deriu
- Polito(BIO)Med Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Italy
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organ, Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Alberto Audenino
- Polito(BIO)Med Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Italy
| | - Diana Massai
- Polito(BIO)Med Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Italy.
| |
Collapse
|
41
|
|
42
|
Borys BS, Le A, Roberts EL, Dang T, Rohani L, Hsu CYM, Wyma AA, Rancourt DE, Gates ID, Kallos MS. Using computational fluid dynamics (CFD) modeling to understand murine embryonic stem cell aggregate size and pluripotency distributions in stirred suspension bioreactors. J Biotechnol 2019; 304:16-27. [PMID: 31394111 DOI: 10.1016/j.jbiotec.2019.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/19/2019] [Accepted: 08/03/2019] [Indexed: 12/15/2022]
Abstract
Computational fluid dynamics (CFD) modeling can be applied to understand hydrodynamics in stirred suspension bioreactors, which can in turn affect cell viability, proliferation, pluripotency and differentiation. In this study, we developed a CFD model to determine the effects of average shear rates and turbulent eddies on the formation and growth of murine embryonic stem cell aggregates. We found a correlation between average eddy size and aggregate size, which depended on bioreactor agitation rates. By relating these computational and biological variables, CFD modeling can predict optimal agitation rates to grow embryonic stem cell aggregates in stirred suspension bioreactors. To examine the effect of hydrodynamics on pluripotency, mESCs cultured in bioreactors under various agitation rates were tested for SSEA-1, Sox-2, and Nanog expression. Cells maintained a minimum of 95% positive expression with no change in the intensity distribution pattern between the different bioreactor conditions. This indicates that the average level of pluripotency marker expression is independent of changes in the hydrodynamic profile and resulting aggregate size distribution. The findings here can be further extended to other cell types that grow as aggregates in stirred suspension bioreactors and offer important insights necessary to realize cell therapies.
Collapse
Affiliation(s)
- Breanna S Borys
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada; Biomedical Engineering Graduate Program, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - An Le
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada; Biomedical Engineering Graduate Program, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Erin L Roberts
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada; Biomedical Engineering Graduate Program, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Tiffany Dang
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada; Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Leili Rohani
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 4N1, Canada
| | - Charlie Yu-Ming Hsu
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 4N1, Canada
| | - Alexander A Wyma
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada; Biomedical Engineering Graduate Program, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Derrick E Rancourt
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N 4N1, Canada
| | - Ian D Gates
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada
| | - Michael S Kallos
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada; Biomedical Engineering Graduate Program, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada; Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
43
|
Nguyen LTB, Odeleye AOO, Chui CY, Baudequin T, Cui Z, Ye H. Development of thermo-responsive polycaprolactone macrocarriers conjugated with Poly(N-isopropyl acrylamide) for cell culture. Sci Rep 2019; 9:3477. [PMID: 30837639 PMCID: PMC6401373 DOI: 10.1038/s41598-019-40242-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 02/12/2019] [Indexed: 01/20/2023] Open
Abstract
Poly(N-isopropyl acrylamide) (PNIPAAm) is a well-known 'smart' material responding to external stimuli such as temperature. PNIPAAm was successfully conjugated to polycaprolactone (PCL) bead surfaces through amidation reaction. Functionalization steps were characterized and confirmed by Fourier transform infrared spectroscopy, X-ray photoelectron spectroscopy, scanning electron microscopy and Energy Dispersion Spectroscopy. PNIPAAm-conjugated PCL allowed human dermal fibroblast cells (HDF) and mesenchymal stem cells (MSC) to adhere, spread, and grow successfully. By reducing the temperature to 30 °C, more than 70% of HDF were detached from PNIPAAm-conjugated PCL macrocarriers with 85% viability. The cell detachment ratio by trypsin treatment was slightly higher than that induced by reduced temperature, however, cell detachment from PNIPAAm-conjugated macrocarriers by lowering the temperature significantly reduced cell death and increased both cell viability and the recovery potential of the detached cells. HDF attachment and detachment were also observed by Live-Dead staining and phase contrast imaging. The expression of extracellular matrix proteins such as Laminin and Fibronectin was also affected by the trypsinization process but not by the reduced temperature process. Taken together, our results showed that thermo-responsive macrocarriers could be a promising alternative method for the non-invasive detachment of cells, in particular for tissue engineering, clinical applications and the use of bioreactors.
Collapse
Affiliation(s)
- Linh T B Nguyen
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| | - Akinlolu O O Odeleye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
- Adaptimmune Limited, 60 Jubilee Avenue, Milton Park, Abingdon, OX14 4RX, UK
| | - Chih-Yao Chui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| | - Timothée Baudequin
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK.
| |
Collapse
|
44
|
Lerman MJ, Muramoto S, Arumugasaamy N, Van Order M, Lembong J, Gerald AG, Gillen G, Fisher JP. Development of surface functionalization strategies for 3D-printed polystyrene constructs. J Biomed Mater Res B Appl Biomater 2019; 107:2566-2578. [PMID: 30821930 DOI: 10.1002/jbm.b.34347] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/18/2019] [Accepted: 02/10/2019] [Indexed: 01/17/2023]
Abstract
There is a growing interest in 3D printing to fabricate culture substrates; however, the surface properties of the scaffold remain pertinent to elicit targeted and expected cell responses. Traditional 2D polystyrene (PS) culture systems typically require surface functionalization (oxidation) to facilitate and encourage cell adhesion. Determining the surface properties which enhance protein adhesion from media and cellular extracellular matrix (ECM) production remains the first step to translating 2D PS systems to a 3D culture surface. Here we show that the presence of carbonyl groups to PS surfaces correlated well with successful adhesion of ECM proteins and sustaining ECM production of deposited human mesenchymal stem cells, if the surface has a water contact angle between 50° and 55°. Translation of these findings to custom-fabricated 3D PS scaffolds reveals carbonyl groups continued to enhance spreading and growth in 3D culture. Cumulatively, these data present a method for 3D printing PS and the design considerations required for understanding cell-material interactions. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B:2566-2578, 2019.
Collapse
Affiliation(s)
- Max J Lerman
- Department of Materials Science and Engineering, University of Maryland, College Park, Maryland.,Surface and Trace Chemical Analysis Group, Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland.,Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
| | - Shin Muramoto
- Surface and Trace Chemical Analysis Group, Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland
| | - Navein Arumugasaamy
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland.,Fischell Department of Bioengineerin, University of Maryland, College Park, Maryland.,Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia
| | - Michael Van Order
- Department of Materials Science and Engineering, University of Maryland, College Park, Maryland
| | - Josephine Lembong
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland.,Fischell Department of Bioengineerin, University of Maryland, College Park, Maryland
| | - Anushka G Gerald
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland.,Fischell Department of Bioengineerin, University of Maryland, College Park, Maryland
| | - Greg Gillen
- Surface and Trace Chemical Analysis Group, Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland
| | - John P Fisher
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland.,Fischell Department of Bioengineerin, University of Maryland, College Park, Maryland
| |
Collapse
|
45
|
Torizal FG, Horiguchi I, Sakai Y. Physiological Microenvironmental Conditions in Different Scalable Culture Systems for Pluripotent Stem Cell Expansion and Differentiation. Open Biomed Eng J 2019. [DOI: 10.2174/1874120701913010041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Human Pluripotent Stem Cells (PSCs) are a valuable cell type that has a wide range of biomedical applications because they can differentiate into many types of adult somatic cell. Numerous studies have examined the clinical applications of PSCs. However, several factors such as bioreactor design, mechanical stress, and the physiological environment have not been optimized. These factors can significantly alter the pluripotency and proliferation properties of the cells, which are important for the mass production of PSCs. Nutritional mass transfer and oxygen transfer must be effectively maintained to obtain a high yield. Various culture systems are currently available for optimum cell propagation by maintaining the physiological conditions necessary for cell cultivation. Each type of culture system using a different configuration with various advantages and disadvantages affecting the mechanical conditions in the bioreactor, such as shear stress. These factors make it difficult to preserve the cellular viability and pluripotency of PSCs. Additional limitations of the culture system for PSCs must also be identified and overcome to maintain the culture conditions and enable large-scale expansion and differentiation of PSCs. This review describes the different physiological conditions in the various culture systems and recent developments in culture technology for PSC expansion and differentiation.
Collapse
|
46
|
Yoshida K, Okada M, Nagasaka R, Sasaki H, Okada M, Kanie K, Kato R. Time-course colony tracking analysis for evaluating induced pluripotent stem cell culture processes. J Biosci Bioeng 2019; 128:209-217. [PMID: 30738731 DOI: 10.1016/j.jbiosc.2019.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/11/2019] [Accepted: 01/16/2019] [Indexed: 01/15/2023]
Abstract
Increasing the yield and maintaining a high quality of induced pluripotent stem cells (iPSCs) is necessary for manufacturing iPSCs at the industrial scale. However, because iPSCs are delicate, it is important to evaluate their quality during processing. To examine the status of cultured iPSCs non-invasively, morphology-based iPSC colony evaluation may be an efficient technology for cellular status monitoring and analysis. In this study, we examined the effectiveness of time-course colony tracking analysis for evaluating the iPSC culture process. Particularly, we obtained detailed time-course data to evaluate the effect of the pipetting technique on cell dissociation before seeding. Although the pipetting process causes severe shear stress to cells, which affects their quality, these effects have not been quantitatively analyzed because of their complex and uncontrollable parameters. By analyzing the heterogeneity and time-course responses of individual colonies, our colony tracking analysis revealed a critically damaged population caused by pipetting stress which could not be detected in conventional bulk analysis. Moreover, by comprehensively analyzing colony tracking data, which links the time-course morphology and marker staining results with each colony, we found that colony morphology is only highly correlated with the undifferentiated marker in the final stage, with a lower correlation in the early stages. Thus, colony tracking analysis provides a way to quantify cellular morphological information when evaluating complex iPSC manufacturing processes.
Collapse
Affiliation(s)
- Kei Yoshida
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furocho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Mika Okada
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furocho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Risako Nagasaka
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furocho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Hiroto Sasaki
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Furocho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Mai Okada
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furocho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Kei Kanie
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furocho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Ryuji Kato
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furocho, Chikusa-ku, Nagoya 464-8602, Japan; Stem Cell Evaluation Technology Research Association (SCA), Hacho-bori, Chuou-ku, Tokyo 104-0032, Japan; Institute of Nano-Life-Systems, Institute of Innovation for Future Society, Nagoya University, Division of Micro-Nano Mechatronics, Furocho, Chikusa-ku, Nagoya 464-8602, Japan.
| |
Collapse
|
47
|
Study of hydrodynamics in wave bioreactors by computational fluid dynamics reveals a resonance phenomenon. Chem Eng Sci 2019. [DOI: 10.1016/j.ces.2018.08.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
48
|
He Q, Ye Z, Zhou Y, Tan WS. Comparative study of mesenchymal stem cells from rat bone marrow and adipose tissue. Turk J Biol 2018; 42:477-489. [PMID: 30983864 PMCID: PMC6451846 DOI: 10.3906/biy-1802-52] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Several therapeutic products based on mesenchymal stem cells (MSCs) have been translated into clinical applications. MSCs should undergo in vitro culture before a sufficient quantity can be achieved. Hence, both expansion kinetics and the biological characteristics of derived cells from primary culture are pertinent to their applications. In the present study, MSCs were isolated from rat bone marrow and adipose tissue (designated as bMSCs and aMSCs, respectively) and cells were comparatively analyzed regarding cell morphology, proliferation, colony formation, differentiation potential, and immunophenotype following the long-term subculture. No apparent differences could be noticed concerning the morphology between bMSCs and aMSCs. The long-term subculture made both types of cells smaller, weakened their colony-forming ability, and stimulated the proliferation rate. However, bMSCs demonstrated better proliferation and colony-forming ability than aMSCs. No significant difference was observed about the expression of some immunophenotypes (i.e. CD29+/CD90+/CD34-/CD45-) regardless of cell types or population doublings. Notably, bMSCs, but not aMSCs, maintained the differentiation potential well after the long-term subculture. The present study demonstrates that MSCs derived from different tissues can be well expanded for the long term, although cells display gradually declined self-renewal and differentiation potentials to different extents depending on the tissue origins.
Collapse
Affiliation(s)
- Qing He
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology , Shanghai , P.R. China
| | - Zhaoyang Ye
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology , Shanghai , P.R. China
| | - Yan Zhou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology , Shanghai , P.R. China
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology , Shanghai , P.R. China
| |
Collapse
|
49
|
Dakhore S, Nayer B, Hasegawa K. Human Pluripotent Stem Cell Culture: Current Status, Challenges, and Advancement. Stem Cells Int 2018; 2018:7396905. [PMID: 30595701 PMCID: PMC6282144 DOI: 10.1155/2018/7396905] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/23/2022] Open
Abstract
Over the past two decades, human embryonic stem cells (hESCs) have gained attention due to their pluripotent and proliferative ability which enables production of almost all cell types in the human body in vitro and makes them an excellent tool to study human embryogenesis and disease, as well as for drug discovery and cell transplantation therapies. Discovery of human-induced pluripotent stem cells (hiPSCs) further expanded therapeutic applications of human pluripotent stem cells (PSCs). hPSCs provide a stable and unlimited original cell source for producing suitable cells and tissues for downstream applications. Therefore, engineering the environment in which these cells are grown, for stable and quality-controlled hPSC maintenance and production, is one of the key factors governing the success of these applications. hPSCs are maintained in a particular niche using specific cell culture components. Ideally, the culture should be free of xenobiotic components to render hPSCs suitable for therapeutic applications. Substantial efforts have been put to identify effective components, and develop culture conditions and protocols, for their large-scale expansion without compromising on quality. In this review, we discuss different media, their components and functions, including specific requirements to maintain the pluripotent and proliferative ability of hPSCs. Understanding the role of culture components would enable the development of appropriate conditions to promote large-scale, quality-controlled expansion of hPSCs thereby increasing their potential applications.
Collapse
Affiliation(s)
- Sushrut Dakhore
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences (NCBS), Bangalore, India
| | - Bhavana Nayer
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences (NCBS), Bangalore, India
| | - Kouichi Hasegawa
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences (NCBS), Bangalore, India
- Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advanced Study, Kyoto University, Japan
| |
Collapse
|
50
|
Lerman MJ, Lembong J, Muramoto S, Gillen G, Fisher JP. The Evolution of Polystyrene as a Cell Culture Material. TISSUE ENGINEERING. PART B, REVIEWS 2018; 24:359-372. [PMID: 29631491 PMCID: PMC6199621 DOI: 10.1089/ten.teb.2018.0056] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 03/21/2018] [Indexed: 01/19/2023]
Abstract
Polystyrene (PS) has brought in vitro cell culture from its humble beginnings to the modern era, propelling dozens of research fields along the way. This review discusses the development of the material, fabrication, and treatment approaches to create the culture material. However, native PS surfaces poorly facilitate cell adhesion and growth in vitro. To overcome this, liquid surface deposition, energetic plasma activation, and emerging functionalization methods transform the surface chemistry. This review seeks to highlight the many potential applications of the first widely accepted polymer growth surface. Although the majority of in vitro research occurs on two-dimensional surfaces, the importance of three-dimensional (3D) culture models cannot be overlooked. The methods to transition PS to specialized 3D culture surfaces are also reviewed. Specifically, casting, electrospinning, 3D printing, and microcarrier approaches to shift PS to a 3D culture surface are highlighted. The breadth of applications of the material makes it impossible to highlight every use, but the aim remains to demonstrate the versatility and potential as both a general and custom cell culture surface. The review concludes with emerging scaffolding approaches and, based on the findings, presents our insights on the future steps for PS as a tissue culture platform.
Collapse
Affiliation(s)
- Max J. Lerman
- Department of Materials Science and Engineering, University of Maryland, College Park, Maryland
- Surface and Trace Chemical Analysis Group, Materials Measurement Lab, National Institute of Standards and Technology, Gaithersburg, Maryland
- NIH/NIBIB Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
| | - Josephine Lembong
- NIH/NIBIB Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Shin Muramoto
- Surface and Trace Chemical Analysis Group, Materials Measurement Lab, National Institute of Standards and Technology, Gaithersburg, Maryland
| | - Greg Gillen
- Surface and Trace Chemical Analysis Group, Materials Measurement Lab, National Institute of Standards and Technology, Gaithersburg, Maryland
| | - John P. Fisher
- NIH/NIBIB Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| |
Collapse
|