1
|
Borys BS, Dang T, Worden H, Larijani L, Corpuz JM, Abraham BD, Gysel EJ, Malinovska J, Krawetz R, Revay T, Argiropoulos B, Rancourt DE, Kallos MS, Jung S. Robust bioprocess design and evaluation of commercial media for the serial expansion of human induced pluripotent stem cell aggregate cultures in vertical-wheel bioreactors. Stem Cell Res Ther 2024; 15:232. [PMID: 39075528 PMCID: PMC11288049 DOI: 10.1186/s13287-024-03819-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 06/27/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND While pluripotent stem cell (PSC) therapies move toward clinical and commercial applications at a rapid rate, manufacturing reproducibility and robustness are notable bottlenecks in regulatory approval. Therapeutic applications of PSCs require large cell quantities to be generated under highly robust, well-defined, and economically viable conditions. Small-scale and short-term process optimization, however, is often performed in a linear fashion that does not account for time needed to verify the bioprocess protocols and analysis methods used. Design of a reproducible and robust bioprocess should be dynamic and include a continuous effort to understand how the process will respond over time and to different stresses before transitioning into large-scale production where stresses will be amplified. METHODS This study utilizes a baseline protocol, developed for the short-term culture of PSC aggregates in Vertical-Wheel® bioreactors, to evaluate key process attributes through long-term (serial passage) suspension culture. This was done to access overall process robustness when performed with various commercially available media and cell lines. Process output variables including growth kinetics, aggregate morphology, harvest efficiency, genomic stability, and functional pluripotency were assessed through short and long-term culture. RESULTS The robust nature of the expansion protocol was demonstrated over a six-day culture period where spherical aggregate formation and expansion were observed with high-fold expansions for all five commercial media tested. Profound differences in cell growth and quality were revealed only through long-term serial expansion and in-vessel dissociation operations. Some commercial media formulations tested demonstrated maintenance of cell growth rates, aggregate morphology, and high harvest recovery efficiencies through three bioreactor serial passages using multiple PSC lines. Exceptional bioprocess robustness was even demonstrated with sustained growth and quality maintenance over 10 serial bioreactor passages. However, some commercial media tested proved less equipped for serial passage cultures in bioreactors as cultures led to cell lysis during dissociation, reduction in growth rates, and a loss of aggregate morphology. CONCLUSIONS This study demonstrates the importance of systematic selection and testing of bioprocess input variables, with multiple bioprocess output variables through serial passages to create a truly reproducible and robust protocol for clinical and commercial PSC production using scalable bioreactor systems.
Collapse
Affiliation(s)
- Breanna S Borys
- Pharmaceutical Production Research Facility, University of Calgary, Calgary, AB, Canada
- PBS Biotech Inc, 4721 Calle Carga, Camarillo, CA, 93012, USA
| | - Tiffany Dang
- Pharmaceutical Production Research Facility, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Hannah Worden
- PBS Biotech Inc, 4721 Calle Carga, Camarillo, CA, 93012, USA
| | - Leila Larijani
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Medical Genetics, University of Calgary, Calgary, AB, Canada
| | - Jessica M Corpuz
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Brett D Abraham
- Pharmaceutical Production Research Facility, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Emilie J Gysel
- Pharmaceutical Production Research Facility, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Julia Malinovska
- Pharmaceutical Production Research Facility, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Roman Krawetz
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Tamas Revay
- Department of Medical Genetics, Alberta Health Services, Alberta Children's Hospital, Calgary, AB, Canada
| | - Bob Argiropoulos
- Department of Medical Genetics, Alberta Health Services, Alberta Children's Hospital, Calgary, AB, Canada
| | - Derrick E Rancourt
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - Michael S Kallos
- Pharmaceutical Production Research Facility, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Sunghoon Jung
- PBS Biotech Inc, 4721 Calle Carga, Camarillo, CA, 93012, USA.
| |
Collapse
|
2
|
Iworima DG, Baker RK, Ellis C, Sherwood C, Zhan L, Rezania A, Piret JM, Kieffer TJ. Metabolic switching, growth kinetics and cell yields in the scalable manufacture of stem cell-derived insulin-producing cells. Stem Cell Res Ther 2024; 15:1. [PMID: 38167219 PMCID: PMC10762849 DOI: 10.1186/s13287-023-03574-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 11/16/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Diabetes is a disease affecting over 500 million people globally due to insulin insufficiency or insensitivity. For individuals with type 1 diabetes, pancreatic islet transplantation can help regulate their blood glucose levels. However, the scarcity of cadaveric donor islets limits the number of people that could receive this therapy. To address this issue, human pluripotent stem cells offer a potentially unlimited source for generating insulin-producing cells through directed differentiation. Several protocols have been developed to make stem cell-derived insulin-producing cells. However, there is a lack of knowledge regarding the bioprocess parameters associated with these differentiation protocols and how they can be utilized to increase the cell yield. METHODS We investigated various bioprocess parameters and quality target product profiles that may influence the differentiation pipeline using a seven-stage protocol in a scalable manner with CellSTACKs and vertical wheel bioreactors (PBS-Minis). RESULTS Cells maintained > 80% viability through all stages of differentiation and appropriately expressed stage-specific markers. During the initial four stages leading up to the development of pancreatic progenitors, there was an increase in cell numbers. Following pancreatic progenitor stage, there was a gradual decrease in the percentage of proliferative cells, as determined by Ki67 positivity, and a significant loss of cells during the period of endocrine differentiation. By minimizing the occurrence of aggregate fusion, we were able to enhance cell yield during the later stages of differentiation. We suggest that glucose utilization and lactate production are cell quality attributes that should be considered during the characterization of insulin-producing cells derived from stem cells. Our findings also revealed a gradual metabolic shift from glycolysis, during the initial four stages of pancreatic progenitor formation, to oxidative phosphorylation later on during endocrine differentiation. Furthermore, the resulting insulin-producing cells exhibited a response to several secretagogues, including high glucose. CONCLUSION This study demonstrates process parameters such as glucose consumption and lactate production rates that may be used to facilitate the scalable manufacture of stem cell-derived insulin-producing cells.
Collapse
Affiliation(s)
- Diepiriye G Iworima
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Robert K Baker
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Cara Ellis
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Chris Sherwood
- Michael Smith Laboratories, The University of British Columbia, Vancouver, BC, Canada
| | - Lisa Zhan
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | | | - James M Piret
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
- Michael Smith Laboratories, The University of British Columbia, Vancouver, BC, Canada
- Department of Chemical and Biological Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Timothy J Kieffer
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada.
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada.
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
3
|
Wu H, Tang X, Wang Y, Wang N, Chen Q, Xie J, Liu S, Zhong Z, Qiu Y, Situ P, Zern MA, Wang J, Chen H, Duan Y. Dextran sulfate prevents excess aggregation of human pluripotent stem cells in 3D culture by inhibiting ICAM1 expression coupled with down-regulating E-cadherin through activating the Wnt signaling pathway. Stem Cell Res Ther 2022; 13:218. [PMID: 35619172 PMCID: PMC9137216 DOI: 10.1186/s13287-022-02890-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/25/2022] [Indexed: 11/29/2022] Open
Abstract
Background Human pluripotent stem cells (hPSCs) have great potential in applications for regenerative medicine and drug development. However, 3D suspension culture systems for clinical-grade hPSC large-scale production have been a major challenge. Accumulating evidence has demonstrated that the addition of dextran sulfate (DS) could prevent excessive adhesion of hPSCs from forming larger aggregates in 3D suspension culture. However, the signaling and molecular mechanisms underlying this phenomenon remain elusive. Methods By using a cell aggregate culture assay and separating big and small aggregates in suspension culture systems, the potential mechanism and downstream target genes of DS were investigated by mRNA sequence analysis, qRT-PCR validation, colony formation assay, and interference assay. Results Since cellular adhesion molecules (CAMs) play important roles in hPSC adhesion and aggregation, we assumed that DS might prevent excess adhesion through affecting the expression of CAMs in hPSCs. As expected, after DS treatment, we found that the expression of CAMs was significantly down-regulated, especially E-cadherin (E-cad) and intercellular adhesion molecule 1 (ICAM1), two highly expressed CAMs in hPSCs. The role of E-cad in the adhesion of hPSCs has been widely investigated, but the function of ICAM1 in hPSCs is hardly understood. In the present study, we demonstrated that ICAM1 exhibited the capacity to promote the adhesion in hPSCs, and this adhesion was suppressed by the treatment with DS. Furthermore, transcriptomic analysis of RNA-seq revealed that DS treatment up-regulated genes related to Wnt signaling resulting in the activation of Wnt signaling in which SLUG, TWIST, and MMP3/7 were highly expressed, and further inhibited the expression of E-cad. Conclusion Our results demonstrated that DS played an important role in controlling the size of hPSC aggregates in 3D suspension culture by inhibiting the expression of ICAM1 coupled with the down-regulation of E-cad through the activation of the Wnt signaling pathway. These results represent a significant step toward developing the expansion of hPSCs under 3D suspension condition in large-scale cultures. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02890-4.
Collapse
Affiliation(s)
- Haibin Wu
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Xianglian Tang
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.,School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510180, People's Republic of China.,Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Health Commission Key Laboratory of Precise Diagnosis and Treatment of Genetic Diseases, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530003, Guangxi, People's Republic of China.,Genetic and Metabolic Central Laboratory, Guangxi Birth Defects Research and Prevention Institute, Nanning, 530003, Guangxi, People's Republic of China
| | - Yiyu Wang
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Ning Wang
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.,School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510180, People's Republic of China
| | - Qicong Chen
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.,School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510180, People's Republic of China
| | - Jinghe Xie
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.,School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510180, People's Republic of China
| | - Shoupei Liu
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Zhiyong Zhong
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.,School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510180, People's Republic of China
| | - Yaqi Qiu
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Ping Situ
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.,School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510180, People's Republic of China
| | - Mark A Zern
- Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, 95817, USA
| | - Jue Wang
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.
| | - Honglin Chen
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China. .,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510180, People's Republic of China. .,Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510180, People's Republic of China. .,Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510180, People's Republic of China.
| | - Yuyou Duan
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, No. 382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China. .,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510180, People's Republic of China. .,Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510180, People's Republic of China. .,Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510180, People's Republic of China.
| |
Collapse
|
4
|
Yuen JSK, Stout AJ, Kawecki NS, Letcher SM, Theodossiou SK, Cohen JM, Barrick BM, Saad MK, Rubio NR, Pietropinto JA, DiCindio H, Zhang SW, Rowat AC, Kaplan DL. Perspectives on scaling production of adipose tissue for food applications. Biomaterials 2022; 280:121273. [PMID: 34933254 PMCID: PMC8725203 DOI: 10.1016/j.biomaterials.2021.121273] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 01/03/2023]
Abstract
With rising global demand for food proteins and significant environmental impact associated with conventional animal agriculture, it is important to develop sustainable alternatives to supplement existing meat production. Since fat is an important contributor to meat flavor, recapitulating this component in meat alternatives such as plant based and cell cultured meats is important. Here, we discuss the topic of cell cultured or tissue engineered fat, growing adipocytes in vitro that could imbue meat alternatives with the complex flavor and aromas of animal meat. We outline potential paths for the large scale production of in vitro cultured fat, including adipogenic precursors during cell proliferation, methods to adipogenically differentiate cells at scale, as well as strategies for converting differentiated adipocytes into 3D cultured fat tissues. We showcase the maturation of knowledge and technology behind cell sourcing and scaled proliferation, while also highlighting that adipogenic differentiation and 3D adipose tissue formation at scale need further research. We also provide some potential solutions for achieving adipose cell differentiation and tissue formation at scale based on contemporary research and the state of the field.
Collapse
Affiliation(s)
- John S K Yuen
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Andrew J Stout
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - N Stephanie Kawecki
- Department of Bioengineering, University of California Los Angeles, 410 Westwood Plaza, Los Angeles, CA, 90095, USA; Department of Integrative Biology & Physiology, University of California Los Angeles, Terasaki Life Sciences Building, 610 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | - Sophia M Letcher
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Sophia K Theodossiou
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Julian M Cohen
- W. M. Keck Science Department, Pitzer College, 925 N Mills Ave, Claremont, CA, 91711, USA
| | - Brigid M Barrick
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Michael K Saad
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Natalie R Rubio
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Jaymie A Pietropinto
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Hailey DiCindio
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Sabrina W Zhang
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Amy C Rowat
- Department of Bioengineering, University of California Los Angeles, 410 Westwood Plaza, Los Angeles, CA, 90095, USA; Department of Integrative Biology & Physiology, University of California Los Angeles, Terasaki Life Sciences Building, 610 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | - David L Kaplan
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA.
| |
Collapse
|
5
|
Torizal FG, Kim SM, Horiguchi I, Inamura K, Suzuki I, Morimura T, Nishikawa M, Sakai Y. Production of homogenous size-controlled human induced pluripotent stem cell aggregates using ring-shaped culture vessel. J Tissue Eng Regen Med 2021; 16:254-266. [PMID: 34923748 DOI: 10.1002/term.3278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 10/23/2021] [Accepted: 12/16/2021] [Indexed: 01/01/2023]
Abstract
Aggregate size is an important parameter that determines the cell fate and quality of the resulting human-induced pluripotent stem cells (hiPSCs). Nowadays, large-scale suspension culture is a common method for scaling-up the biomanufacturing of hiPSCs to realize their practical application. However, this culture system exhibits a complex hydrodynamic condition resulting from the different mixing conditions of culture media, which potentially produce non-uniform aggregates, which may decrease the quality of the cell yield. Here, we performed expansion in a ring-shaped culture vessel and compared it with three other suspension-based culture systems to evaluate the uniformity and characteristics of hiPSC aggregates. Morphologically, the hiPSC aggregates formed and expanded in the ring-shaped culture vessel, resulting in small and uniform aggregates compared to the other culture systems. This aggregate population showed a decent mass transfer required for the exchange of biochemical substances, such as nutrients, growth factors, oxygen, and waste metabolic products, inside the aggregates. Thus, better metabolic performance and pluripotency markers were achieved in this system. Interestingly, all culture systems used in this study showed different tendencies in embryoid body differentiation. The smaller aggregates produced by sphere ring and dish bag tended to differentiate toward ectodermal and mesodermal lineages, while predominantly larger aggregates from the 6-well plates and spinner flask exhibited more potential for endodermal lineage. Our study demonstrates the production of a decent homogenous aggregate population by providing equal hydrodynamic force through the ring-shaped culture vessel design, which may be further upscaled to produce a large number of hiPSCs for clinical applications.
Collapse
Affiliation(s)
- Fuad Gandhi Torizal
- Department of Chemical Systems Engineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Japan.,Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Japan
| | - Seong Min Kim
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Japan
| | - Ikki Horiguchi
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Japan
| | - Kousuke Inamura
- Department of Chemical Systems Engineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Japan
| | - Ikumi Suzuki
- Division of Biotechnology Industrial Equipments, Fukoku Ltd, Saitama, Japan
| | - Takashi Morimura
- Division of Biotechnology Industrial Equipments, Fukoku Ltd, Saitama, Japan
| | - Masaki Nishikawa
- Department of Chemical Systems Engineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Japan
| | - Yasuyuki Sakai
- Department of Chemical Systems Engineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Japan.,Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Japan
| |
Collapse
|
6
|
Rivera-Ordaz A, Peli V, Manzini P, Barilani M, Lazzari L. Critical Analysis of cGMP Large-Scale Expansion Process in Bioreactors of Human Induced Pluripotent Stem Cells in the Framework of Quality by Design. BioDrugs 2021; 35:693-714. [PMID: 34727354 PMCID: PMC8561684 DOI: 10.1007/s40259-021-00503-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2021] [Indexed: 10/28/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs) are manufactured as advanced therapy medicinal products for tissue replacement applications. With this aim, the feasibility of hiPSC large-scale expansion in existing bioreactor systems under current good manufacturing practices (cGMP) has been tested. Yet, these attempts have lacked a paradigm shift in culture settings and technologies tailored to hiPSCs, which jeopardizes their clinical translation. The best approach for industrial scale-up of high-quality hiPSCs is to design their manufacturing process by following quality-by-design (QbD) principles: a scientific, risk-based framework for process design based on relating product and process attributes to product quality. In this review, we analyzed the hiPSC expansion manufacturing process implementing the QbD approach in the use of bioreactors, stressing the decisive role played by the cell quantity, quality and costs, drawing key QbD concepts directly from the guidelines of the International Council for Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use.
Collapse
Affiliation(s)
- Araceli Rivera-Ordaz
- Laboratory of Regenerative Medicine-Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | - Valeria Peli
- Laboratory of Regenerative Medicine-Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | - Paolo Manzini
- Laboratory of Regenerative Medicine-Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | - Mario Barilani
- Laboratory of Regenerative Medicine-Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.
| | - Lorenza Lazzari
- Laboratory of Regenerative Medicine-Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| |
Collapse
|
7
|
Suong DNA, Imamura K, Inoue I, Kabai R, Sakamoto S, Okumura T, Kato Y, Kondo T, Yada Y, Klein WL, Watanabe A, Inoue H. Induction of inverted morphology in brain organoids by vertical-mixing bioreactors. Commun Biol 2021; 4:1213. [PMID: 34686776 PMCID: PMC8536773 DOI: 10.1038/s42003-021-02719-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/28/2021] [Indexed: 12/19/2022] Open
Abstract
Organoid technology provides an opportunity to generate brain-like structures by recapitulating developmental steps in the manner of self-organization. Here we examined the vertical-mixing effect on brain organoid structures using bioreactors and established inverted brain organoids. The organoids generated by vertical mixing showed neurons that migrated from the outer periphery to the inner core of organoids, in contrast to orbital mixing. Computational analysis of flow dynamics clarified that, by comparison with orbital mixing, vertical mixing maintained the high turbulent energy around organoids, and continuously kept inter-organoid distances by dispersing and adding uniform rheological force on organoids. To uncover the mechanisms of the inverted structure, we investigated the direction of primary cilia, a cellular mechanosensor. Primary cilia of neural progenitors by vertical mixing were aligned in a multidirectional manner, and those by orbital mixing in a bidirectional manner. Single-cell RNA sequencing revealed that neurons of inverted brain organoids presented a GABAergic character of the ventral forebrain. These results suggest that controlling fluid dynamics by biomechanical engineering can direct stem cell differentiation of brain organoids, and that inverted brain organoids will be applicable for studying human brain development and disorders in the future. Dang Ngoc Anh Suong et al find that vertical mixing generates iPSC-derived brain organoids displaying an inverted structure with neurons localising at the centre and neural progenitors at the outside. This study illustrates the influence of fluid mechanics relevant to the direction of primary cilia on stem cell differentiation.
Collapse
Affiliation(s)
- Dang Ngoc Anh Suong
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
| | - Keiko Imamura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan.,Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Ikuyo Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Ryotaro Kabai
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | - Yoshikazu Kato
- Mixing Technology Laboratory, SATAKE Chemical Equipment Manufacturing Ltd., Saitama, Japan
| | - Takayuki Kondo
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan.,Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Yuichiro Yada
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
| | - William L Klein
- Department of Neurobiology, Northwestern University, Evanston, IL, 60208, USA
| | - Akira Watanabe
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan. .,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan. .,Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan. .,Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan.
| |
Collapse
|
8
|
Tang X, Wu H, Xie J, Wang N, Chen Q, Zhong Z, Qiu Y, Wang J, Li X, Situ P, Lai L, Zern MA, Chen H, Duan Y. The combination of dextran sulphate and polyvinyl alcohol prevents excess aggregation and promotes proliferation of pluripotent stem cells in suspension culture. Cell Prolif 2021; 54:e13112. [PMID: 34390064 PMCID: PMC8450127 DOI: 10.1111/cpr.13112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/13/2021] [Accepted: 07/24/2021] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES For clinical applications of cell-based therapies, a large quantity of human pluripotent stem cells (hPSCs) produced in standardized and scalable culture processes is required. Currently, microcarrier-free suspension culture shows potential for large-scale expansion of hPSCs; however, hPSCs tend to aggregate during culturing leading to a negative effect on cell yield. To overcome this problem, we developed a novel protocol to effectively control the sizes of cell aggregates and enhance the cell proliferation during the expansion of hPSCs in suspension. MATERIALS AND METHODS hPSCs were expanded in suspension culture supplemented with polyvinyl alcohol (PVA) and dextran sulphate (DS), and 3D suspension culture of hPSCs formed cell aggregates under static or dynamic conditions. The sizes of cell aggregates and the cell proliferation as well as the pluripotency of hPSCs after expansion were assessed using cell counting, size analysis, real-time quantitative polymerase chain reaction, flow cytometry analysis, immunofluorescence staining, embryoid body formation, teratoma formation and transcriptome sequencing. RESULTS Our results demonstrated that the addition of DS alone effectively prevented hPSC aggregation, while the addition of PVA significantly enhanced hPSC proliferation. The combination of PVA and DS not only promoted cell proliferation of hPSCs but also produced uniform and size-controlled cell aggregates. Moreover, hPSCs treated with PVA, or DS or a combination, maintained the pluripotency and were capable of differentiating into all three germ layers. mRNA-seq analysis demonstrated that the combination of PVA and DS significantly promoted hPSC proliferation and prevented cell aggregation through improving energy metabolism-related processes, regulating cell growth, cell proliferation and cell division, as well as reducing the adhesion among hPSC aggregates by affecting expression of genes related to cell adhesion. CONCLUSIONS Our results represent a significant step towards developing a simple and robust approach for the expansion of hPSCs in large scale.
Collapse
Affiliation(s)
- Xianglian Tang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, China.,Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Haibin Wu
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Jinghe Xie
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, China.,Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Ning Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, China.,Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Qicong Chen
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, China.,Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhiyong Zhong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, China.,Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Yaqi Qiu
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Jue Wang
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiajing Li
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ping Situ
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Liangxue Lai
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Mark A Zern
- Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, USA
| | - Honglin Chen
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China.,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China.,Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, China.,Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, China.,Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| | - Yuyou Duan
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China.,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China.,Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, China.,Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, China.,Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| |
Collapse
|
9
|
Manstein F, Ullmann K, Kropp C, Halloin C, Triebert W, Franke A, Farr CM, Sahabian A, Haase A, Breitkreuz Y, Peitz M, Brüstle O, Kalies S, Martin U, Olmer R, Zweigerdt R. High density bioprocessing of human pluripotent stem cells by metabolic control and in silico modeling. Stem Cells Transl Med 2021; 10:1063-1080. [PMID: 33660952 PMCID: PMC8235132 DOI: 10.1002/sctm.20-0453] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 01/18/2021] [Accepted: 01/24/2021] [Indexed: 12/13/2022] Open
Abstract
To harness the full potential of human pluripotent stem cells (hPSCs) we combined instrumented stirred tank bioreactor (STBR) technology with the power of in silico process modeling to overcome substantial, hPSC‐specific hurdles toward their mass production. Perfused suspension culture (3D) of matrix‐free hPSC aggregates in STBRs was applied to identify and control process‐limiting parameters including pH, dissolved oxygen, glucose and lactate levels, and the obviation of osmolality peaks provoked by high density culture. Media supplements promoted single cell‐based process inoculation and hydrodynamic aggregate size control. Wet lab‐derived process characteristics enabled predictive in silico modeling as a new rational for hPSC cultivation. Consequently, hPSC line‐independent maintenance of exponential cell proliferation was achieved. The strategy yielded 70‐fold cell expansion in 7 days achieving an unmatched density of 35 × 106 cells/mL equivalent to 5.25 billion hPSC in 150 mL scale while pluripotency, differentiation potential, and karyotype stability was maintained. In parallel, media requirements were reduced by 75% demonstrating the outstanding increase in efficiency. Minimal input to our in silico model accurately predicts all main process parameters; combined with calculation‐controlled hPSC aggregation kinetics, linear process upscaling is also enabled and demonstrated for up to 500 mL scale in an independent bioreactor system. Thus, by merging applied stem cell research with recent knowhow from industrial cell fermentation, a new level of hPSC bioprocessing is revealed fueling their automated production for industrial and therapeutic applications.
Collapse
Affiliation(s)
- Felix Manstein
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Kevin Ullmann
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Christina Kropp
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Caroline Halloin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Wiebke Triebert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Annika Franke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Clara-Milena Farr
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Anais Sahabian
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Alexandra Haase
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Yannik Breitkreuz
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | - Michael Peitz
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany.,Cell Programming Core Facility, University of Bonn Medical Faculty, Bonn, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | - Stefan Kalies
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany.,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Hannover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Ruth Olmer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| |
Collapse
|
10
|
Qian L, TCW J. Human iPSC-Based Modeling of Central Nerve System Disorders for Drug Discovery. Int J Mol Sci 2021; 22:1203. [PMID: 33530458 PMCID: PMC7865494 DOI: 10.3390/ijms22031203] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/07/2023] Open
Abstract
A high-throughput drug screen identifies potentially promising therapeutics for clinical trials. However, limitations that persist in current disease modeling with limited physiological relevancy of human patients skew drug responses, hamper translation of clinical efficacy, and contribute to high clinical attritions. The emergence of induced pluripotent stem cell (iPSC) technology revolutionizes the paradigm of drug discovery. In particular, iPSC-based three-dimensional (3D) tissue engineering that appears as a promising vehicle of in vitro disease modeling provides more sophisticated tissue architectures and micro-environmental cues than a traditional two-dimensional (2D) culture. Here we discuss 3D based organoids/spheroids that construct the advanced modeling with evolved structural complexity, which propels drug discovery by exhibiting more human specific and diverse pathologies that are not perceived in 2D or animal models. We will then focus on various central nerve system (CNS) disease modeling using human iPSCs, leading to uncovering disease pathogenesis that guides the development of therapeutic strategies. Finally, we will address new opportunities of iPSC-assisted drug discovery with multi-disciplinary approaches from bioengineering to Omics technology. Despite technological challenges, iPSC-derived cytoarchitectures through interactions of diverse cell types mimic patients' CNS and serve as a platform for therapeutic development and personalized precision medicine.
Collapse
Affiliation(s)
- Lu Qian
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Ronald Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Julia TCW
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Ronald Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
11
|
Ghasemian M, Layton C, Nampe D, zur Nieden NI, Tsutsui H, Princevac M. Hydrodynamic characterization within a spinner flask and a rotary wall vessel for stem cell culture. Biochem Eng J 2020. [DOI: 10.1016/j.bej.2020.107533] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
12
|
Jiao Z, Fu C, Li L, Wang Z, Wang Y, Shi X, Zhang P. Microcarriers with Controllable Size via Electrified Liquid Jets and Phase Separation Technique Promote Cell Proliferation and Osteogenic Differentiation. ACS APPLIED BIO MATERIALS 2019; 2:4134-4141. [DOI: 10.1021/acsabm.9b00746] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Zixue Jiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
- University of Chinese Academy of Sciences, Beijing 100039, PR China
| | - Chuan Fu
- Department of Orthopaedic Surgery, The Second Hospital of Jilin University, Changchun 130021, PR China
| | - Linlong Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Zongliang Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Yu Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Xincui Shi
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Peibiao Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
- University of Chinese Academy of Sciences, Beijing 100039, PR China
| |
Collapse
|
13
|
Goto-Silva L, Ayad NME, Herzog IL, Silva NP, Lamien B, Orlande HRB, da Costa Souza A, Ribeiro S, Martins M, Domont GB, Junqueira M, Tovar-Moll F, Rehen SK. Computational fluid dynamic analysis of physical forces playing a role in brain organoid cultures in two different multiplex platforms. BMC DEVELOPMENTAL BIOLOGY 2019; 19:3. [PMID: 30841924 PMCID: PMC6404276 DOI: 10.1186/s12861-019-0183-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 01/31/2019] [Indexed: 12/21/2022]
Abstract
Background Organoid cultivation in suspension culture requires agitation at low shear stress to allow for nutrient diffusion, which preserves tissue structure. Multiplex systems for organoid cultivation have been proposed, but whether they meet similar shear stress parameters as the regularly used spinner flask and its correlation with the successful generation of brain organoids has not been determined. Results Here we used computational fluid dynamics (CFD) to simulate two multiplex culture conditions: steering plates on an orbital shaker and the use of a previously described bioreactor. The bioreactor had low speed and high shear stress regions that may affect cell aggregate growth, depending on volume, whereas the computed variables of the steering plates were closer to those of the spinning flask. Conclusion Our protocol improves the initial steps of the standard brain organoid formation, and the produced organoids displayed regionalized brain structures, including retinal pigmented cells. Overall, we conclude that suspension culture on orbital steering plates is a cost-effective practical alternative to previously described platforms for the cultivation of brain organoids for research and multiplex testing. Electronic supplementary material The online version of this article (10.1186/s12861-019-0183-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Livia Goto-Silva
- D'Or Institute for Research and Education (IDOR), Rua Diniz Cordeiro, 30 - Botafogo, Rio de Janeiro, RJ, 22281-100, Brazil
| | - Nadia M E Ayad
- D'Or Institute for Research and Education (IDOR), Rua Diniz Cordeiro, 30 - Botafogo, Rio de Janeiro, RJ, 22281-100, Brazil
| | - Iasmin L Herzog
- Department of Mechanical Engineering, Politecnica/COPPE - Federal University of Rio de Janeiro, UFRJ, Av. Horácio Macedo, 2030, Cidade Universitária, Rio de Janeiro, RJ, 21941-914, Brazil
| | - Nilton P Silva
- Department of Mechanical Engineering, Politecnica/COPPE - Federal University of Rio de Janeiro, UFRJ, Av. Horácio Macedo, 2030, Cidade Universitária, Rio de Janeiro, RJ, 21941-914, Brazil
| | - Bernard Lamien
- Department of Mechanical Engineering, Politecnica/COPPE - Federal University of Rio de Janeiro, UFRJ, Av. Horácio Macedo, 2030, Cidade Universitária, Rio de Janeiro, RJ, 21941-914, Brazil
| | - Helcio R B Orlande
- Department of Mechanical Engineering, Politecnica/COPPE - Federal University of Rio de Janeiro, UFRJ, Av. Horácio Macedo, 2030, Cidade Universitária, Rio de Janeiro, RJ, 21941-914, Brazil
| | - Annie da Costa Souza
- Brain Institute, Federal University of Rio Grande do Norte, Av. Nascimento de Castro, 2155, Natal, RN, 59056-450, Brazil
| | - Sidarta Ribeiro
- Brain Institute, Federal University of Rio Grande do Norte, Av. Nascimento de Castro, 2155, Natal, RN, 59056-450, Brazil
| | - Michele Martins
- Proteomics Unit, Institute of Chemistry, Federal University of Rio de Janeiro, UFRJ, Av. Athos da Silveira Ramos 149, Rio de Janeiro, 21941-909, Brazil
| | - Gilberto B Domont
- Proteomics Unit, Institute of Chemistry, Federal University of Rio de Janeiro, UFRJ, Av. Athos da Silveira Ramos 149, Rio de Janeiro, 21941-909, Brazil
| | - Magno Junqueira
- Proteomics Unit, Institute of Chemistry, Federal University of Rio de Janeiro, UFRJ, Av. Athos da Silveira Ramos 149, Rio de Janeiro, 21941-909, Brazil
| | - Fernanda Tovar-Moll
- D'Or Institute for Research and Education (IDOR), Rua Diniz Cordeiro, 30 - Botafogo, Rio de Janeiro, RJ, 22281-100, Brazil.,Institute of Biomedical Sciences, Federal University of Rio de Janeiro, UFRJ, Av. Carlos Chagas Filho 373, Bloco K, Cidade Universitária, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Stevens K Rehen
- D'Or Institute for Research and Education (IDOR), Rua Diniz Cordeiro, 30 - Botafogo, Rio de Janeiro, RJ, 22281-100, Brazil. .,Institute of Biomedical Sciences, Federal University of Rio de Janeiro, UFRJ, Av. Carlos Chagas Filho 373, Bloco K, Cidade Universitária, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
14
|
Davis BM, Loghin ER, Conway KR, Zhang X. Automated Closed-System Expansion of Pluripotent Stem Cell Aggregates in a Rocking-Motion Bioreactor. SLAS Technol 2018; 23:364-373. [PMID: 29481762 DOI: 10.1177/2472630318760745] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pluripotent stem cell suspension aggregates have proven to be an efficient and phenotypically stable means for expansion and directed differentiation. Bioreactor systems with automation of perfusion, fluidization, and gas exchange are essential for scaling up pluripotent stem cell cultures. Since stem cell pluripotency and differentiation are affected by both chemical and physical signals, we investigated a low-shear method for the expansion of cells in a rocking-motion bioreactor. The rocking motion drives continual mixing and aeration, and the single-use disposable bioreactors avoid issues around contamination during seeding, medium exchange, passage, and cell harvest. Serial passaging from a 150 mL to a 1 L scale was demonstrated, achieving cell densities of up to 4 million cells/mL. In an average of 13 experiments, pluripotent stem cell aggregates expanded 5.7-fold (with maximal 9.5-fold expansion) and maintained 97% viability over 4 days in a rocking bioreactor culture. In seven experiments with improved culture conditions, the average expansion was 6.8-fold. Maintenance of pluripotency was confirmed by differentiation to all three germ layers and surface marker expression, and the expanded aggregates maintained a stable normal karyotype. The automation associated with the rocking platform bioreactor required no user intervention during the 4-day culture, providing hands-off expansion of pluripotent stem cells.
Collapse
Affiliation(s)
- Brian M Davis
- 1 GE Global Research Center, Biosciences Organization, Niskayuna, NY, USA
| | - Evelina R Loghin
- 1 GE Global Research Center, Biosciences Organization, Niskayuna, NY, USA
| | - Kenneth R Conway
- 1 GE Global Research Center, Biosciences Organization, Niskayuna, NY, USA
| | - Xiaohua Zhang
- 2 GE Global Research Center, Mechanical Systems Organization, Niskayuna, NY, USA
| |
Collapse
|