1
|
Wu X, Yang Y, Pan Y, Wang Y, Lian X, Dong C, Wang S, Wang A, Lei Y. Collagen Nanofiber Reinforced Alginate Hydrogel Tube Microbioreactors for Cell Culture. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.23.650245. [PMID: 40313920 PMCID: PMC12045347 DOI: 10.1101/2025.04.23.650245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
The large-scale production of mammalian cells is pivotal for various applications; however, current bioreactor technologies encounter significant technical and economic challenges. Scaling up cell cultures remains problematic due to excessive cell aggregation, shear stress-induced cell death, batch-to-batch inconsistencies, and limited scalability. We propose that engineering a cell-friendly microenvironment can enhance culture efficiency. Previously, we developed alginate hydrogel microtubes (AlgTubes) that significantly improved cell density and growth rates. Nevertheless, AlgTubes lack adhesion sites essential for anchorage-dependent cells and frequently break, causing cell leakage and production inconsistencies. To address these limitations, this study reinforced AlgTubes with collagen nanofibers, creating collagen-alginate hybrid hydrogel microtubes (ColAlgTubes). Utilizing a novel micro-extruder, we efficiently produced cell-loaded ColAlgTubes. Collagen formed a dense nanofiber network interwoven with the alginate mesh, enhancing the hydrogel's mechanical properties while providing cell adhesion sites. ColAlgTubes protected cells from hydrodynamic stress and maintained cell mass within a 400 μm diameter, ensuring efficient nutrient exchange and waste removal. This optimized microenvironment resulted in high cell viability, rapid proliferation, and exceptional yields of 5×10 8 cells/mL - 200 times higher than conventional culture methods. With their scalability, cost-effectiveness, and efficiency, ColAlgTubes offers a transformative solution for large-scale cell production with broad applications in biotechnology, regenerative medicine, and therapeutic manufacturing.
Collapse
|
2
|
Vicente P, Inocêncio LR, Ullate‐Agote A, Louro AF, Jacinto J, Gamelas B, Iglesias‐García O, Martin‐Uriz PS, Aguirre‐Ruiz P, Ríos‐Muñoz GR, Fernández‐Santos ME, van Mil A, Sluijter JPG, Prósper F, Vega MMM, Alves PM, Serra M. Billion-Scale Expansion of Functional hiPSC-Derived Cardiomyocytes in Bioreactors Through Oxygen Control and Continuous Wnt Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410510. [PMID: 39846380 PMCID: PMC11923921 DOI: 10.1002/advs.202410510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/28/2024] [Indexed: 01/24/2025]
Abstract
Generation of upscaled quantities of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM), for therapeutic or testing applications, is both expensive and time-consuming. Herein, a scalable bioprocess for hiPSC-CM expansion in stirred-tank bioreactors (STB) is developed. By combining the continuous activation of the Wnt pathway, through perfusion of CHIR99021, within a mild hypoxia environment, the expansion of hiPSC-CM as aggregates is maximized, reaching 4 billion of pure hiPSC-CM in 2L STB. In particular, the importance of i) controlling the dissolved oxygen at 10% O2 to reduce reactive oxygen species production and upregulate genes involved in cell proliferation, resulting in higher expansion rates (tenfold) compared to normoxic conditions, and ii) maintaining constant power input per volume as a scale-up criteria is demonstrated. After expansion, hiPSC-CM further mature in culture, revealing more mature transcriptional signatures, higher sarcomere alignment and improved calcium handling. This new bioprocess opens the door to time- and cost-effective generation of hiPSC-CM.
Collapse
Affiliation(s)
- Pedro Vicente
- iBETInstituto de Biologia Experimental e TecnológicaApartado 12Oeiras2780901Portugal
- Instituto de Tecnologia Química e Biológica António XavierUniversidade de Nova de LisboaAv. da RepúblicaOeiras2780157Portugal
| | - Lara R. Inocêncio
- iBETInstituto de Biologia Experimental e TecnológicaApartado 12Oeiras2780901Portugal
- Instituto de Tecnologia Química e Biológica António XavierUniversidade de Nova de LisboaAv. da RepúblicaOeiras2780157Portugal
| | - Asier Ullate‐Agote
- Program of Biomedical EngineeringTechnological Innovation DivisionCIMA Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA)Pamplona31008Spain
| | - Ana F. Louro
- iBETInstituto de Biologia Experimental e TecnológicaApartado 12Oeiras2780901Portugal
- Instituto de Tecnologia Química e Biológica António XavierUniversidade de Nova de LisboaAv. da RepúblicaOeiras2780157Portugal
| | - João Jacinto
- iBETInstituto de Biologia Experimental e TecnológicaApartado 12Oeiras2780901Portugal
- Instituto de Tecnologia Química e Biológica António XavierUniversidade de Nova de LisboaAv. da RepúblicaOeiras2780157Portugal
| | - Beatriz Gamelas
- iBETInstituto de Biologia Experimental e TecnológicaApartado 12Oeiras2780901Portugal
- Instituto de Tecnologia Química e Biológica António XavierUniversidade de Nova de LisboaAv. da RepúblicaOeiras2780157Portugal
| | - Olalla Iglesias‐García
- Program of Biomedical EngineeringTechnological Innovation DivisionCIMA Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA)Pamplona31008Spain
| | - Patxi San Martin‐Uriz
- Hemato‐oncology ProgramCancer DivisionCIMA Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA)Pamplona31008Spain
| | - Paula Aguirre‐Ruiz
- Hemato‐oncology ProgramCancer DivisionCIMA Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA)Pamplona31008Spain
| | - Gonzalo R. Ríos‐Muñoz
- Bioengineering DepartmentUniversidad Carlos III de MadridMadrid28911Spain
- Department of CardiologyGregorio Marañón Health Research Institute (IiSGM)Hospital General Universitario Gregorio MarañónMadrid28007Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Instituto Carlos IIIMadrid28029Spain
| | - María Eugenia Fernández‐Santos
- Department of CardiologyGregorio Marañón Health Research Institute (IiSGM)Hospital General Universitario Gregorio MarañónMadrid28007Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Instituto Carlos IIIMadrid28029Spain
| | - Alain van Mil
- Regenerative Medicine Center UtrechtLaboratory of Experimental Cardiology, Circulatory Health Research Center, University UtrechtUniversity Medical Center UtrechtUniversity UtrechtUtrecht3508 GAThe Netherlands
| | - Joost P. G. Sluijter
- Regenerative Medicine Center UtrechtLaboratory of Experimental Cardiology, Circulatory Health Research Center, University UtrechtUniversity Medical Center UtrechtUniversity UtrechtUtrecht3508 GAThe Netherlands
| | - Felipe Prósper
- Hematology and Cell TherapyClínica Universidad de NavarraPamplona31008Spain
- Hemato‐oncology ProgramCancer DivisionCIMA Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA)Pamplona31008Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)Madrid28029Spain
| | - Manuel M Mazo Vega
- Program of Biomedical EngineeringTechnological Innovation DivisionCIMA Universidad de Navarra, and Instituto de Investigación Sanitaria de Navarra (IdiSNA)Pamplona31008Spain
- Hematology and Cell TherapyClínica Universidad de NavarraPamplona31008Spain
| | - Paula M. Alves
- iBETInstituto de Biologia Experimental e TecnológicaApartado 12Oeiras2780901Portugal
- Instituto de Tecnologia Química e Biológica António XavierUniversidade de Nova de LisboaAv. da RepúblicaOeiras2780157Portugal
| | - Margarida Serra
- iBETInstituto de Biologia Experimental e TecnológicaApartado 12Oeiras2780901Portugal
- Instituto de Tecnologia Química e Biológica António XavierUniversidade de Nova de LisboaAv. da RepúblicaOeiras2780157Portugal
| |
Collapse
|
3
|
Yang Y, Wu X, Pan Y, Wang Y, Lian X, Dong C, Liu W, Wang S, Lei Y. Collagen Hydrogel Tube Microbioreactors for Cell and Tissue Manufacturing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631570. [PMID: 39829742 PMCID: PMC11741382 DOI: 10.1101/2025.01.08.631570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The production of mammalian cells in large quantities is essential for various applications. However, scaling up cell culture using existing bioreactors poses significant technical challenges and high costs. To address this, we previously developed an innovative 3D culture system, known as the AlgTube cell culture system, for high-density cell cultivation. This system involves processing cells into microscale alginate hydrogel tubes, which are suspended in the culture medium within a vessel. These hydrogel tubes shield cells from hydrodynamic stress and maintain the cell mass below 400 µm in diameter, facilitating efficient mass transport and creating a favorable microenvironment for cell growth. Under optimized conditions, AlgTubes supported long-term culture with high cell viability, rapid expansion (1000-fold increase over 9 days per passage), and high yield (5×10⁸ cells/mL), offering significant advantages over conventional methods. Despite these benefits, AlgTubes have critical drawbacks. They are mechanically fragile, with frequent breakage during culture leading to cell leakage and production failures. Additionally, many cell types exhibit poor growth due to the inability to adhere to the alginate surface, making alginate hydrogel microtubes unsuitable for industrial-scale cell production. To overcome these challenges, we developed a novel collagen hydrogel tube-based microbioreactor system in this work. This system provides enhanced robustness and adhesion, enabling scalable, cost-effective, and efficient cell production for a wide range of applications.
Collapse
|
4
|
Hashemi M, Finklea FB, Hammons H, Tian Y, Young N, Kim E, Halloin C, Triebert W, Zweigerdt R, Mitra AK, Lipke EA. Hydrogel microsphere stem cell encapsulation enhances cardiomyocyte differentiation and functionality in scalable suspension system. Bioact Mater 2025; 43:423-440. [PMID: 39399838 PMCID: PMC11471139 DOI: 10.1016/j.bioactmat.2024.08.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 10/15/2024] Open
Abstract
A reliable suspension-based platform for scaling engineered cardiac tissue (ECT) production from human induced pluripotent stem cells (hiPSCs) is crucial for regenerative therapies. Here, we compared the production and functionality of ECTs formed using our scaffold-based, engineered tissue microsphere differentiation approach with those formed using the prevalent scaffold-free aggregate platform. We utilized a microfluidic system for the rapid (1 million cells/min), high density (30, 40, 60 million cells/ml) encapsulation of hiPSCs within PEG-fibrinogen hydrogel microspheres. HiPSC-laden microspheres and aggregates underwent suspension-based cardiac differentiation in chemically defined media. In comparison to aggregates, microspheres maintained consistent size and shape initially, over time, and within and between batches. Initial size and shape coefficients of variation for microspheres were eight and three times lower, respectively, compared to aggregates. On day 10, microsphere cardiomyocyte (CM) content was 27 % higher and the number of CMs per initial hiPSC was 250 % higher than in aggregates. Contraction and relaxation velocities of microspheres were four and nine times higher than those of aggregates, respectively. Microsphere contractile functionality also improved with culture time, whereas aggregate functionality remained unchanged. Additionally, microspheres displayed improved β-adrenergic signaling responsiveness and uniform calcium transient propagation. Transcriptomic analysis revealed that while both microspheres and aggregates demonstrated similar gene regulation patterns associated with cardiomyocyte differentiation, heart development, cardiac muscle contraction, and sarcomere organization, the microspheres exhibited more pronounced transcriptional changes over time. Taken together, these results highlight the capability of the microsphere platform for scaling up biomanufacturing of ECTs in a suspension-based culture platform.
Collapse
Affiliation(s)
| | - Ferdous B. Finklea
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Hanna Hammons
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Yuan Tian
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Nathan Young
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Emma Kim
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Caroline Halloin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Wiebke Triebert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Amit Kumar Mitra
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, United States
| | - Elizabeth A. Lipke
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| |
Collapse
|
5
|
Gwon K, Dharmesh E, Nguyen KM, Schornack AMR, de Hoyos-Vega JM, Ceylan H, Stybayeva G, Peterson QP, Revzin A. Designing magnetic microcapsules for cultivation and differentiation of stem cell spheroids. MICROSYSTEMS & NANOENGINEERING 2024; 10:127. [PMID: 39261472 PMCID: PMC11390961 DOI: 10.1038/s41378-024-00747-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/03/2024] [Accepted: 06/05/2024] [Indexed: 09/13/2024]
Abstract
Human pluripotent stem cells (hPSCs) represent an excellent cell source for regenerative medicine and tissue engineering applications. However, there remains a need for robust and scalable differentiation of stem cells into functional adult tissues. In this paper, we sought to address this challenge by developing magnetic microcapsules carrying hPSC spheroids. A co-axial flow-focusing microfluidic device was employed to encapsulate stem cells in core-shell microcapsules that also contained iron oxide magnetic nanoparticles (MNPs). These microcapsules exhibited excellent response to an external magnetic field and could be held at a specific location. As a demonstration of utility, magnetic microcapsules were used for differentiating hPSC spheroids as suspension cultures in a stirred bioreactor. Compared to standard suspension cultures, magnetic microcapsules allowed for more efficient media change and produced improved differentiation outcomes. In the future, magnetic microcapsules may enable better and more scalable differentiation of hPSCs into adult cell types and may offer benefits for cell transplantation.
Collapse
Affiliation(s)
- Kihak Gwon
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
- Department of Biofibers and Biomaterials Science, Kyungpook National University, Daegu, Republic of Korea.
| | - Ether Dharmesh
- Department of Biomedical Engineering, Saint Louis University, St. Louis, MO, USA
| | - Kianna M Nguyen
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | | | - Jose M de Hoyos-Vega
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Hakan Ceylan
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Scottsdale, AZ, USA
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Quinn P Peterson
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
6
|
Suong DNA, Imamura K, Kato Y, Inoue H. Design of neural organoids engineered by mechanical forces. IBRO Neurosci Rep 2024; 16:190-195. [PMID: 38328799 PMCID: PMC10847990 DOI: 10.1016/j.ibneur.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/19/2024] [Indexed: 02/09/2024] Open
Abstract
Neural organoids consist of three-dimensional tissue derived from pluripotent stem cells that could recapitulate key features of the human brain. During the past decade, organoid technology has evolved in the field of human brain science by increasing the quality and applicability of its products. Among them, a novel approach involving the design of neural organoids engineered by mechanical forces has emerged. This review describes previous approaches for the generation of neural organoids, the engineering of neural organoids by mechanical forces, and future challenges for the application of mechanical forces in the design of neural organoids.
Collapse
Affiliation(s)
- Dang Ngoc Anh Suong
- iPSC‑Based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Keiko Imamura
- iPSC‑Based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
- 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
| | - Yoshikazu Kato
- Mixing Technology Laboratory, SATAKE MultiMix Corporation, Saitama, Japan
| | - Haruhisa Inoue
- iPSC‑Based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
- 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
| |
Collapse
|
7
|
Wang K, Xie W, Harcum SW. Metabolic regulatory network kinetic modeling with multiple isotopic tracers for iPSCs. Biotechnol Bioeng 2024; 121:1336-1354. [PMID: 38037741 DOI: 10.1002/bit.28609] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/26/2023] [Accepted: 11/15/2023] [Indexed: 12/02/2023]
Abstract
The rapidly expanding market for regenerative medicines and cell therapies highlights the need to advance the understanding of cellular metabolisms and improve the prediction of cultivation production process for human induced pluripotent stem cells (iPSCs). In this paper, a metabolic kinetic model was developed to characterize the underlying mechanisms of iPSC culture process, which can predict cell response to environmental perturbation and support process control. This model focuses on the central carbon metabolic network, including glycolysis, pentose phosphate pathway, tricarboxylic acid cycle, and amino acid metabolism, which plays a crucial role to support iPSC proliferation. Heterogeneous measures of extracellular metabolites and multiple isotopic tracers collected under multiple conditions were used to learn metabolic regulatory mechanisms. Systematic cross-validation confirmed the model's performance in terms of providing reliable predictions on cellular metabolism and culture process dynamics under various culture conditions. Thus, the developed mechanistic kinetic model can support process control strategies to strategically select optimal cell culture conditions at different times, ensure cell product functionality, and facilitate large-scale manufacturing of regenerative medicines and cell therapies.
Collapse
Affiliation(s)
- Keqi Wang
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Wei Xie
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Sarah W Harcum
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
8
|
Zheng H, Harcum SW, Pei J, Xie W. Stochastic biological system-of-systems modelling for iPSC culture. Commun Biol 2024; 7:39. [PMID: 38191636 PMCID: PMC10774284 DOI: 10.1038/s42003-023-05653-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 11/30/2023] [Indexed: 01/10/2024] Open
Abstract
Large-scale manufacturing of induced pluripotent stem cells (iPSCs) is essential for cell therapies and regenerative medicines. Yet, iPSCs form large cell aggregates in suspension bioreactors, resulting in insufficient nutrient supply and extra metabolic waste build-up for the cells located at the core. Since subtle changes in micro-environment can lead to a heterogeneous cell population, a novel Biological System-of-Systems (Bio-SoS) framework is proposed to model cell-to-cell interactions, spatial and metabolic heterogeneity, and cell response to micro-environmental variation. Building on stochastic metabolic reaction network, aggregation kinetics, and reaction-diffusion mechanisms, the Bio-SoS model characterizes causal interdependencies at individual cell, aggregate, and cell population levels. It has a modular design that enables data integration and improves predictions for different monolayer and aggregate culture processes. In addition, a variance decomposition analysis is derived to quantify the impact of factors (i.e., aggregate size) on cell product health and quality heterogeneity.
Collapse
Affiliation(s)
- Hua Zheng
- Mechanical and Industrial Engineering, Northeastern University, Boston, MA, 02115, USA
| | | | - Jinxiang Pei
- Mechanical and Industrial Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Wei Xie
- Mechanical and Industrial Engineering, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
9
|
Alsobaie S, Alsobaie T, Alshammary AF, Abudawood M, Mantalaris A. Alginate Beads as a Promising Tool for Successful Production of Viable and Pluripotent Human-Induced Pluripotent Stem Cells in a 3D Culture System. Stem Cells Cloning 2023; 16:61-73. [PMID: 37790697 PMCID: PMC10544263 DOI: 10.2147/sccaa.s409139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/13/2023] [Indexed: 10/05/2023] Open
Abstract
Purpose Two-dimensional (2D)-based cell culture systems, limited by their inherent heterogeneity and scalability, are a bottleneck in the production of high-quality cells for downstream biomedical applications. Finding the optimal conditions for large-scale stem cell culture while maintaining good cellular status is challenging. The aim of this study was to assess the effects of three-dimensional (3D) culture on the viability, proliferation, self-renewal, and differentiation of human induced pluripotent stem cells (IPSCs). Patients and Methods Various culture conditions were evaluated to determine the optimal conditions to maintain the viability and proliferation of human IPSCs in a 3D environment: static versus dynamic culture, type of adhesion protein added to alginate (Matrigel™ versus gelatin), and the addition of Y-27632t on long-term 3D culture. The proliferation ability of the cells was evaluated via the MTS proliferation assay; the expression levels of the pluripotency markers Nanog and Oct3/4, PAX6 as an ectoderm marker, and laminin-5 and fibronectin as markers of extracellular matrix synthesis were assessed; and HIF1α and HIF2α levels were measured using quantitative reverse transcription polymerase chain reaction. Results Using a high-aspect-ratio vessel bioreactor with a gentle, low-sheer, and low-turbulence environment with sufficient oxygenation and effective mass transfer of nutrients and waste, we verified its ability to promote cell proliferation and self-renewal. The findings showed that human IPSCs have the ability to maintain pluripotency in a feeder-free system and by inhibiting ROCK signaling and using hypoxia to improve single-cell viability in 3D culture. Furthermore, these cells demonstrated increased self-renewal and proliferation when inoculated as single cells in 3D alginate beads by adding RI during the culture period. Conclusion Dynamic 3D culture is desirable for the large-scale expansion of undifferentiated human IPSCs.
Collapse
Affiliation(s)
- Sarah Alsobaie
- Department of Clinical Laboratory Science, King Saud University, Riyadh, Saudi Arabia
| | - Tamador Alsobaie
- Biological Systems Engineering Laboratory, Department of Chemical Engineering, Imperial College London, London, UK
| | - Amal F Alshammary
- Department of Clinical Laboratory Science, King Saud University, Riyadh, Saudi Arabia
| | - Manal Abudawood
- Department of Clinical Laboratory Science, King Saud University, Riyadh, Saudi Arabia
| | - Athanasios Mantalaris
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
10
|
Cohen PJR, Luquet E, Pletenka J, Leonard A, Warter E, Gurchenkov B, Carrere J, Rieu C, Hardouin J, Moncaubeig F, Lanero M, Quelennec E, Wurtz H, Jamet E, Demarco M, Banal C, Van Liedekerke P, Nassoy P, Feyeux M, Lefort N, Alessandri K. Engineering 3D micro-compartments for highly efficient and scale-independent expansion of human pluripotent stem cells in bioreactors. Biomaterials 2023; 295:122033. [PMID: 36764194 DOI: 10.1016/j.biomaterials.2023.122033] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 12/12/2022] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Human pluripotent stem cells (hPSCs) have emerged as the most promising cellular source for cell therapies. To overcome the scale-up limitations of classical 2D culture systems, suspension cultures have been developed to meet the need for large-scale culture in regenerative medicine. Despite constant improvements, current protocols that use microcarriers or generate cell aggregates only achieve moderate amplification performance. Here, guided by reports showing that hPSCs can self-organize in vitro into cysts reminiscent of the epiblast stage in embryo development, we developed a physio-mimetic approach for hPSC culture. We engineered stem cell niche microenvironments inside microfluidics-assisted core-shell microcapsules. We demonstrate that lumenized three-dimensional colonies significantly improve viability and expansion rates while maintaining pluripotency compared to standard hPSC culture platforms such as 2D cultures, microcarriers, and aggregates. By further tuning capsule size and culture conditions, we scale up this method to industrial-scale stirred tank bioreactors and achieve an unprecedented hPSC amplification rate of 277-fold in 6.5 days. In brief, our findings indicate that our 3D culture system offers a suitable strategy both for basic stem cell biology experiments and for clinical applications.
Collapse
Affiliation(s)
- Philippe J R Cohen
- Université Paris Cité, Imagine Institute, IPSC Core Facility, INSERM UMR U1163, F-75015, Paris, France; Treefrog Therapeutics, F-33600, Pessac, France.
| | | | | | | | | | | | | | | | | | | | | | - Eddy Quelennec
- Université Paris Cité, Imagine Institute, IPSC Core Facility, INSERM UMR U1163, F-75015, Paris, France; Treefrog Therapeutics, F-33600, Pessac, France
| | | | | | | | - Celine Banal
- Université Paris Cité, Imagine Institute, IPSC Core Facility, INSERM UMR U1163, F-75015, Paris, France
| | - Paul Van Liedekerke
- Inria Paris & Sorbonne Université LJLL, 2 Rue Simone IFF, F-75012, Paris, France
| | - Pierre Nassoy
- LP2N, Laboratoire Photonique Numérique et Nanosciences, Univ. Bordeaux, F-33400, Talence, France; Institut D'Optique Graduate School & CNRS UMR 5298, F-33400, Talence, France
| | | | - Nathalie Lefort
- Université Paris Cité, Imagine Institute, IPSC Core Facility, INSERM UMR U1163, F-75015, Paris, France
| | | |
Collapse
|
11
|
Zong RQ, Zhang HY, Li XY, Li YR, Chen Y. Overexpressed Histocompatibility Minor 13 was Associated with Liver Hepatocellular Carcinoma Progression and Prognosis. Genet Res (Camb) 2022; 2022:7067743. [PMID: 36262249 PMCID: PMC9550386 DOI: 10.1155/2022/7067743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 12/03/2022] Open
Abstract
Among primary liver carcinoma cases, the proportion of liver hepatocellular carcinoma (LIHC) cases is 75%-85%. Current treatments for LIHC include chemotherapy, surgical excision, and liver transplantation, which are effective for early LIHC treatment. Nevertheless, the early symptoms of liver carcinoma are atypical, so a large proportion of LIHC patients are diagnosed at an advanced stage. Histocompatibility minor 13 (HM13), located in the endoplasmic reticulum, is responsible for catalysing the hydrolysis of some signal peptides after cleavage from the precursor protein. Here, we studied the role of HM13 in LIHC development through bioinformatics analysis. Database analysis showed that HM13 was of great significance for LIHC tumorigenesis. Compared to normal liver tissues, HM13 expression was increased to a greater extent in LIHC tissues. After analysis of Kaplan‒Meier plotter and Gene Expression Profiling Interactive Analysis (GEPIA) datasets, we discovered that highly expressed HM13 exhibited an association with shorter overall survival (OS), disease-free survival (DFS), and disease-specific survival (DSS). We conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses to analyse HM13-related genes, and the data indicated that these genes obviously participated in rRNA processing, ribosome biogenesis, spliceosome, Huntington's disease, and ATP-dependent helicase activity. The Cell Counting Kit-8 (CCK-8) assay and Transwell assay showed that reducing HM13 expression hindered LIHC cell proliferation, migration, and invasion. In conclusion, these findings indicate that HM13 is a biomarker and is related to the poor prognosis of LIHC. Our results are conducive to discovering new targets for LIHC treatment.
Collapse
Affiliation(s)
- Rui-Qing Zong
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Intensive Care Medicine, Eastern Hepatobiliary Surgery Hospital, The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Hong-Yan Zhang
- Department of Intensive Care Medicine, Eastern Hepatobiliary Surgery Hospital, The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiao-Ying Li
- Department of Intensive Care Medicine, Eastern Hepatobiliary Surgery Hospital, The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yi-ran Li
- Department of Intensive Care Medicine, Eastern Hepatobiliary Surgery Hospital, The Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Ying Chen
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Dai Y, Cui X, Zhang G, Mohsin A, Xu H, Zhuang Y, Guo M. Development of a novel feeding regime for large scale production of human umbilical cord mesenchymal stem/stromal cells. Cytotechnology 2022; 74:351-369. [DOI: 10.1007/s10616-022-00523-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 01/23/2022] [Indexed: 12/21/2022] Open
|
13
|
Miranda CC, Akenhead ML, Silva TP, Derr MA, Vemuri MC, Cabral JMS, Fernandes TG. A Dynamic 3D Aggregate-Based System for the Successful Expansion and Neural Induction of Human Pluripotent Stem Cells. Front Cell Neurosci 2022; 16:838217. [PMID: 35308123 PMCID: PMC8928726 DOI: 10.3389/fncel.2022.838217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/04/2022] [Indexed: 11/13/2022] Open
Abstract
The demand for large cell numbers for cellular therapies and drug screening applications requires the development of scalable platforms capable of generating high-quality populations of tissue-specific cells derived from human pluripotent stem cells (hPSCs). Here, we studied the ability of Gibco StemScale PSC Suspension Medium to promote the efficient expansion of hPSC cultures as aggregates grown in suspension. We tested human induced pluripotent stem cell (hiPSC) growth in 6-well plates (on orbital shaker platforms) and single-use vertical-wheel bioreactors for a total of three consecutive passages. Up to a 9-fold increase in cell number was observed over 5 days per passage, with a cumulative fold change up to 600 in 15 days. Additionally, we compared neural induction of hiPSCs by using a dual SMAD inhibition protocol with a commercially available neural induction medium, which can potentially yield more than a 30-fold change, including neural progenitor induction and expansion. This system can also be adapted toward the generation of floor plate progenitors, which yields up to an 80-fold change in cell number and generates FOXA2-positive populations. In summary, we developed platforms for hiPSC expansion and neural induction into different brain regions that provide scalability toward producing clinically relevant cell numbers.
Collapse
Affiliation(s)
- Cláudia C. Miranda
- Department of Bioengineering and iBB – Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Michael L. Akenhead
- Thermo Fisher Scientific, Cell Biology, Life Sciences Solutions, Frederick, MD, United States
| | - Teresa P. Silva
- Department of Bioengineering and iBB – Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Michael A. Derr
- Thermo Fisher Scientific, Cell Biology, Life Sciences Solutions, Frederick, MD, United States
| | - Mohan C. Vemuri
- Thermo Fisher Scientific, Cell Biology, Life Sciences Solutions, Frederick, MD, United States
| | - Joaquim M. S. Cabral
- Department of Bioengineering and iBB – Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Tiago G. Fernandes
- Department of Bioengineering and iBB – Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- *Correspondence: Tiago G. Fernandes,
| |
Collapse
|
14
|
Lee B, Jung S, Hashimura Y, Lee M, Borys BS, Dang T, Kallos MS, Rodrigues CAV, Silva TP, Cabral JMS. Cell Culture Process Scale-Up Challenges for Commercial-Scale Manufacturing of Allogeneic Pluripotent Stem Cell Products. Bioengineering (Basel) 2022; 9:bioengineering9030092. [PMID: 35324781 PMCID: PMC8945133 DOI: 10.3390/bioengineering9030092] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 01/01/2023] Open
Abstract
Allogeneic cell therapy products, such as therapeutic cells derived from pluripotent stem cells (PSCs), have amazing potential to treat a wide variety of diseases and vast numbers of patients globally. However, there are various challenges related to manufacturing PSCs in single-use bioreactors, particularly at larger volumetric scales. This manuscript addresses these challenges and presents potential solutions to alleviate the anticipated bottlenecks for commercial-scale manufacturing of high-quality therapeutic cells derived from PSCs.
Collapse
Affiliation(s)
- Brian Lee
- PBS Biotech, Inc., Camarillo, CA 93012, USA; (S.J.); (Y.H.); (M.L.); (B.S.B.)
- Correspondence:
| | - Sunghoon Jung
- PBS Biotech, Inc., Camarillo, CA 93012, USA; (S.J.); (Y.H.); (M.L.); (B.S.B.)
| | - Yas Hashimura
- PBS Biotech, Inc., Camarillo, CA 93012, USA; (S.J.); (Y.H.); (M.L.); (B.S.B.)
| | - Maximilian Lee
- PBS Biotech, Inc., Camarillo, CA 93012, USA; (S.J.); (Y.H.); (M.L.); (B.S.B.)
| | - Breanna S. Borys
- PBS Biotech, Inc., Camarillo, CA 93012, USA; (S.J.); (Y.H.); (M.L.); (B.S.B.)
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB T2N 1N4, Canada; (T.D.); (M.S.K.)
| | - Tiffany Dang
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB T2N 1N4, Canada; (T.D.); (M.S.K.)
| | - Michael S. Kallos
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB T2N 1N4, Canada; (T.D.); (M.S.K.)
| | - Carlos A. V. Rodrigues
- iBB–Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal; (C.A.V.R.); (T.P.S.); (J.M.S.C.)
| | - Teresa P. Silva
- iBB–Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal; (C.A.V.R.); (T.P.S.); (J.M.S.C.)
| | - Joaquim M. S. Cabral
- iBB–Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal; (C.A.V.R.); (T.P.S.); (J.M.S.C.)
| |
Collapse
|
15
|
Nebel S, Lux M, Kuth S, Bider F, Dietrich W, Egger D, Boccaccini AR, Kasper C. Alginate Core-Shell Capsules for 3D Cultivation of Adipose-Derived Mesenchymal Stem Cells. Bioengineering (Basel) 2022; 9:66. [PMID: 35200419 PMCID: PMC8869374 DOI: 10.3390/bioengineering9020066] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are primary candidates in tissue engineering and stem cell therapies due to their intriguing regenerative and immunomodulatory potential. Their ability to self-assemble into three-dimensional (3D) aggregates further improves some of their therapeutic properties, e.g., differentiation potential, secretion of cytokines, and homing capacity after administration. However, high hydrodynamic shear forces and the resulting mechanical stresses within commercially available dynamic cultivation systems can decrease their regenerative properties. Cells embedded within a polymer matrix, however, lack cell-to-cell interactions found in their physiological environment. Here, we present a "semi scaffold-free" approach to protect the cells from high shear forces by a physical barrier, but still allow formation of a 3D structure with in vivo-like cell-to-cell contacts. We highlight a relatively simple method to create core-shell capsules by inverse gelation. The capsules consist of an outer barrier made from sodium alginate, which allows for nutrient and waste diffusion and an inner compartment for direct cell-cell interactions. Next to capsule characterization, a harvesting procedure was established and viability and proliferation of human adipose-derived MSCs were investigated. In the future, this encapsulation and cultivation technique might be used for MSC-expansion in scalable dynamic bioreactor systems, facilitating downstream procedures, such as cell harvest and differentiation into mature tissue grafts.
Collapse
Affiliation(s)
- Sabrina Nebel
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences BOKU Vienna, 1190 Vienna, Austria; (S.N.); (M.L.); (D.E.)
| | - Manuel Lux
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences BOKU Vienna, 1190 Vienna, Austria; (S.N.); (M.L.); (D.E.)
| | - Sonja Kuth
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (S.K.); (F.B.); (A.R.B.)
| | - Faina Bider
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (S.K.); (F.B.); (A.R.B.)
| | - Wolf Dietrich
- Department of Gynecology and Obstetrics, Karl Landsteiner University of Health Sciences, Alter Ziegelweg 10, 3430 Tulln, Austria;
| | - Dominik Egger
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences BOKU Vienna, 1190 Vienna, Austria; (S.N.); (M.L.); (D.E.)
| | - Aldo R. Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (S.K.); (F.B.); (A.R.B.)
| | - Cornelia Kasper
- Institute of Cell and Tissue Culture Technologies, Department of Biotechnology, University of Natural Resources and Life Sciences BOKU Vienna, 1190 Vienna, Austria; (S.N.); (M.L.); (D.E.)
| |
Collapse
|
16
|
Matthys OB, McDevitt TC. Self-Assembled Heterotypic Cardiac Spheroids from Human Pluripotent Stem Cells. Methods Mol Biol 2022; 2485:39-53. [PMID: 35618897 DOI: 10.1007/978-1-0716-2261-2_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Engineered cardiac tissue models aim to recapitulate the multicellular composition of the native myocardium by incorporating multiple tissue-relevant cell populations. Here, we describe the process of generating self-assembled cardiac microtissue spheroids comprised of heterotypic cardiac cell types. The absence of exogenous extracellular matrix (ECM) or scaffolding makes microtissue assembly dependent upon intercellular adhesion interactions over cell-ECM interactions, analogous to early development. Therefore, this approach creates a 3D platform to study how multicellular heterotypic interactions impact tissue structure, function, and phenotype.
Collapse
Affiliation(s)
- Oriane B Matthys
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, San Francisco, CA, USA
- Gladstone Institutes, San Francisco, CA, USA
| | - Todd C McDevitt
- Gladstone Institutes, San Francisco, CA, USA.
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA.
| |
Collapse
|
17
|
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
|
18
|
Kinney MA. Finding the volume dial in stem cell manufacturing: Bioinspired and bioengineered approaches to scale up. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021. [DOI: 10.1016/j.cobme.2021.100356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
19
|
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
|
20
|
|
21
|
Dang T, Borys BS, Kanwar S, Colter J, Worden H, Blatchford A, Croughan MS, Hossan T, Rancourt DE, Lee B, Kallos MS, Jung S. Computational fluid dynamic characterization of vertical‐wheel bioreactors used for effective scale‐up of human induced pluripotent stem cell aggregate culture. CAN J CHEM ENG 2021. [DOI: 10.1002/cjce.24253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Tiffany Dang
- 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
| | - 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
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering University of Calgary Calgary Alberta Canada
- PBS Biotech Inc. Camarillo California USA
| | - Shivek Kanwar
- Pharmaceutical Production Research Facility, Schulich School of Engineering University of Calgary Calgary Alberta Canada
- Department of Mechanical and Manufacturing Engineering, Schulich School of Engineering University of Calgary Calgary Alberta Canada
| | - James Colter
- 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
| | | | | | | | - Tareq Hossan
- Department of Biochemistry and Molecular Biology, 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
| | - Brian Lee
- PBS Biotech Inc. Camarillo California USA
| | - 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
|
22
|
Petry F, Salzig D. Impact of Bioreactor Geometry on Mesenchymal Stem Cell Production in Stirred‐Tank Bioreactors. CHEM-ING-TECH 2021. [DOI: 10.1002/cite.202100041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Florian Petry
- University of Applied Sciences Mittelhessen Institute of Bioprocess Engineering and Pharmaceutical Technology Wiesenstraße 14 35390 Giessen Germany
| | - Denise Salzig
- University of Applied Sciences Mittelhessen Institute of Bioprocess Engineering and Pharmaceutical Technology Wiesenstraße 14 35390 Giessen Germany
| |
Collapse
|
23
|
Ajiteru O, Choi KY, Lim TH, Kim DY, Hong H, Lee YJ, Lee JS, Lee H, Suh YJ, Sultan MT, Lee OJ, Kim SH, Park CH. A digital light processing 3D printed magnetic bioreactor system using silk magnetic bioink. Biofabrication 2021; 13. [PMID: 33887719 DOI: 10.1088/1758-5090/abfaee] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/22/2021] [Indexed: 12/13/2022]
Abstract
Among various bioreactors used in the field of tissue engineering and regenerative medicine, a magnetic bioreactor is more capable of providing steady force to the cells while avoiding direct manipulation of the materials. However, most of them are complex and difficult to fabricate, with drawbacks in terms of consistency and biocompatibility. In this study, a magnetic bioreactor system and a magnetic hydrogel were manufactured by single-stage three-dimensional (3D) printing with digital light processing (DLP) technique for differentiation of myoblast cells. The hydrogel was composed of a magnetic part containing iron oxide and glycidyl-methacrylated silk fibroin, and a cellular part printed by adding mouse myoblast cell (C2C12) to gelatin glycidyl methacrylate, that was placed in the magnetic bioreactor system to stimulate the cells in the hydrogel. The composite hydrogel was steadily printed by a one-stage layering technique using a DLP printer. The magnetic bioreactor offered mechanical stretching of the cells in the hydrogel in 3D ways, so that the cellular differentiation could be executed in three dimensions just like the human environment. Cell viability, as well as gene expression using quantitative reverse transcription-polymerase chain reaction, were assessed after magneto-mechanical stimulation of the myoblast cell-embedded hydrogel in the magnetic bioreactor system. Comparison with the control group revealed that the magnetic bioreactor system accelerated differentiation of mouse myoblast cells in the hydrogel and increased myotube diameter and lengthin vitro. The DLP-printed magnetic bioreactor and the hydrogel were simply manufactured and easy-to-use, providing an efficient environment for applying noninvasive mechanical force via FDA-approved silk fibroin and iron oxide biocomposite hydrogel, to stimulate cells without any evidence of cytotoxicity, demonstrating the potential for application in muscle tissue engineering.
Collapse
Affiliation(s)
- Olatunji Ajiteru
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Kyu Young Choi
- Department of Otorhinolaryngology-Head and Neck Surgery, Hallym University College of Medicine, Kangnam Sacred Heart Hospital, Seoul 07441, Republic of Korea
| | - Tae Hyeon Lim
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Do Yeon Kim
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Heesun Hong
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Young Jin Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Ji Seung Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Hanna Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Ye Ji Suh
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Md Tipu Sultan
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Ok Joo Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Soon Hee Kim
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Chan Hum Park
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, School of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
24
|
Wang Z, Zuo F, Liu Q, Wu X, Du Q, Lei Y, Wu Z, Lin H. Comparative Study of Human Pluripotent Stem Cell-Derived Endothelial Cells in Hydrogel-Based Culture Systems. ACS OMEGA 2021; 6:6942-6952. [PMID: 33748608 PMCID: PMC7970572 DOI: 10.1021/acsomega.0c06187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 02/19/2021] [Indexed: 06/12/2023]
Abstract
Human pluripotent stem cell (hPSC)-derived endothelial cells (ECs) are promising cell sources for drug discovery, tissue engineering, and studying or treating vascular diseases. However, hPSC-ECs derived from different culture methods display different phenotypes. Herein, we made a detailed comparative study of hPSC-ECs from three different culture systems (e.g., 2D, 3D PNIPAAm-PEG hydrogel, and 3D alginate hydrogel cultures) based on our previous reports. We expanded hPSCs and differentiated them into ECs in three culture systems. Both 3D hydrogel systems could mimic an in vivo physiologically relevant microenvironment to protect cells from shear force and prevent cell agglomeration, leading to a high culture efficiency and a high volumetric yield. We demonstrated that hPSC-ECs produced from both hydrogel systems had similar results as 2D-ECs. The transcriptome analysis showed that PEG-ECs and alginate-ECs displayed a functional phenotype due to their higher gene expressions in vasculature development, extracellular matrix, angiogenesis, and glycolysis, while 2D-ECs showed a proliferative phenotype due to their higher gene expressions in cell proliferation. Taken together, both PEG- and alginate-hydrogel systems will significantly advance the applications of hPSC-ECs in various biomedical fields.
Collapse
Affiliation(s)
- Zhanqi Wang
- Department
of Vascular Surgery, Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Fuxing Zuo
- Department
of Neurosurgery, National Cancer Center/National Clinical Research
Center for Cancer/Cancer Hospital, Chinese
Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qing Liu
- Department
of Obstetrics, Beijing Obstetrics and Gynecology
Hospital Capital Medical University, Beijing 100006, China
| | - Xuesheng Wu
- Department
of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qian Du
- Department
of Biological Systems Engineering, University
of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| | - Yuguo Lei
- Department
of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| | - Zhangmin Wu
- Department
of Vascular Surgery, Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Haishuang Lin
- Department
of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
25
|
Abstract
Induced pluripotent stem cells (iPSC) open up the unique perspective of manufacturing cell products for drug development and regenerative medicine in tissue-, disease- and patient-specific forms. iPSC can be multiplied almost without restriction and differentiated into cell types of all organs. The basis for clinical use of iPSC is a high number of cells (approximately 7 × 107 cells per treatment), which must be produced cost-effectively while maintaining reproducible and high quality. Compared to manual cell production, the automation of cell production offers a unique chance of reliable reproducibility of cells in addition to cost reduction and increased throughput. StemCellFactory is a prototype for a fully automated production of iPSC. However, in addition to the already tested functionality of the system, it must be shown that this automation brings necessary economic advantages. This paper presents that fully automated stem cell production offers economic advantages in addition to increased throughput and better quality. First, biological and technological basics for a fully automated production of iPSC are presented. Second, the basics for profitability calculation are presented. Third, profitability of both manual and automated production are calculated. Finally, different scenarios effecting the profitability of manual and automated production are compared.
Collapse
|
26
|
Muckom RJ, Sampayo RG, Johnson HJ, Schaffer DV. Advanced Materials to Enhance Central Nervous System Tissue Modeling and Cell Therapy. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2002931. [PMID: 33510596 PMCID: PMC7840150 DOI: 10.1002/adfm.202002931] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Indexed: 05/04/2023]
Abstract
The progressively deeper understanding of mechanisms underlying stem cell fate decisions has enabled parallel advances in basic biology-such as the generation of organoid models that can further one's basic understanding of human development and disease-and in clinical translation-including stem cell based therapies to treat human disease. Both of these applications rely on tight control of the stem cell microenvironment to properly modulate cell fate, and materials that can be engineered to interface with cells in a controlled and tunable manner have therefore emerged as valuable tools for guiding stem cell growth and differentiation. With a focus on the central nervous system (CNS), a broad range of material solutions that have been engineered to overcome various hurdles in constructing advanced organoid models and developing effective stem cell therapeutics is reviewed. Finally, regulatory aspects of combined material-cell approaches for CNS therapies are considered.
Collapse
Affiliation(s)
- Riya J Muckom
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94704, USA
| | - Rocío G Sampayo
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94704, USA
| | - Hunter J Johnson
- Department of Bioengineering, UC Berkeley, Berkeley, CA 94704, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94704, USA
| |
Collapse
|
27
|
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]
|
28
|
Wyrobnik TA, Ducci A, Micheletti M. Advances in human mesenchymal stromal cell-based therapies - Towards an integrated biological and engineering approach. Stem Cell Res 2020; 47:101888. [PMID: 32688331 DOI: 10.1016/j.scr.2020.101888] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 12/14/2022] Open
Abstract
Recent advances of stem cell-based therapies in clinical trials have raised the need for large-scale manufacturing platforms that can supply clinically relevant doses to meet an increasing demand. Promising results have been reported using stirred-tank bioreactors, where human Mesenchymal Stromal Cells (hMSCs) were cultured in suspension on microcarriers (MCs), although the formation of microcarrier-cell-aggregates might still limit mass transfer and determine a heterogeneous distribution of hMSCs. A variety of MCs, bioreactor-impeller configurations, and agitation conditions have been established in an attempt to overcome the trade-off of ensuring good suspension while keeping the stresses to a minimum. While understanding and controlling the fluid flow environment of bioreactors has been initially under-appreciated, it has recently gained in popularity in the mission of providing ideal culture environments across different scales. This review article aims to provide a comprehensive overview of how rigorous engineering characterisation studies improved the outcome of biological process development and scale-up efforts. Reconciling these two disciplines is crucial to propose tailored bioprocessing solutions that can provide improved growth environments across a range of scales for the allogeneic cell therapies of the future.
Collapse
Affiliation(s)
- Tom A Wyrobnik
- Department of Biochemical Engineering, UCL, Gower Street, London WC1E 6BT, UK
| | - Andrea Ducci
- Department of Mechanical Engineering, UCL, Torrington Place, London WC1E 7JE, UK
| | - Martina Micheletti
- Department of Biochemical Engineering, UCL, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
29
|
Garzon Dasgupta AK, Martyanov AA, Filkova AA, Panteleev MA, Sveshnikova AN. Development of a Simple Kinetic Mathematical Model of Aggregation of Particles or Clustering of Receptors. Life (Basel) 2020; 10:E97. [PMID: 32604803 PMCID: PMC7345685 DOI: 10.3390/life10060097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 06/24/2020] [Indexed: 12/24/2022] Open
Abstract
The process of clustering of plasma membrane receptors in response to their agonist is the first step in signal transduction. The rate of the clustering process and the size of the clusters determine further cell responses. Here we aim to demonstrate that a simple 2-differential equation mathematical model is capable of quantitative description of the kinetics of 2D or 3D cluster formation in various processes. Three mathematical models based on mass action kinetics were considered and compared with each other by their ability to describe experimental data on GPVI or CR3 receptor clustering (2D) and albumin or platelet aggregation (3D) in response to activation. The models were able to successfully describe experimental data without losing accuracy after switching between complex and simple models. However, additional restrictions on parameter values are required to match a single set of parameters for the given experimental data. The extended clustering model captured several properties of the kinetics of cluster formation, such as the existence of only three typical steady states for this system: unclustered receptors, receptor dimers, and clusters. Therefore, a simple kinetic mass-action-law-based model could be utilized to adequately describe clustering in response to activation both in 2D and in 3D.
Collapse
Affiliation(s)
- Andrei K. Garzon Dasgupta
- Faculty of Physics, Lomonosov Moscow State University, 1/2 Leninskie gory, 119991 Moscow, Russia; (A.K.G.D.); (A.A.M.); (A.A.F.); (M.A.P.)
- National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, 117198 Moscow, Russia
| | - Alexey A. Martyanov
- Faculty of Physics, Lomonosov Moscow State University, 1/2 Leninskie gory, 119991 Moscow, Russia; (A.K.G.D.); (A.A.M.); (A.A.F.); (M.A.P.)
- National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, 117198 Moscow, Russia
- Institute for Biochemical Physics (IBCP), Russian Academy of Sciences (RAS), Russian Federation, Kosyigina 4, 119334 Moscow, Russia
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., 109029 Moscow, Russia
| | - Aleksandra A. Filkova
- Faculty of Physics, Lomonosov Moscow State University, 1/2 Leninskie gory, 119991 Moscow, Russia; (A.K.G.D.); (A.A.M.); (A.A.F.); (M.A.P.)
- National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, 117198 Moscow, Russia
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., 109029 Moscow, Russia
| | - Mikhail A. Panteleev
- Faculty of Physics, Lomonosov Moscow State University, 1/2 Leninskie gory, 119991 Moscow, Russia; (A.K.G.D.); (A.A.M.); (A.A.F.); (M.A.P.)
- National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, 117198 Moscow, Russia
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., 109029 Moscow, Russia
- Faculty of Biological and Medical Physics, Moscow Institute of Physics and Technology, 9 Institutskii per., 141700 Dolgoprudnyi, Russia
| | - Anastasia N. Sveshnikova
- Faculty of Physics, Lomonosov Moscow State University, 1/2 Leninskie gory, 119991 Moscow, Russia; (A.K.G.D.); (A.A.M.); (A.A.F.); (M.A.P.)
- National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, 117198 Moscow, Russia
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., 109029 Moscow, Russia
- Department of Normal Physiology, Sechenov First Moscow State Medical University, 8/2 Trubetskaya St., 119991 Moscow, Russia
| |
Collapse
|
30
|
Raman S, Srinivasan G, Brookhouser N, Nguyen T, Henson T, Morgan D, Cutts J, Brafman DA. A Defined and Scalable Peptide-Based Platform for the Generation of Human Pluripotent Stem Cell-Derived Astrocytes. ACS Biomater Sci Eng 2020; 6:3477-3490. [PMID: 32550261 PMCID: PMC7284803 DOI: 10.1021/acsbiomaterials.0c00067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/06/2020] [Indexed: 01/07/2023]
Abstract
![]()
Astrocytes
comprise the most abundant cell type in the central
nervous system (CNS) and play critical roles in maintaining neural
tissue homeostasis. In addition, astrocyte dysfunction and death has
been implicated in numerous neurological disorders such as multiple
sclerosis, Alzheimer’s disease, amyotrophic lateral sclerosis
(ALS), and Parkinson’s disease (PD). As such, there is much
interest in using human pluripotent stem cell (hPSC)-derived astrocytes
for drug screening, disease modeling, and regenerative medicine applications.
However, current protocols for generation of astrocytes from hPSCs
are limited by the use of undefined xenogeneic components and two-dimensional
(2D) culture surfaces, which limits their downstream applications
where large-quantities of cells generated under defined conditions
are required. Here, we report the use of a completely synthetic, peptide-based
substrate that allows for the differentiation of highly pure populations
of astrocytes from several independent hPSC lines, including those
derived from patients with neurodegenerative disease. This substrate,
which we demonstrate is compatible with both conventional 2D culture
formats and scalable microcarrier (MC)-based technologies, leads to
the generation of cells that express high levels of canonical astrocytic
markers as well as display properties characteristic of functionally
mature cells including production of apolipoprotein E (ApoE), responsiveness
to inflammatory stimuli, ability to take up amyloid-β (Aβ),
and appearance of robust calcium transients. Finally, we show that
these astrocytes can be cryopreserved without any loss of functionality.
In the future, we anticipate that these methods will enable the development
of bioprocesses for the production of hPSC-derived astrocytes needed
for biomedical research and clinical applications.
Collapse
Affiliation(s)
- Sreedevi Raman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Gayathri Srinivasan
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Nicholas Brookhouser
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States.,Graduate Program in Clinical Translational Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona 85004, United States
| | - Toan Nguyen
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Tanner Henson
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Daylin Morgan
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Joshua Cutts
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - David A Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| |
Collapse
|
31
|
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
|
32
|
Polanco A, Kuang B, Yoon S. Bioprocess Technologies that Preserve the Quality of iPSCs. Trends Biotechnol 2020; 38:1128-1140. [PMID: 32941792 DOI: 10.1016/j.tibtech.2020.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/16/2022]
Abstract
Large-scale production of induced pluripotent stem cells (iPSCs) is essential for the treatment of a variety of clinical indications. However, culturing enough iPSCs for clinical applications is problematic due to their sensitive pluripotent state and dependence on a supporting matrix. Developing stem cell bioprocessing strategies that are scalable and meet clinical needs requires incorporating methods that measure and monitor intrinsic markers of cell differentiation state, developmental status, and viability in real time. In addition, proper cell culture modalities that nurture the growth of high-quality stem cells in suspension are critical for industrial scale-up. In this review, we present an overview of cell culture media, suspension modalities, and monitoring techniques that preserve the quality and pluripotency of iPSCs during initiation, expansion, and manufacturing.
Collapse
Affiliation(s)
- Ashli Polanco
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, USA
| | - Bingyu Kuang
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, USA
| | - Seongkyu Yoon
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, MA, USA.
| |
Collapse
|
33
|
Lee B, Borys BS, Kallos MS, Rodrigues CAV, Silva TP, Cabral JMS. Challenges and Solutions for Commercial Scale Manufacturing of Allogeneic Pluripotent Stem Cell Products. Bioengineering (Basel) 2020; 7:E31. [PMID: 32231012 PMCID: PMC7355837 DOI: 10.3390/bioengineering7020031] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 01/09/2023] Open
Abstract
Allogeneic cell therapy products, such as therapeutic cells derived from pluripotent stem cells (PSCs), have amazing potential to treat a wide variety of diseases and vast numbers of patients globally. However, there are various challenges related to the manufacturing of PSCs in large enough quantities to meet commercial needs. This manuscript addresses the challenges for the process development of PSCs production in a bioreactor, and also presents a scalable bioreactor technology that can be a possible solution to remove the bottleneck for the large-scale manufacturing of high-quality therapeutic cells derived from PSCs.
Collapse
Affiliation(s)
- Brian Lee
- PBS Biotech, Inc., Camarillo, CA 93012, USA
| | - Breanna S. Borys
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB T2N 1N4, Canada; (B.S.B.); (M.S.K.)
| | - Michael S. Kallos
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Dr. NW, Calgary, AB T2N 1N4, Canada; (B.S.B.); (M.S.K.)
| | - Carlos A. V. Rodrigues
- Ibb—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal; (C.A.V.R.); (T.P.S.); (J.M.S.C.)
| | - Teresa P. Silva
- Ibb—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal; (C.A.V.R.); (T.P.S.); (J.M.S.C.)
| | - Joaquim M. S. Cabral
- Ibb—Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal; (C.A.V.R.); (T.P.S.); (J.M.S.C.)
| |
Collapse
|
34
|
Barzegari A, Gueguen V, Omidi Y, Ostadrahimi A, Nouri M, Pavon‐Djavid G. The role of Hippo signaling pathway and mechanotransduction in tuning embryoid body formation and differentiation. J Cell Physiol 2020; 235:5072-5083. [DOI: 10.1002/jcp.29455] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 01/06/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Abolfazl Barzegari
- Department of Medical Biotechnology, Faculty of Advanced Medical SciencesTabriz University of Medical Sciences Tabriz Iran
| | - Virginie Gueguen
- INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular BioengineeringUniversité Paris 13 Paris France
| | - Yadollah Omidi
- Research Center for Pharmaceutical NanotechnologyTabriz University of Medical Sciences Tabriz Iran
- Department of Pharmaceutics, Faculty of PharmacyTabriz University of Medical Sciences Tabriz Iran
| | - Alireza Ostadrahimi
- Nutrition Research CenterTabriz University of Medical Sciences Tabriz Iran
- Department of Clinical Nutrition, Faculty of Nutrition and Food SciencesTabriz University of Medical Sciences Tabriz Iran
| | - Mohammad Nouri
- Department of Medical Biotechnology, Faculty of Advanced Medical SciencesTabriz University of Medical Sciences Tabriz Iran
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of MedicineTabriz University of Medical Sciences Tabriz Iran
| | - Graciela Pavon‐Djavid
- INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular BioengineeringUniversité Paris 13 Paris France
| |
Collapse
|
35
|
Hashida A, Uemura T, Kino-Oka M. Kinetics on aggregate behaviors of human induced pluripotent stem cells in static suspension and rotating flow cultures. J Biosci Bioeng 2019; 129:494-501. [PMID: 31826834 DOI: 10.1016/j.jbiosc.2019.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/06/2019] [Accepted: 11/11/2019] [Indexed: 12/31/2022]
Abstract
Understanding of kinetics on aggregate behaviors of human induced pluripotent stem cells (hiPSCs) is critical knowledge for culture design because aggregate behaviors are considered to affect cell growth. In this study, we elucidated kinetics on aggregate behaviors of two types of hiPSCs (253G1 and 201B7 lines) to clarify the influence of aggregate behaviors on cell growth by comparing aggregate morphology, size of cell aggregates, and kinetic parameters in 72 h culture under static and floating conditions, which were realized by multi-dimple plate and rotating wall vessel, respectively. In the case of 253G1 line under floating condition, aggregate number decreased and size increased drastically during culture time, t = 0-24 h due to coalescence between cell aggregates. The apparent specific growth rate decreased after t = 24 h although cell number and aggregate size gradually increased under static condition. In the case of 201B7 line under floating condition, cell and aggregate number, and aggregate size kept constant levels during t = 24-72 h due to collapse of cell aggregates by stripping of single cells from aggregate, suggesting that specific death rate increased after t = 24 h despite constant levels of apparent specific growth rate and aggregate number under static condition. Our kinetic analysis concluded that excessive increase of aggregate size due to coalescence and cell death due to collapse critically affected growth of hiPSCs in suspension culture.
Collapse
Affiliation(s)
- Akihiro Hashida
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshimasa Uemura
- Department of Precise and Science Technology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
36
|
Xu Y, Chen C, Hellwarth PB, Bao X. Biomaterials for stem cell engineering and biomanufacturing. Bioact Mater 2019; 4:366-379. [PMID: 31872161 PMCID: PMC6909203 DOI: 10.1016/j.bioactmat.2019.11.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/09/2019] [Accepted: 11/20/2019] [Indexed: 12/15/2022] Open
Abstract
Recent years have witnessed the expansion of tissue failures and diseases. The uprising of regenerative medicine converges the sight onto stem cell-biomaterial based therapy. Tissue engineering and regenerative medicine proposes the strategy of constructing spatially, mechanically, chemically and biologically designed biomaterials for stem cells to grow and differentiate. Therefore, this paper summarized the basic properties of embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and adult stem cells. The properties of frequently used biomaterials were also described in terms of natural and synthetic origins. Particularly, the combination of stem cells and biomaterials for tissue repair applications was reviewed in terms of nervous, cardiovascular, pancreatic, hematopoietic and musculoskeletal system. Finally, stem-cell-related biomanufacturing was envisioned and the novel biofabrication technologies were discussed, enlightening a promising route for the future advancement of large-scale stem cell-biomaterial based therapeutic manufacturing.
Collapse
Affiliation(s)
| | | | | | - Xiaoping Bao
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, West Lafayette, IN, 47907, USA
| |
Collapse
|
37
|
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
|
38
|
Tachibana K. N-cadherin-mediated aggregate formation; cell detachment by Trypsin-EDTA loses N-cadherin and delays aggregate formation. Biochem Biophys Res Commun 2019; 516:414-418. [PMID: 31227216 DOI: 10.1016/j.bbrc.2019.06.067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 06/13/2019] [Indexed: 12/31/2022]
Abstract
Although cell aggregates/spheroids are useful tools in various fields of cell biology, the mechanism for aggregate formation is not fully resolved yet. Here I show the involvement of N-cadherin in the quick formation of packed aggregates in suspension culture. HEK293T cells detached from substratum by Trypsin alone quickly formed packed aggregates in suspension. This aggregate formation was inhibited by the down-regulation of N-cadherin. Meanwhile, aggregate formation of cells detached by Trypsin-EDTA was significantly delayed. N-cadherin was transiently lost by Trypsin-EDTA-treatment, and the re-expression of N-cadherin corresponded to delayed aggregate formation. Furthermore, packed phenotype was not observed in the absence of N-cadherin. These findings indicate that N-cadherin mediates quick formation of packed aggregates/spheroids in suspension culture.
Collapse
Affiliation(s)
- Kouichi Tachibana
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8565, Japan.
| |
Collapse
|
39
|
Greuel S, Hanci G, Böhme M, Miki T, Schubert F, Sittinger M, Mandenius CF, Zeilinger K, Freyer N. Effect of inoculum density on human-induced pluripotent stem cell expansion in 3D bioreactors. Cell Prolif 2019; 52:e12604. [PMID: 31069891 PMCID: PMC6668975 DOI: 10.1111/cpr.12604] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/15/2019] [Accepted: 02/20/2019] [Indexed: 12/18/2022] Open
Abstract
Objective For optimized expansion of human‐induced pluripotent stem cells (hiPSCs) with regards to clinical applications, we investigated the influence of the inoculum density on the expansion procedure in 3D hollow‐fibre bioreactors. Materials and Methods Analytical‐scale bioreactors with a cell compartment volume of 3 mL or a large‐scale bioreactor with a cell compartment volume of 17 mL were used and inoculated with either 10 × 106 or 50 × 106 hiPSCs. Cells were cultured in bioreactors over 15 days; daily measurements of biochemical parameters were performed. At the end of the experiment, the CellTiter‐Blue® Assay was used for culture activity evaluation and cell quantification. Also, cell compartment sections were removed for gene expression and immunohistochemistry analysis. Results The results revealed significantly higher values for cell metabolism, cell activity and cell yields when using the higher inoculation number, but also a more distinct differentiation. As large inoculation numbers require cost and time‐extensive pre‐expansion, low inoculation numbers may be used preferably for long‐term expansion of hiPSCs. Expansion of hiPSCs in the large‐scale bioreactor led to a successful production of 5.4 × 109 hiPSCs, thereby achieving sufficient cell amounts for clinical applications. Conclusions In conclusion, the results show a significant effect of the inoculum density on cell expansion, differentiation and production of hiPSCs, emphasizing the importance of the inoculum density for downstream applications of hiPSCs. Furthermore, the bioreactor technology was successfully applied for controlled and scalable production of hiPSCs for clinical use.
Collapse
Affiliation(s)
- Selina Greuel
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Güngör Hanci
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Mike Böhme
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Toshio Miki
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | | | - Michael Sittinger
- Tissue Engineering Laboratory, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Carl-Fredrik Mandenius
- Division of Biotechnology, Department of Physics, Chemistry and Biology (IFM), Linköping University, Linköping, Sweden
| | - Katrin Zeilinger
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nora Freyer
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
40
|
Kato Y, Matsumoto T, Kino-Oka M. Effect of liquid flow by pipetting during medium change on deformation of hiPSC aggregates. Regen Ther 2019; 12:20-26. [PMID: 31890763 PMCID: PMC6933458 DOI: 10.1016/j.reth.2019.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/19/2019] [Accepted: 03/21/2019] [Indexed: 12/17/2022] Open
Abstract
Introduction Maintaining the pluripotency and homogeneity of human induced pluripotent stem cells (hiPSCs) requires stable culture conditions with consistent medium change. In this study, we evaluated the performance of medium change by machine vs. medium change performed manually in terms of their impact on the aggregate shape of hiPSCs. Methods Aggregates of two hiPSC lines (1383D2 and Tic) were cultured, and the medium change was conducted either manually or with a machine. The populational homogeneity in aggregate shape was determined based on the projected aggregate area for size expansion as well as the circularity for spherical morphology. Results In the case of manually performed medium changes, the size of 1383D2 aggregates expanded homogeneously, maintaining its spherical morphology as culture duration increased, while spherical morphology was deformed in Tic aggregates, which had a heterogeneous population in terms of shape. In the case of medium change performed by a machine under a low flux of liquid flow, cultures of both aggregates showed homogeneous populations without deformation, although a high flux led to a heterogeneous population. The heterogeneous population observed in manually performed medium change was caused by the low stability of motion. In addition, time-lapse observation revealed that the Tic aggregates underwent tardive deformation with cellular protrusions from the aggregate surface after medium change with high flux. Histological analysis revealed a spatial heterogeneity of collagen type I inside 1383D2 aggregates, which had a shell structure with strong formation of collagen type I at the periphery of the aggregates, while Tic aggregates did not have a shell structure, suggesting that the shell structure prevented aggregate deformation. Conclusion Medium change by a machine led to a homogeneous population of aggregate shapes. Liquid flow caused tardive deformation of aggregates, but the shell structure of collagen type I in aggregates maintained its spherical shape. Mechanization of medium change leads to homogeneous shape of iPSC aggregates. Tardive change of aggregates was observed. Collagen type I distribution in aggregates induces shell structure formation.
Collapse
Affiliation(s)
- Yuma Kato
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Takuya Matsumoto
- Institute for Innovation, Ajinomoto Co., Inc., 1-1 Suzukicho, Kawasaki-ku, Kawasaki, Kanagawa, 210-8681, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamada-oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
41
|
Lin H, Du Q, Li Q, Wang O, Wang Z, Liu K, Akert L, Zhang C, Chung S, Duan B, Lei Y. Differentiating human pluripotent stem cells into vascular smooth muscle cells in three dimensional thermoreversible hydrogels. Biomater Sci 2019; 7:347-361. [PMID: 30483691 DOI: 10.1039/c8bm01128a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Vascular smooth muscle cells (VSMCs) are of great value and are needed in large quantities for tissue engineering, drug screening, disease modeling and cell-based therapies. However, getting high quantity VSMCs remains a challenge. Here, we report a method for the scalable manufacturing of VSMCs from human pluripotent stem cells (hPSCs). hPSCs are expanded and differentiated into VSMCs in a three dimensional (3D) thermoreversible hydrogel. The hydrogel not only acts as a 3D scaffold for cells to grow, but also protects cells from hydrodynamic stresses in the culture vessel and prevents cells from excessive aggregation. Together, the hydrogel creates a cell-friendly microenvironment, leading to high culture efficiency. We show that VSMCs can be generated in 10 days with high viability (>90%), high purity (>80%) and high yield (∼2.0 × 107 cells per mL hydrogel) in the hydrogel scaffold. The generated VSMCs have normal functions. Genome-wide gene expression analysis shows VSMCs made in the hydrogel (i.e. 3D-VSMCs) have higher expression of genes related to vasculature development and glycolysis compared to VSMCs made in the conventional 2D cultures (i.e. 2D-VSMCs), while 2D-VSMCs have higher expression of genes related to cell proliferation. This simple, defined and efficient method is scalable for manufacturing hPSC-VSMCs for various biomedical applications.
Collapse
Affiliation(s)
- Haishuang Lin
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Nebraska, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lin H, Qiu X, Du Q, Li Q, Wang O, Akert L, Wang Z, Anderson D, Liu K, Gu L, Zhang C, Lei Y. Engineered Microenvironment for Manufacturing Human Pluripotent Stem Cell-Derived Vascular Smooth Muscle Cells. Stem Cell Reports 2019; 12:84-97. [PMID: 30527760 PMCID: PMC6335449 DOI: 10.1016/j.stemcr.2018.11.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 12/18/2022] Open
Abstract
Human pluripotent stem cell-derived vascular smooth muscle cells (hPSC-VSMCs) are of great value for disease modeling, drug screening, cell therapies, and tissue engineering. However, producing a high quantity of hPSC-VSMCs with current cell culture technologies remains very challenging. Here, we report a scalable method for manufacturing hPSC-VSMCs in alginate hydrogel microtubes (i.e., AlgTubes), which protect cells from hydrodynamic stresses and limit cell mass to <400 μm to ensure efficient mass transport. The tubes provide cells a friendly microenvironment, leading to extremely high culture efficiency. We have shown that hPSC-VSMCs can be generated in 10 days with high viability, high purity, and high yield (∼5.0 × 108 cells/mL). Phenotype and gene expression showed that VSMCs made in AlgTubes and VSMCs made in 2D cultures were similar overall. However, AlgTube-VSMCs had higher expression of genes related to vasculature development and angiogenesis, and 2D-VSMCs had higher expression of genes related to cell death and biosynthetic processes.
Collapse
Affiliation(s)
- Haishuang Lin
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Xuefeng Qiu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qian Du
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Qiang Li
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; Biomedical Engineering Program, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Ou Wang
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; Biomedical Engineering Program, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Leonard Akert
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Zhanqi Wang
- Department of Vascular Surgery, Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing 100029, China
| | - Dirk Anderson
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Kan Liu
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Linxia Gu
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Chi Zhang
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Yuguo Lei
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; Biomedical Engineering Program, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
43
|
Human Pluripotent Stem Cells: Applications and Challenges for Regenerative Medicine and Disease Modeling. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2019; 171:189-224. [PMID: 31740987 DOI: 10.1007/10_2019_117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years, human pluripotent stem (hPS) cells have started to emerge as a potential tool with application in fields such as regenerative medicine, disease modeling, and drug screening. In particular, the ability to differentiate human-induced pluripotent stem (hiPS) cells into different cell types and to mimic structures and functions of a specific target organ, resourcing to organoid technology, has introduced novel model systems for disease recapitulation while offering a powerful tool to provide a faster and reproducible approach in the process of drug discovery. All these technologies are expected to improve the overall quality of life of the humankind. Here, we highlight the main applications of hiPS cells and the main challenges associated with the translation of hPS cell derivatives into clinical settings and other biomedical applications, such as the costs of the process and the ability to mimic the complexity of the in vivo systems. Moreover, we focus on the bioprocessing approaches that can be applied towards the production of high numbers of cells as well as their efficient differentiation into the final product and further purification.
Collapse
|
44
|
Lin H, Du Q, Li Q, Wang O, Wang Z, Elowsky C, Liu K, Zhang C, Chung S, Duan B, Lei Y. Manufacturing human pluripotent stem cell derived endothelial cells in scalable and cell-friendly microenvironments. Biomater Sci 2019; 7:373-388. [DOI: 10.1039/c8bm01095a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alginate hydrogel tubes are designed for the scalable expansion of human pluripotent stem cells and efficient differentiation into endothelial cells.
Collapse
Affiliation(s)
- Haishuang Lin
- Department of Chemical and Biomolecular Engineering
- University of Nebraska-Lincoln
- USA
| | - Qian Du
- Department of Biological Systems Engineering
- University of Nebraska-Lincoln
- USA
| | - Qiang Li
- Department of Chemical and Biomolecular Engineering
- University of Nebraska-Lincoln
- USA
- Biomedical Engineering Program
- University of Nebraska-Lincoln
| | - Ou Wang
- Department of Chemical and Biomolecular Engineering
- University of Nebraska-Lincoln
- USA
- Biomedical Engineering Program
- University of Nebraska-Lincoln
| | - Zhanqi Wang
- Department of Vascular Surgery
- Beijing Anzhen Hospital of Capital Medical University
- Beijing Institute of Heart Lung and Blood Vessel Diseases
- Beijing
- China
| | - Christian Elowsky
- Department of Agronomy and Horticulture
- University of Nebraska-Lincoln
- USA
| | - Kan Liu
- Department of Biological Systems Engineering
- University of Nebraska-Lincoln
- USA
| | - Chi Zhang
- Department of Biological Systems Engineering
- University of Nebraska-Lincoln
- USA
| | - Soonkyu Chung
- Department of Nutrition and Health Sciences
- University of Nebraska-Lincoln
- Lincoln
- USA
| | - Bin Duan
- Mary and Dick Holland Regenerative Medicine Program
- University of Nebraska Medical Center
- Omaha
- USA
| | - Yuguo Lei
- Department of Chemical and Biomolecular Engineering
- University of Nebraska-Lincoln
- USA
- Biomedical Engineering Program
- University of Nebraska-Lincoln
| |
Collapse
|
45
|
Cummins KA, Crampton AL, Wood DK. A High-Throughput Workflow to Study Remodeling of Extracellular Matrix-Based Microtissues. Tissue Eng Part C Methods 2018; 25:25-36. [PMID: 30430922 DOI: 10.1089/ten.tec.2018.0290] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
IMPACT STATEMENT The described microtissue-microwell workflow is uniquely suited for high-throughput study of extracellular matrix (ECM) remodeling at the molecular, cellular, and tissue levels and demonstrates possibilities of studying progressive, heterogeneous diseases in a way that is meaningful for drug discovery and development. We outline several assays that can be utilized in studying tissue-level diseases and functions that involve cell-ECM interactions and ECM remodeling (e.g., cancer, fibrosis, wound healing) in pursuit of an improved three-dimensional cell culturing system. Finally, we demonstrate the ability to cryopreserve cells encapsulated in microtissue constructs while remaining highly viable, proliferative, and retaining cell functions that are involved in ECM remodeling.
Collapse
Affiliation(s)
- Katherine A Cummins
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Alexandra L Crampton
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - David K Wood
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| |
Collapse
|
46
|
Alloush MM, Liermann M, Zedan A, Oweis GF. A Novel Pulse Damper for Endothelial Cell Flow Bioreactors. Cardiovasc Eng Technol 2018; 10:95-111. [PMID: 30488177 DOI: 10.1007/s13239-018-00394-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 11/13/2018] [Indexed: 11/28/2022]
Abstract
PURPOSE Peristaltic pumps (PP) are favored in flow bioreactors for their non-contact sterile design. But they produce pulsatile flow, which is consequential for the cultured cells. A novel pulse damper (PD) is reported for pulsatility elimination. METHODS The PD design was implemented to target static pressure pulsatility and flow rate (velocity) pulsatility from a PP. Damping effectiveness was tested in a macro-scale, closed-loop recirculating bioreactor mimicking the aortic arch at flow rates up to (4 L/min). Time-resolved particle image velocimetry was used to characterize the velocity field. Endothelial cells (EC) were grown in the bioreactor, and subjected to continuous flow for 15 min with or without PD. RESULTS The PD was found to be nearly 90% effective at reducing pulsatility. The EC exposed to low PP flow without PD exhibited distress signaling in the form of increased ERK1/2 phosphorylation (2.5 folds) when compared to those exposed to the same flow with PD. At high pump flow without PD, the cells detached and did not survive, while they were perfectly healthy with PD. CONCLUSIONS Flow pulsatility from PP causes EC distress at low flow and cell detachment at high flow. Elevated temporal shear stress gradient combined with elevated shear stress magnitude at high flow are believed to be the cause of cell detachment and death. The proposed PD design was effective at minimizing the hemodynamic stressors in the pump's output, demonstrably reducing cell distress. Adoption of the proposed PD design in flow bioreactors should improve experimental protocols.
Collapse
Affiliation(s)
- M M Alloush
- Department of Mechanical Engineering, MS Faculty of Engineering and Architecture, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon
| | - M Liermann
- Department of Mechanical Engineering, MS Faculty of Engineering and Architecture, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon.,Danfoss Power Solutions, Krokamp 35, 24539, Neumünster, Germany
| | - A Zedan
- Department of Physiology, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon
| | - G F Oweis
- Department of Mechanical Engineering, MS Faculty of Engineering and Architecture, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon.
| |
Collapse
|
47
|
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
|
48
|
Lin H, Du Q, Li Q, Wang O, Wang Z, Liu K, Elowsky C, Zhang C, Lei Y. Hydrogel-Based Bioprocess for Scalable Manufacturing of Human Pluripotent Stem Cell-Derived Neural Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2018; 10:29238-29250. [PMID: 30091584 DOI: 10.1021/acsami.8b05780] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Neural stem cells derived from human pluripotent stem cells (hPSC-NSCs) are of great value for modeling diseases, developing drugs, and treating neurological disorders. However, manufacturing high-quantity and -quality hPSC-NSCs, especially for clinical applications, remains a challenge. Here, we report a chemically defined, high-yield, and scalable bioprocess for manufacturing hPSC-NSCs. hPSCs are expanded and differentiated into NSCs in microscale tubes made with alginate hydrogels. The tubes are used to isolate cells from the hydrodynamic stresses in the culture vessel and limit the radial diameter of the cell mass to less than 400 μm to ensure efficient mass transport during the culture. The hydrogel tubes provide uniform, reproducible, and cell-friendly microspaces and microenvironments for cells. With this new technology, we showed that hPSC-NSCs could be produced in 12 days with high viability (∼95%), high purity (>90%), and high yield (∼5 × 108 cells/mL of microspace). The volumetric yield is about 250 times more than the current state-of-the-art. Whole transcriptome analysis and quantitative real-time polymerase chain reaction showed that hPSC-NSCs made by this process had a similar gene expression to hPSC-NSCs made by the conventional culture technology. The produced hPSC-NSCs could mature into both neurons and glial cells in vitro and in vivo. The process developed in this paper can be used to produce large numbers of hPSC-NSCs for various biomedical applications in the future.
Collapse
Affiliation(s)
| | | | | | | | - Zhanqi Wang
- Department of Vascular Surgery, Beijing Anzhen Hospital of Capital Medical University , Beijing Institute of Heart Lung and Blood Vessel Diseases , Beijing 100029 , China
| | | | | | | | | |
Collapse
|
49
|
A Scalable and Efficient Bioprocess for Manufacturing Human Pluripotent Stem Cell-Derived Endothelial Cells. Stem Cell Reports 2018; 11:454-469. [PMID: 30078557 PMCID: PMC6092882 DOI: 10.1016/j.stemcr.2018.07.001] [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] [Received: 02/02/2018] [Revised: 07/05/2018] [Accepted: 07/09/2018] [Indexed: 02/08/2023] Open
Abstract
Endothelial cells (ECs) are of great value for cell therapy, tissue engineering, and drug discovery. Obtaining high-quantity and -quality ECs remains very challenging. Here, we report a method for the scalable manufacturing of ECs from human pluripotent stem cells (hPSCs). hPSCs are expanded and differentiated into ECs in a 3D thermoreversible PNIPAAm-PEG hydrogel. The hydrogel protects cells from hydrodynamic stresses in the culture vessel and prevents cells from excessive agglomeration, leading to high-culture efficiency including high-viability (>90%), high-purity (>80%), and high-volumetric yield (2.0 × 107 cells/mL). These ECs (i.e., 3D-ECs) had similar properties as ECs made using 2D culture systems (i.e., 2D-ECs). Genome-wide gene expression analysis showed that 3D-ECs had higher expression of genes related to vasculature development, extracellular matrix, and glycolysis, while 2D-ECs had higher expression of genes related to cell proliferation. hPSCs can be differentiated into endothelial cells in 3D thermoreversible hydrogels The differentiation efficiency is similar to this in 2D cultures The global gene expression and phenotypes are similar to ECs made in 2D cultures
Collapse
|
50
|
Ting S, Lam A, Tong G, Chen A, Wei H, Wu J, Lam YN, Reuveny S, Oh S. Meticulous optimization of cardiomyocyte yields in a 3-stage continuous integrated agitation bioprocess. Stem Cell Res 2018; 31:161-173. [DOI: 10.1016/j.scr.2018.07.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/12/2018] [Accepted: 07/23/2018] [Indexed: 01/15/2023] Open
|