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
|
Francis N, Aho J, Ben-Nun IF, Bharti K, Dianat N, Makovoz B, Nouri P, Rothberg J, Song H, Zamilpa R, Lakshmipathy U, Allickson J. Scaling up pluripotent stem cell-based therapies - considerations, current challenges and emerging technologies: perspectives from the ISCT Emerging Regenerative Medicine Working Group. Cytotherapy 2025:S1465-3249(25)00678-4. [PMID: 40353785 DOI: 10.1016/j.jcyt.2025.04.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/21/2025] [Accepted: 04/03/2025] [Indexed: 05/14/2025]
Abstract
Approval of induced pluripotent stem cells (iPSCs) for the manufacture of cell therapies to support clinical trials is now becoming realized after more than 20 years of research and development. However, manufacturing these therapies at the scale required for patient treatment, as well as for key clinical trial enabling activities such as preclinical and stability studies, remains a challenge. In 2022 the International Society for Cell and Gene Therapy (ISCT) established a Working Group on Emerging Regenerative Medicine Technologies, an area in which iPSC-derived technologies are expected to play a key role. In this article, the Working Group provides an overview of the considerations and challenges facing stem cell therapy developers, including development-stage specific manufacturing processes, the decision on when to implement automation, the choice of technology and different requirements of expansion and differentiation aspects of the process, and the integration of automation for both manufacturing and analytics in an end-to-end manufacturing process. The role of scalable manufacturing technologies in the application of quality-by-design approaches to product development, and the use of design-of-experiment approaches for increased product characterization, is discussed. Finally, we provide an in-depth review of the different technologies that have been used for expansion and differentiation of iPSC-derived therapies to date, including compatibility with good manufacturing practice requirements and process analytical technologies. We hope that this overview will summarize the existing knowledge in the field and reduce the challenges that therapy developers face in translating their research into clinical and commercial scale manufacturing.
Collapse
Affiliation(s)
- Natalie Francis
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, UK.
| | - Joy Aho
- Product Management and Strategy, NMDP BioTherapies, Minneapolis, USA
| | | | - Kapil Bharti
- National Eye Institute, National Institute of Health, Bethesda, Maryland, USA
| | - Noushin Dianat
- R&D Cell Therapy, Sartorius Stedim Biotech, Göttingen, Germany
| | - Bar Makovoz
- Product Management, Cellino, Cambridge, Massachusetts, USA
| | - Parivash Nouri
- Pluripotent Stem Cells Product Management, Miltenyi Biotec, Bergisch, Gladbach, Germany
| | - Janet Rothberg
- Process & Analytical Development, CCRM, MaRS Centre, West Tower, Toronto, Ontario, Canada
| | - Hannah Song
- Center for Cellular Engineering, National Institute of Health, Rockville, Maryland, USA
| | | | - Uma Lakshmipathy
- Patheon Cell & Gene Translation Services, Thermo Fisher Scientific, San Diego, California, USA
| | - Julie Allickson
- Clinic's Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, Minnesota, USA
| |
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
|
Grattoni A, Korbutt G, Tomei AA, García AJ, Pepper AR, Stabler C, Brehm M, Papas K, Citro A, Shirwan H, Millman JR, Melero-Martin J, Graham M, Sefton M, Ma M, Kenyon N, Veiseh O, Desai TA, Nostro MC, Marinac M, Sykes M, Russ HA, Odorico J, Tang Q, Ricordi C, Latres E, Mamrak NE, Giraldo J, Poznansky MC, de Vos P. Harnessing cellular therapeutics for type 1 diabetes mellitus: progress, challenges, and the road ahead. Nat Rev Endocrinol 2025; 21:14-30. [PMID: 39227741 PMCID: PMC11938328 DOI: 10.1038/s41574-024-01029-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/06/2024] [Indexed: 09/05/2024]
Abstract
Type 1 diabetes mellitus (T1DM) is a growing global health concern that affects approximately 8.5 million individuals worldwide. T1DM is characterized by an autoimmune destruction of pancreatic β cells, leading to a disruption in glucose homeostasis. Therapeutic intervention for T1DM requires a complex regimen of glycaemic monitoring and the administration of exogenous insulin to regulate blood glucose levels. Advances in continuous glucose monitoring and algorithm-driven insulin delivery devices have improved the quality of life of patients. Despite this, mimicking islet function and complex physiological feedback remains challenging. Pancreatic islet transplantation represents a potential functional cure for T1DM but is hindered by donor scarcity, variability in harvested cells, aggressive immunosuppressive regimens and suboptimal clinical outcomes. Current research is directed towards generating alternative cell sources, improving transplantation methods, and enhancing cell survival without chronic immunosuppression. This Review maps the progress in cell replacement therapies for T1DM and outlines the remaining challenges and future directions. We explore the state-of-the-art strategies for generating replenishable β cells, cell delivery technologies and local targeted immune modulation. Finally, we highlight relevant animal models and the regulatory aspects for advancing these technologies towards clinical deployment.
Collapse
Affiliation(s)
- Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Surgery, Houston Methodist Hospital, Houston, TX, USA.
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, USA.
| | - Gregory Korbutt
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Alice A Tomei
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrew R Pepper
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Cherie Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
- Diabetes Institute, University of Florida, Gainesville, FL, USA
| | - Michael Brehm
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Klearchos Papas
- Department of Surgery, The University of Arizona, Tucson, AZ, USA
| | - Antonio Citro
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Haval Shirwan
- Department of Pediatrics, Ellis Fischel Cancer Center, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Jeffrey R Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Juan Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Melanie Graham
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, USA
| | - Michael Sefton
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Norma Kenyon
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Tejal A Desai
- University of California, San Francisco, Department of Bioengineering and Therapeutic Sciences, San Francisco, CA, USA
- Brown University, School of Engineering, Providence, RI, USA
| | - M Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | | | - Megan Sykes
- Department of Medicine, Columbia Center for Translational Immunology, Columbia University, New York, NY, USA
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
- Department of Surgery, Columbia University, New York, NY, USA
| | - Holger A Russ
- Diabetes Institute, University of Florida, Gainesville, FL, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Jon Odorico
- UW Health Transplant Center, Madison, WI, USA
- Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Qizhi Tang
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA
- Department of Surgery, University of California San Francisco, San Francisco, CA, US
- Gladstone Institute of Genomic Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Esther Latres
- Research Department, Breakthrough T1D, New York, NY, USA
| | | | - Jaime Giraldo
- Research Department, Breakthrough T1D, New York, NY, USA.
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, Netherlands.
| |
Collapse
|
5
|
Govara S, Hosseinalipour SM, Soleimani M. Quantitative investigation of the effect of mechanical and geometrical factors of a laboratory-scale bioreactor using a vibrating agitator on mammalian cell culture indices. Bioprocess Biosyst Eng 2025; 48:85-101. [PMID: 39373731 DOI: 10.1007/s00449-024-03095-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/29/2024] [Indexed: 10/08/2024]
Abstract
Mammalian cell cultures in laboratories are performed in static and dynamic methods, and cell growth indices are higher in dynamic mode. In this study, a lab-scale stirred bioreactor using a vibrating disc and a suitable setup has been introduced for dynamic cell culture, which creates proper mixing at low shear stress. 15 experiments have been done by Raji cell in batch mode using Box-Behnken design to quantitatively investigate the effect of mechanical and geometrical factors of this bioreactor on cell culture indices. Three structural factors, including disc diameter, vibration amplitude, and the height of the disc placement have been selected as the main factors. Three cell growth indices including the specific growth rate, the maximum cell concentration, and productivity have been considered as biological responses. Resulting models predict the value of each index under different settings of the factors with good accuracy. Results show that the disc diameter has the greatest effect among the investigated factors. Also, the specific growth rate, the natural logarithm of the maximum cell concentration, and productivity are about 0.033 (1/h), 13.2, and 5133 (cells/hmL), respectively by using a 25 (mm) disc with a vibration amplitude of 2.5 up to 3 (mm), and a placement height of 40 up to 60 (mm).
Collapse
Affiliation(s)
- Sepehr Govara
- School of Mechanical Engineering, Iran University of Science and Technology, Resalat Square, Hengam Street, Tehran, Tehran, Iran
| | - S M Hosseinalipour
- School of Mechanical Engineering, Iran University of Science and Technology, Resalat Square, Hengam Street, Tehran, Tehran, Iran.
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Tehran, Iran
| |
Collapse
|
6
|
Ueki M, Suzuki T, Kato Y. Large-scale cultivation of human iPS cells in bioreactor with reciprocal mixing. J Biosci Bioeng 2024; 137:149-155. [PMID: 38185598 DOI: 10.1016/j.jbiosc.2023.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/28/2023] [Accepted: 12/12/2023] [Indexed: 01/09/2024]
Abstract
A substantial number of human iPS cells (hiPSCs) is needed for cell therapy to be successful against various diseases. We previously reported on a bioreactor with reciprocal mixing that produces specific physical properties that differ from those of conventional bioreactors with rotary paddle stirring. Moreover, such reactors not only provide a homogeneous environment but also allow the control of spheroid size by changing the mixing speed. In this study, we applied this bioreactor to the large-scale cultivation of hiPSCs. Approximately 10 billion hiPSCs were obtained from 2.0 L of culture, and the high expression of pluripotency markers was maintained. Our findings indicate that a bioreactor with reciprocal mixing can be used for large-scale hiPSC cultivation.
Collapse
Affiliation(s)
- Masashi Ueki
- Glycometabolic Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.
| | - Tadashi Suzuki
- Glycometabolic Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yoshikazu Kato
- Glycometabolic Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Mixing Technology Laboratory, Satake Multimix Corporation, 60 Niizo, Toda, Saitama 335-0021, Japan
| |
Collapse
|
7
|
Komosa ER, Lin WH, Mahadik B, Bazzi MS, Townsend D, Fisher JP, Ogle BM. A novel perfusion bioreactor promotes the expansion of pluripotent stem cells in a 3D-bioprinted tissue chamber. Biofabrication 2023; 16:014101. [PMID: 37906964 PMCID: PMC10636629 DOI: 10.1088/1758-5090/ad084a] [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: 10/09/2022] [Revised: 10/15/2023] [Accepted: 10/31/2023] [Indexed: 11/02/2023]
Abstract
While the field of tissue engineering has progressed rapidly with the advent of 3D bioprinting and human induced pluripotent stem cells (hiPSCs), impact is limited by a lack of functional, thick tissues. One way around this limitation is to 3D bioprint tissues laden with hiPSCs. In this way, the iPSCs can proliferate to populate the thick tissue mass prior to parenchymal cell specification. Here we design a perfusion bioreactor for an hiPSC-laden, 3D-bioprinted chamber with the goal of proliferating the hiPSCs throughout the structure prior to differentiation to generate a thick tissue model. The bioreactor, fabricated with digital light projection, was optimized to perfuse the interior of the hydrogel chamber without leaks and to provide fluid flow around the exterior as well, maximizing nutrient delivery throughout the chamber wall. After 7 days of culture, we found that intermittent perfusion (15 s every 15 min) at 3 ml min-1provides a 1.9-fold increase in the density of stem cell colonies in the engineered tissue relative to analogous chambers cultured under static conditions. We also observed a more uniform distribution of colonies within the tissue wall of perfused structures relative to static controls, reflecting a homogeneous distribution of nutrients from the culture media. hiPSCs remained pluripotent and proliferative with application of fluid flow, which generated wall shear stresses averaging ∼1.0 dyn cm-2. Overall, these promising outcomes following perfusion of a stem cell-laden hydrogel support the production of multiple tissue types with improved thickness, and therefore increased function and utility.
Collapse
Affiliation(s)
- Elizabeth R Komosa
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States of America
- NIBIB/NIH Center for Engineering Complex Tissues, College Park, MD, United States of America
| | - Wei-Han Lin
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States of America
| | - Bhushan Mahadik
- NIBIB/NIH Center for Engineering Complex Tissues, College Park, MD, United States of America
- Fishell Department of Bioengineering, University of Maryland, College Park, MD, United States of America
| | - Marisa S Bazzi
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, United States of America
| | - DeWayne Townsend
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States of America
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States of America
| | - John P Fisher
- NIBIB/NIH Center for Engineering Complex Tissues, College Park, MD, United States of America
- Fishell Department of Bioengineering, University of Maryland, College Park, MD, United States of America
| | - Brenda M Ogle
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States of America
- NIBIB/NIH Center for Engineering Complex Tissues, College Park, MD, United States of America
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States of America
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States of America
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN, United States of America
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States of America
| |
Collapse
|
8
|
Hogrebe NJ, Ishahak M, Millman JR. Developments in stem cell-derived islet replacement therapy for treating type 1 diabetes. Cell Stem Cell 2023; 30:530-548. [PMID: 37146579 PMCID: PMC10167558 DOI: 10.1016/j.stem.2023.04.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/20/2023] [Accepted: 04/05/2023] [Indexed: 05/07/2023]
Abstract
The generation of islet-like endocrine clusters from human pluripotent stem cells (hPSCs) has the potential to provide an unlimited source of insulin-producing β cells for the treatment of diabetes. In order for this cell therapy to become widely adopted, highly functional and well-characterized stem cell-derived islets (SC-islets) need to be manufactured at scale. Furthermore, successful SC-islet replacement strategies should prevent significant cell loss immediately following transplantation and avoid long-term immune rejection. This review highlights the most recent advances in the generation and characterization of highly functional SC-islets as well as strategies to ensure graft viability and safety after transplantation.
Collapse
Affiliation(s)
- Nathaniel J Hogrebe
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63130, USA.
| | - Matthew Ishahak
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63130, USA
| | - Jeffrey R Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO 63130, USA; Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive, St. Louis, MO 63130, USA.
| |
Collapse
|
9
|
In Vitro 3D Modeling of Neurodegenerative Diseases. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010093. [PMID: 36671665 PMCID: PMC9855033 DOI: 10.3390/bioengineering10010093] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/29/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023]
Abstract
The study of neurodegenerative diseases (such as Alzheimer's disease, Parkinson's disease, Huntington's disease, or amyotrophic lateral sclerosis) is very complex due to the difficulty in investigating the cellular dynamics within nervous tissue. Despite numerous advances in the in vivo study of these diseases, the use of in vitro analyses is proving to be a valuable tool to better understand the mechanisms implicated in these diseases. Although neural cells remain difficult to obtain from patient tissues, access to induced multipotent stem cell production now makes it possible to generate virtually all neural cells involved in these diseases (from neurons to glial cells). Many original 3D culture model approaches are currently being developed (using these different cell types together) to closely mimic degenerative nervous tissue environments. The aim of these approaches is to allow an interaction between glial cells and neurons, which reproduces pathophysiological reality by co-culturing them in structures that recapitulate embryonic development or facilitate axonal migration, local molecule exchange, and myelination (to name a few). This review details the advantages and disadvantages of techniques using scaffolds, spheroids, organoids, 3D bioprinting, microfluidic systems, and organ-on-a-chip strategies to model neurodegenerative diseases.
Collapse
|
10
|
Park S, Avera AD, Kim Y. BIOMANUFACTURING OF GLIOBLASTOMA ORGANOIDS EXHIBITING HIERARCHICAL AND SPATIALLY ORGANIZED TUMOR MICROENVIRONMENT VIA TRANSDIFFERENTIATION. Biotechnol Bioeng 2022; 119:3252-3274. [DOI: 10.1002/bit.28191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/14/2022] [Accepted: 07/20/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Seungjo Park
- Department of Chemical and Biological EngineeringThe University of AlabamaTuscaloosaAlabama
| | - Alexandra D. Avera
- Department of Chemical and Biological EngineeringThe University of AlabamaTuscaloosaAlabama
| | - Yonghyun Kim
- Department of Chemical and Biological EngineeringThe University of AlabamaTuscaloosaAlabama
| |
Collapse
|
11
|
Cacciamali A, Villa R, Dotti S. 3D Cell Cultures: Evolution of an Ancient Tool for New Applications. Front Physiol 2022; 13:836480. [PMID: 35936888 PMCID: PMC9353320 DOI: 10.3389/fphys.2022.836480] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 06/14/2022] [Indexed: 12/12/2022] Open
Abstract
Recently, research is undergoing a drastic change in the application of the animal model as a unique investigation strategy, considering an alternative approach for the development of science for the future. Although conventional monolayer cell cultures represent an established and widely used in vitro method, the lack of tissue architecture and the complexity of such a model fails to inform true biological processes in vivo. Recent advances in cell culture techniques have revolutionized in vitro culture tools for biomedical research by creating powerful three-dimensional (3D) models to recapitulate cell heterogeneity, structure and functions of primary tissues. These models also bridge the gap between traditional two-dimensional (2D) single-layer cultures and animal models. 3D culture systems allow researchers to recreate human organs and diseases in one dish and thus holds great promise for many applications such as regenerative medicine, drug discovery, precision medicine, and cancer research, and gene expression studies. Bioengineering has made an important contribution in the context of 3D systems using scaffolds that help mimic the microenvironments in which cells naturally reside, supporting the mechanical, physical and biochemical requirements for cellular growth and function. We therefore speak of models based on organoids, bioreactors, organ-on-a-chip up to bioprinting and each of these systems provides its own advantages and applications. All of these techniques prove to be excellent candidates for the development of alternative methods for animal testing, as well as revolutionizing cell culture technology. 3D systems will therefore be able to provide new ideas for the study of cellular interactions both in basic and more specialized research, in compliance with the 3R principle. In this review, we provide a comparison of 2D cell culture with 3D cell culture, provide details of some of the different 3D culture techniques currently available by discussing their strengths as well as their potential applications.
Collapse
Affiliation(s)
| | | | - Silvia Dotti
- *Correspondence: Andrea Cacciamali, ; Silvia Dotti,
| |
Collapse
|
12
|
Knock E, Julian LM. Building on a Solid Foundation: Adding Relevance and Reproducibility to Neurological Modeling Using Human Pluripotent Stem Cells. Front Cell Neurosci 2021; 15:767457. [PMID: 34867204 PMCID: PMC8637745 DOI: 10.3389/fncel.2021.767457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/20/2021] [Indexed: 11/25/2022] Open
Abstract
The brain is our most complex and least understood organ. Animal models have long been the most versatile tools available to dissect brain form and function; however, the human brain is highly distinct from that of standard model organisms. In addition to existing models, access to human brain cells and tissues is essential to reach new frontiers in our understanding of the human brain and how to intervene therapeutically in the face of disease or injury. In this review, we discuss current and developing culture models of human neural tissue, outlining advantages over animal models and key challenges that remain to be overcome. Our principal focus is on advances in engineering neural cells and tissue constructs from human pluripotent stem cells (PSCs), though primary human cell and slice culture are also discussed. By highlighting studies that combine animal models and human neural cell culture techniques, we endeavor to demonstrate that clever use of these orthogonal model systems produces more reproducible, physiological, and clinically relevant data than either approach alone. We provide examples across a range of topics in neuroscience research including brain development, injury, and cancer, neurodegenerative diseases, and psychiatric conditions. Finally, as testing of PSC-derived neurons for cell replacement therapy progresses, we touch on the advancements that are needed to make this a clinical mainstay.
Collapse
Affiliation(s)
- Erin Knock
- Research and Development, STEMCELL Technologies Inc., Vancouver, BC, Canada.,Department of Biological Sciences, Faculty of Science, Simon Fraser University, Burnaby, BC, Canada
| | - Lisa M Julian
- Department of Biological Sciences, Faculty of Science, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
13
|
Rios-Galacho M, Martinez-Moreno D, López-Ruiz E, Galvez-Martin P, Marchal JA. An overview on the manufacturing of functional and mature cellular skin substitutes. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:1035-1052. [PMID: 34652978 DOI: 10.1089/ten.teb.2021.0131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
There are different types of skin diseases due to chronic injuries that impede the natural healing process of the skin. Tissue engineering (TE) has focused on the development of bioengineered skin or skin substitutes that cover the wound, providing the necessary care to restore the functionality of injured skin. There are two types of substitutes: acellular skin substitutes (ASSs), which offer a low response of the body, and cellular skin substitutes (CSSs), which incorporate living cells and appear as a great alternative in the treatment of skin injuries due to them presenting a greater interaction and integration with the rest of the body. For the development of a CSS, it is necessary to select the most suitable biomaterials, cell components, and methodology of biofabrication for the wound to be treated. Moreover, these CSSs are immature substitutes that must undergo a maturing process in specific bioreactors, guaranteeing their functionality. The bioreactor simulates the natural state of maturation of the skin by controlling parameters such as temperature, pressure, or humidity, allowing a homogeneous maturation of the CSSs in an aseptic environment. The use of bioreactors not only contributes to the maturation of the CSSs, but also offers a new way of obtaining large sections of skin substitutes or natural skin from small portions acquired from the patient, donor, or substitute. Based on the innovation of this technology and the need to develop efficient CSSs, this work offers an update on bioreactor technology in the field of skin regeneration.
Collapse
Affiliation(s)
| | | | - Elena López-Ruiz
- Universidad de Jaen, 16747, Department of Health Sciences, Jaen, Andalucía, Spain;
| | | | - Juan Antonio Marchal
- University of Granada, humqn Anatomy and embriology, avd del conocimiento nº 11, Granada, Granada, Spain, 18016;
| |
Collapse
|
14
|
Advancing Regenerative Medicine Through the Development of Scaffold, Cell Biology, Biomaterials and Strategies of Smart Material. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2021. [DOI: 10.1007/s40883-021-00227-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
15
|
Ueki M, Tansho N, Sato M, Kanamori H, Kato Y. Improved cultivation of Chinese hamster ovary cells in bioreactor with reciprocal mixing. J Biosci Bioeng 2021; 132:531-536. [PMID: 34474981 DOI: 10.1016/j.jbiosc.2021.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/21/2021] [Accepted: 08/16/2021] [Indexed: 12/14/2022]
Abstract
We have constructed a new bioreactor with reciprocal mixing that is better suited for the cultivation of delicate animal cells. In-silico simulation (computational fluid dynamics) suggested both maximum and average shear stresses in the bioreactor with reciprocal mixing to be remarkably lower than in a conventional bioreactor with rotary mixing. Although we could not find any difference in growth speed and cell density between the bioreactors with reciprocal and rotary mixing, we did find cell viability in the reciprocal-mixing bioreactor to be retained longer than in the rotary-paddle bioreactor. This implied that cell culture in a bioreactor with reciprocal mixing could be prolonged for the production of target proteins. Leakage of lactate dehydrogenase activity into the culture medium was suppressed much more in the reciprocal-mixing bioreactor than in the rotary-paddle one. Production of human tissue plasminogen activator in the former system was also observed to be much higher than in the latter. Therefore, a bioreactor with reciprocal mixing was concluded to be better suited for the cultivation of animal cells and efficient production of proteins, such as antibody drugs and various growth factors.
Collapse
Affiliation(s)
- Masashi Ueki
- Animal Cell Incubator Laboratory, RIKEN Innovation Center, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Glycometabolic Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.
| | - Noriyuki Tansho
- Animal Cell Incubator Laboratory, RIKEN Innovation Center, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Mixing Technology Laboratory, Satake Chemical Equipment Mfg., Ltd., 60 Niizo, Toda, Saitama 335-0021, Japan
| | - Makoto Sato
- Animal Cell Incubator Laboratory, RIKEN Innovation Center, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Mixing Technology Laboratory, Satake Chemical Equipment Mfg., Ltd., 60 Niizo, Toda, Saitama 335-0021, Japan
| | - Hisayuki Kanamori
- Animal Cell Incubator Laboratory, RIKEN Innovation Center, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Mixing Technology Laboratory, Satake Chemical Equipment Mfg., Ltd., 60 Niizo, Toda, Saitama 335-0021, Japan
| | - Yoshikazu Kato
- Animal Cell Incubator Laboratory, RIKEN Innovation Center, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Mixing Technology Laboratory, Satake Chemical Equipment Mfg., Ltd., 60 Niizo, Toda, Saitama 335-0021, Japan
| |
Collapse
|
16
|
Yamamoto R, Kino-Oka M. Design of suspension culture system with bubble sparging for human induced pluripotent stem cells in a plastic fluid. J Biosci Bioeng 2021; 132:190-197. [PMID: 34052116 DOI: 10.1016/j.jbiosc.2021.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 11/29/2022]
Abstract
Bubble sparging has been used to supply oxygen to large-scale bioreactor systems. However, sparged bubbles cause cell death by rupturing due to shear stress, and the foam layer carries a risk of contamination. Large-scale culture of human induced pluripotent stem cells (hiPSCs) is required for manufacturing, but hiPSCs show high sensitivity to shear stress, and also, aseptic processing is important for their expansion. In this study, a culture system with bubble sparging for hiPSC proliferation was designed using a plastic fluid as a culture medium. The rising bubble velocity in the plastic fluid decreased and was lower than that in a Newtonian fluid when the time interval between bubbles generation, Δt, was greater than 0.14 s. Under this condition, aggregate distribution in the plastic fluid was maintained without liquid flow. Although large aeration induced aggregate coalescence and growth inhibition, the apparent specific growth rate at Δt > 0.14 s increased with an increase in the aeration rate, and the maximum value was similar to that of the conventional suspension culture in a stirred bioreactor system. The gas hold-up in the plastic fluid was higher than that in a Newtonian fluid because of the lower rising bubble velocity, which leads to the suppression of bubble sparging. Therefore, our results indicated that using a plastic fluid leads to a more efficient oxygen supply without agitation in a spatial-temporal phase-transition culture system.
Collapse
Affiliation(s)
- Riku Yamamoto
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
17
|
Hydrodynamics and mass transfer in spinner flasks: Implications for large scale cultured meat production. Biochem Eng J 2021. [DOI: 10.1016/j.bej.2020.107864] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
18
|
Jeske R, Lewis S, Tsai AC, Sanders K, Liu C, Yuan X, Li Y. Agitation in a Microcarrier-based Spinner Flask Bioreactor Modulates Homeostasis of Human Mesenchymal Stem Cells. Biochem Eng J 2021; 168. [PMID: 33967591 DOI: 10.1016/j.bej.2021.107947] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Human mesenchymal stem cells (hMSCs) are well known in cell therapy due to their secretion of trophic factors, multipotent differentiation potential, and ability for self-renewal. As a result, the number of clinical trials has been steadily increasing over the last decade highlighting the need for in vitro systems capable of producing large quantities of cells to meet growing demands. However, hMSCs are highly sensitive to microenvironment conditions, including shear stress caused by dynamic bioreactor systems, and can lead to alteration of cellular homeostasis. In this study, hMSCs were expanded on microcarriers within a 125 mL spinner flask bioreactor system. Our results demonstrate a three-fold expansion over seven days. Furthermore, our results show that culturing hMSCs in the microcarrier-based suspension bioreactor (compared to static planar culture) results in smaller cell size and higher levels of reactive oxidative species (ROS) and ROS regulator Sirtuin-3, which have implications on the nicotinamide adenine dinucleotide metabolic pathway and metabolic homeostasis. In addition, hMSCs in the bioreactor showed the increased Prostaglandin E2 secretion as well as reduced the Indoleamine-pyrrole 2,3-dioxygenase secretion upon stimulus with interferon gamma. The results of this study provide understanding of potential hMSC physiology alterations impacted by bioreactor microenvironment during scalable production of hMSCs for biomanufacturing and clinical trials.
Collapse
Affiliation(s)
- Richard Jeske
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| | - Shaquille Lewis
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| | - Ang-Chen Tsai
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| | - Kevin Sanders
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| | - Chang Liu
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| | - Xuegang Yuan
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States.,The National High Magnetic Field Laboratory, Florida State University, Tallahassee, Florida
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| |
Collapse
|
19
|
Marchesi N, Barbieri A, Fahmideh F, Govoni S, Ghidoni A, Parati G, Vanoli E, Pascale A, Calvillo L. Use of dual-flow bioreactor to develop a simplified model of nervous-cardiovascular systems crosstalk: A preliminary assessment. PLoS One 2020; 15:e0242627. [PMID: 33253266 PMCID: PMC7703955 DOI: 10.1371/journal.pone.0242627] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic conditions requiring long-term rehabilitation therapies, such as hypertension, stroke, or cancer, involve complex interactions between various systems/organs of the body and mutual influences, thus implicating a multiorgan approach. The dual-flow IVTech LiveBox2 bioreactor is a recently developed inter-connected dynamic cell culture model able to mimic organ crosstalk, since cells belonging to different organs can be connected and grown under flow conditions in a more physiological environment. This study aims to setup for the first time a 2-way connected culture of human neuroblastoma cells, SH-SY5Y, and Human Coronary Artery Smooth Muscle Cells, HCASMC through a dual-flow IVTech LiveBox2 bioreactor, in order to represent a simplified model of nervous-cardiovascular systems crosstalk, possibly relevant for the above-mentioned diseases. The system was tested by treating the cells with 10nM angiotensin II (AngII) inducing PKCβII/HuR/VEGF pathway activation, since AngII and PKCβII/HuR/VEGF pathway are relevant in cardiovascular and neuroscience research. Three different conditions were applied: 1- HCASMC and SH-SY5Y separately seeded in petri dishes (static condition); 2- the two cell lines separately seeded under flow (dynamic condition); 3- the two lines, seeded in dynamic conditions, connected, each maintaining its own medium, with a membrane as interface for biohumoral changes between the two mediums, and then treated. We detected that only in condition 3 there was a synergic AngII-dependent VEGF production in SH-SY5Y cells coupled to an AngII-dependent PKCβII/HuR/VEGF pathway activation in HCASMC, consistent with the observed physiological response in vivo. HCASMC response to AngII seems therefore to be generated by/derived from the reciprocal cell crosstalk under the dynamic inter-connection ensured by the dual flow LiveBox 2 bioreactor. This system can represent a useful tool for studying the crosstalk between organs, helpful for instance in rehabilitation research or when investigating chronic diseases; further, it offers the advantageous opportunity of cultivating each cell line in its own medium, thus mimicking, at least in part, distinct tissue milieu.
Collapse
Affiliation(s)
- Nicoletta Marchesi
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Annalisa Barbieri
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Foroogh Fahmideh
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Stefano Govoni
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Alice Ghidoni
- Center for Cardiac Arrhythmias of Genetic Origin, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Gianfranco Parati
- Department of Cardiovascular, Neural and Metabolic Sciences, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy
| | - Emilio Vanoli
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Cardiovascular Department, IRCCS Multimedica, Sesto San Giovanni, Italy
| | - Alessia Pascale
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Laura Calvillo
- Department of Cardiovascular, Neural and Metabolic Sciences, IRCCS Istituto Auxologico Italiano, Milan, Italy
| |
Collapse
|
20
|
Aggarwal V, Miranda O, Johnston PA, Sant S. Three dimensional engineered models to study hypoxia biology in breast cancer. Cancer Lett 2020; 490:124-142. [PMID: 32569616 PMCID: PMC7442747 DOI: 10.1016/j.canlet.2020.05.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/13/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022]
Abstract
Breast cancer is the second leading cause of mortality among women worldwide. Despite the available therapeutic regimes, variable treatment response is reported among different breast cancer subtypes. Recently, the effects of the tumor microenvironment on tumor progression as well as treatment responses have been widely recognized. Hypoxia and hypoxia inducible factors in the tumor microenvironment have long been known as major players in tumor progression and survival. However, the majority of our understanding of hypoxia biology has been derived from two dimensional (2D) models. Although many hypoxia-targeted therapies have elicited promising results in vitro and in vivo, these results have not been successfully translated into clinical trials. These limitations of 2D models underscore the need to develop and integrate three dimensional (3D) models that recapitulate the complex tumor-stroma interactions in vivo. This review summarizes role of hypoxia in various hallmarks of cancer progression. We then compare traditional 2D experimental systems with novel 3D tissue-engineered models giving accounts of different bioengineering platforms available to develop 3D models and how these 3D models are being exploited to understand the role of hypoxia in breast cancer progression.
Collapse
Affiliation(s)
- Vaishali Aggarwal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Oshin Miranda
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Paul A Johnston
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; UPMC-Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; UPMC-Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
21
|
Samaras JJ, Micheletti M, Ducci A. Flow, suspension, and mixing dynamics in
DASGIP
bioreactors: Part 1. AIChE J 2020. [DOI: 10.1002/aic.17014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jasmin J. Samaras
- Department of Biochemical Engineering University College London London UK
| | - Martina Micheletti
- Department of Biochemical Engineering University College London London UK
| | - Andrea Ducci
- Department of Mechanical Engineering University College London London UK
| |
Collapse
|
22
|
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
|
23
|
Parrotta EI, Lucchino V, Scaramuzzino L, Scalise S, Cuda G. Modeling Cardiac Disease Mechanisms Using Induced Pluripotent Stem Cell-Derived Cardiomyocytes: Progress, Promises and Challenges. Int J Mol Sci 2020; 21:E4354. [PMID: 32575374 PMCID: PMC7352327 DOI: 10.3390/ijms21124354] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are a class of disorders affecting the heart or blood vessels. Despite progress in clinical research and therapy, CVDs still represent the leading cause of mortality and morbidity worldwide. The hallmarks of cardiac diseases include heart dysfunction and cardiomyocyte death, inflammation, fibrosis, scar tissue, hyperplasia, hypertrophy, and abnormal ventricular remodeling. The loss of cardiomyocytes is an irreversible process that leads to fibrosis and scar formation, which, in turn, induce heart failure with progressive and dramatic consequences. Both genetic and environmental factors pathologically contribute to the development of CVDs, but the precise causes that trigger cardiac diseases and their progression are still largely unknown. The lack of reliable human model systems for such diseases has hampered the unraveling of the underlying molecular mechanisms and cellular processes involved in heart diseases at their initial stage and during their progression. Over the past decade, significant scientific advances in the field of stem cell biology have literally revolutionized the study of human disease in vitro. Remarkably, the possibility to generate disease-relevant cell types from induced pluripotent stem cells (iPSCs) has developed into an unprecedented and powerful opportunity to achieve the long-standing ambition to investigate human diseases at a cellular level, uncovering their molecular mechanisms, and finally to translate bench discoveries into potential new therapeutic strategies. This review provides an update on previous and current research in the field of iPSC-driven cardiovascular disease modeling, with the aim of underlining the potential of stem-cell biology-based approaches in the elucidation of the pathophysiology of these life-threatening diseases.
Collapse
|
24
|
Suraiya AB, Hun ML, Truong VX, Forsythe JS, Chidgey AP. Gelatin-Based 3D Microgels for In Vitro T Lineage Cell Generation. ACS Biomater Sci Eng 2020; 6:2198-2208. [PMID: 33455336 DOI: 10.1021/acsbiomaterials.9b01610] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
T cells are predominantly produced by the thymus and play a significant role in maintaining our adaptive immune system. Physiological involution of the thymus occurs gradually with age, compromising naive T cell output, which can have severe clinical complications. Also, T cells are utilized as therapeutic agents in cancer immunotherapies. Therefore, there is an increasing need for strategies aimed at generating naive T cells. The majority of in vitro T cell generation studies are performed in two-dimensional (2D) cultures, which ignore the physiological thymic microenvironment and are not scalable; therefore, we applied a new three-dimensional (3D) approach. Here, we use a gelatin-based 3D microgel system for T lineage induction by co-culturing OP9-DL4 cells and mouse fetal-liver-derived hematopoietic stem cells (HSCs). Flow cytometric analysis revealed that microgel co-cultures supported T lineage induction similar to 2D cultures while providing a 3D environment. We also encapsulated mouse embryonic thymic epithelial cells (TECs) within the microgels to provide a defined 3D culture platform. The microgel system supported TEC maintenance and retained their phenotype. Together, these data show that our microgel system has the capacity for TEC maintenance and induction of in vitro T lineage differentiation with potential for scalability.
Collapse
Affiliation(s)
- Anisha B Suraiya
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia.,Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia
| | - Michael L Hun
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia
| | - Vinh X Truong
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia
| | - John S Forsythe
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia
| | - Ann P Chidgey
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, Melbourne 3800, Australia
| |
Collapse
|
25
|
A conceptual air-lift reactor design for large scale animal cell cultivation in the context of in vitro meat production. Chem Eng Sci 2020. [DOI: 10.1016/j.ces.2019.115269] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
26
|
Brancato V, Oliveira JM, Correlo VM, Reis RL, Kundu SC. Could 3D models of cancer enhance drug screening? Biomaterials 2019; 232:119744. [PMID: 31918229 DOI: 10.1016/j.biomaterials.2019.119744] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/29/2019] [Accepted: 12/25/2019] [Indexed: 02/06/2023]
Abstract
Cancer is a multifaceted pathology, where cellular and acellular players interact to drive cancer progression and, in the worst-case, metastasis. The current methods to investigate the heterogeneous nature of cancer are inadequate, since they rely on 2D cell cultures and animal models. The cell line-based drug efficacy and toxicity assays are not able to predict the tumor response to anti-cancer agents and it is already widely discussed how molecular pathway are not recapitulated in vitro so called flat biology. On the other side, animal models often fail to detect the side-effects of drugs, mimic the metastatic progression or the interaction between cancer and immune system, due to biologic difference in human and animals. Moreover, ethical and regulatory issues limit animal experimentation. Every year pharma/biotech companies lose resources in drug discovery and testing processes that are successful only in 5% of the cases. There is an urgent need to validate accurate and predictive platforms in order to enhance drug-testing process taking into account the physiopathology of the tumor microenvironment. Three dimensional in vitro tumor models could enhance drug manufactures in developing effective drugs for cancer diseases. The 3D in vitro cancer models can improve the predictability of toxicity and drug sensitivity in cancer. Despite the demonstrated advantages of 3D in vitro disease systems when compared to 2D culture and animal models, they still do not reach the standardization required for preclinical trials. This review highlights in vitro models that may be used as preclinical models, accelerating the drug development process towards more precise and personalized standard of care for cancer patients. We describe the state-of-the art of 3D in vitro culture systems, with a focus on how these different approaches could be coupled in order to achieve a compromise between standardization and reliability in recapitulating tumor microenvironment and drug response.
Collapse
Affiliation(s)
- Virginia Brancato
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| | - Joaquim Miguel Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - Vitor Manuel Correlo
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - Rui Luis Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - Subhas C Kundu
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
27
|
Hu X, Xu J, Li W, Li L, Parungao R, Wang Y, Zheng S, Nie Y, Liu T, Song K. Therapeutic "Tool" in Reconstruction and Regeneration of Tissue Engineering for Osteochondral Repair. Appl Biochem Biotechnol 2019; 191:785-809. [PMID: 31863349 DOI: 10.1007/s12010-019-03214-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
Abstract
Repairing osteochondral defects to restore joint function is a major challenge in regenerative medicine. However, with recent advances in tissue engineering, the development of potential treatments is promising. In recent years, in addition to single-layer scaffolds, double-layer or multilayer scaffolds have been prepared to mimic the structure of articular cartilage and subchondral bone for osteochondral repair. Although there are a range of different cells such as umbilical cord stem cells, bone marrow mesenchyml stem cell, and others that can be used, the availability, ease of preparation, and the osteogenic and chondrogenic capacity of these cells are important factors that will influence its selection for tissue engineering. Furthermore, appropriate cell proliferation and differentiation of these cells is also key for the optimal repair of osteochondral defects. The development of bioreactors has enhanced methods to stimulate the proliferation and differentiation of cells. In this review, we summarize the recent advances in tissue engineering, including the development of layered scaffolds, cells, and bioreactors that have changed the approach towards the development of novel treatments for osteochondral repair.
Collapse
Affiliation(s)
- Xueyan Hu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Jie Xu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Wenfang Li
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian, 116024, China.,Key Laboratory of Biological Medicines, Universities of Shandong Province Weifang Key Laboratory of Antibody Medicines, School of Bioscience and Technology, Weifang Medical University, Weifang, 261053, China
| | - Liying Li
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Roxanne Parungao
- Burns Research Group, ANZAC Research Institute, University of Sydney, Concord, NSW, 2139, Australia
| | - Yiwei Wang
- Burns Research Group, ANZAC Research Institute, University of Sydney, Concord, NSW, 2139, Australia
| | - Shuangshuang Zheng
- Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou, 450000, China
| | - Yi Nie
- Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou, 450000, China. .,Key Laboratory of Green Process and Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Tianqing Liu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian, 116024, China.
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian, 116024, China.
| |
Collapse
|
28
|
Tee CA, Yang Z, Yin L, Wu Y, Han J, Lee EH. Improved zonal chondrocyte production protocol integrating size-based inertial spiral microchannel separation and dynamic microcarrier culture for clinical application. Biomaterials 2019; 220:119409. [DOI: 10.1016/j.biomaterials.2019.119409] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/30/2019] [Accepted: 08/02/2019] [Indexed: 10/26/2022]
|
29
|
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
|
30
|
Gelse K, Biggemann J, Stumpf M, Halmheu M, Grüneboom A, Kleyer A, Scholtysek C, Pachowsky ML, Hueber A, Krönke G, Fey T. Modular Lattice Constructs for Biological Joint Resurfacing. Tissue Eng Part A 2019; 25:1053-1062. [DOI: 10.1089/ten.tea.2018.0231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Kolja Gelse
- Department of Orthopedic and Trauma Surgery, University Hospital Erlangen, Erlangen, Germany
| | - Jonas Biggemann
- Department of Materials Science (Glass and Ceramics), University of Erlangen-Nuernberg, Erlangen, Germany
| | - Martin Stumpf
- Department of Materials Science (Glass and Ceramics), University of Erlangen-Nuernberg, Erlangen, Germany
| | - Melissa Halmheu
- Department of Materials Science (Glass and Ceramics), University of Erlangen-Nuernberg, Erlangen, Germany
| | - Anika Grüneboom
- Department of Internal Medicine 3—Rheumatology and Immunology, University Hospital Erlangen, Erlangen, Germany
| | - Arnd Kleyer
- Department of Internal Medicine 3—Rheumatology and Immunology, University Hospital Erlangen, Erlangen, Germany
| | - Carina Scholtysek
- Department of Internal Medicine 3—Rheumatology and Immunology, University Hospital Erlangen, Erlangen, Germany
| | - Milena L. Pachowsky
- Department of Orthopedic and Trauma Surgery, University Hospital Erlangen, Erlangen, Germany
| | - Axel Hueber
- Department of Internal Medicine 3—Rheumatology and Immunology, University Hospital Erlangen, Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3—Rheumatology and Immunology, University Hospital Erlangen, Erlangen, Germany
| | - Tobias Fey
- Department of Materials Science (Glass and Ceramics), University of Erlangen-Nuernberg, Erlangen, Germany
- Frontier Research Institute for Materials Science, Nagoya Institute of Technology, Nagoya, Japan
| |
Collapse
|
31
|
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
|
32
|
Saberi A, Zhang S, van den Bersselaar C, Kandail H, den Toonder JMJ, Kurniawan NA. A stirring system using suspended magnetically-actuated pillars for controlled cell clustering. SOFT MATTER 2019; 15:1435-1443. [PMID: 30666323 DOI: 10.1039/c8sm01957f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Controlled stirring of a solution is a household task in most laboratories. However, most stirring methods are perturbative or require vessels with predefined shapes and sizes. Here we propose a novel stirring system based on suspended magnetically-actuated pillars (SMAPs), inspired by the ability of biological flagella and cilia to generate flow. We fabricated flexible, millimeter-scale magnetic pillars grafted on transparent polydimethylsiloxane (PDMS) substrates and built a simple actuation setup to control the motion of the pillars remotely. We tested the system with a standard 24-well plate routinely used in most research laboratories and demonstrate that the magnetic actuation results in robust bending of the pillars and large-scale fluid flow in the wells. Quantitative analysis using computational fluid dynamics modeling indicates that the flow profile in the well can be tuned by modulating the applied magnetic field and the geometries of the well and the pillar. Finally, we show that, by employing the stirring system in a standard cell culture plate, we were able to obtain controlled clustering of cells. The SMAP stirring system is therefore a promising cost-effective and scalable stirring approach for various types of studies involving colloids as well as soft and biological materials.
Collapse
Affiliation(s)
- Aref Saberi
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | - Shuaizhong Zhang
- Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands and Institute for Complex Molecular Systems, Eindhoven, The Netherlands
| | - Carola van den Bersselaar
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | - Harkamaljot Kandail
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | - Jaap M J den Toonder
- Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands and Institute for Complex Molecular Systems, Eindhoven, The Netherlands
| | - Nicholas A Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands. and Institute for Complex Molecular Systems, Eindhoven, The Netherlands
| |
Collapse
|
33
|
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: 21] [Impact Index Per Article: 3.5] [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
|
34
|
Ahmed S, Chauhan VM, Ghaemmaghami AM, Aylott JW. New generation of bioreactors that advance extracellular matrix modelling and tissue engineering. Biotechnol Lett 2019; 41:1-25. [PMID: 30368691 PMCID: PMC6313369 DOI: 10.1007/s10529-018-2611-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/26/2018] [Indexed: 12/12/2022]
Abstract
Bioreactors hold a lot of promise for tissue engineering and regenerative medicine applications. They have multiple uses including cell cultivation for therapeutic production and for in vitro organ modelling to provide a more physiologically relevant environment for cultures compared to conventional static conditions. Bioreactors are often used in combination with scaffolds as the nutrient flow can enhance oxygen and diffusion throughout the 3D constructs to prevent the formation of necrotic cores. A variety of scaffolds have been fabricated to achieve a structural architecture that mimic native extracellular matrix. Future developments of in vitro models will incorporate the ability to non-invasively monitor the cellular microenvironment to enhance the understanding of in vitro conditions. This review details current advancements in bioreactor and scaffold systems and provides insight on how in vitro models can be augmented for future biomedical applications.
Collapse
Affiliation(s)
- Shehnaz Ahmed
- School of Pharmacy, University of Nottingham, Boots Sciences Building, University Park, Nottingham, UK
| | - Veeren M. Chauhan
- School of Pharmacy, University of Nottingham, Boots Sciences Building, University Park, Nottingham, UK
| | - Amir M. Ghaemmaghami
- School of Life Sciences, University of Nottingham, Life Sciences Building, University Park, Nottingham, NG7 2RD UK
| | - Jonathan W. Aylott
- School of Pharmacy, University of Nottingham, Boots Sciences Building, University Park, Nottingham, UK
| |
Collapse
|
35
|
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
|
36
|
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
|
37
|
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: 18] [Impact Index Per Article: 2.6] [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
|
38
|
Lin H, Li Q, Wang O, Rauch J, Harm B, Viljoen HJ, Zhang C, Van Wyk E, Zhang C, Lei Y. Automated Expansion of Primary Human T Cells in Scalable and Cell-Friendly Hydrogel Microtubes for Adoptive Immunotherapy. Adv Healthc Mater 2018; 7:e1701297. [PMID: 29749707 DOI: 10.1002/adhm.201701297] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 03/15/2018] [Indexed: 01/17/2023]
Abstract
Adoptive immunotherapy is a highly effective strategy for treating many human cancers, such as melanoma, cervical cancer, lymphoma, and leukemia. Here, a novel cell culture technology is reported for expanding primary human T cells for adoptive immunotherapy. T cells are suspended and cultured in microscale alginate hydrogel tubes (AlgTubes) that are suspended in the cell culture medium in a culture vessel. The hydrogel tubes protect cells from hydrodynamic stresses and confine the cell mass less than 400 µm (in radial diameter) to ensure efficient mass transport, creating a cell-friendly microenvironment for growing T cells. This system is simple, scalable, highly efficient, defined, cost-effective, and compatible with current good manufacturing practices. Under optimized culture conditions, the AlgTubes enable culturing T cells with high cell viability, low DNA damage, high growth rate (≈320-fold expansion over 14 days), high purity (≈98% CD3+), and high yield (≈3.2 × 108 cells mL-1 hydrogel). All offer considerable advantages compared to current T cell culturing approaches. This new culture technology can significantly reduce the culture volume, time, and cost, while increasing the production.
Collapse
Affiliation(s)
- Haishuang Lin
- Department of Chemical and Biomolecular Engineering; University of Nebraska; Lincoln 68588 NE USA
| | - Qiang Li
- Department of Chemical and Biomolecular Engineering; University of Nebraska; Lincoln 68588 NE USA
- Biomedical Engineering Program; University of Nebraska; Lincoln 68588 NE USA
| | - Ou Wang
- Department of Chemical and Biomolecular Engineering; University of Nebraska; Lincoln 68588 NE USA
- Biomedical Engineering Program; University of Nebraska; Lincoln 68588 NE USA
| | - Jack Rauch
- Department of Chemical and Biomolecular Engineering; University of Nebraska; Lincoln 68588 NE USA
| | - Braden Harm
- Department of Chemical and Biomolecular Engineering; University of Nebraska; Lincoln 68588 NE USA
| | - Hendrik J. Viljoen
- Department of Chemical and Biomolecular Engineering; University of Nebraska; Lincoln 68588 NE USA
| | - Chi Zhang
- Department of Radiation Oncology; College of Medicine; University of Nebraska Medical Center; Omaha 68198 NE USA
| | | | - Chi Zhang
- School of Biological Science; University of Nebraska; Lincoln 68588 NE USA
| | - Yuguo Lei
- Department of Chemical and Biomolecular Engineering; University of Nebraska; Lincoln 68588 NE USA
- Biomedical Engineering Program; University of Nebraska; Lincoln 68588 NE USA
- Mary and Dick Holland Regenerative Medicine Program; University of Nebraska Medical Center; Omaha 68198 NE USA
- Fred & Pamela Buffett Cancer Center; University of Nebraska Medical Center; Omaha 68106 NE USA
| |
Collapse
|
39
|
Jafarkhani M, Salehi Z, Kowsari-Esfahan R, Shokrgozar MA, Rezaa Mohammadi M, Rajadas J, Mozafari M. Strategies for directing cells into building functional hearts and parts. Biomater Sci 2018; 6:1664-1690. [PMID: 29767196 DOI: 10.1039/c7bm01176h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025]
Abstract
The increasing population of patients with heart disease and the limited availability of organs for transplantation have encouraged multiple strategies to fabricate healthy implantable cardiac tissues. One of the main challenges in cardiac tissue engineering is to direct cell behaviors to form functional three-dimensional (3D) biomimetic constructs. This article provides a brief review on various cell sources used in cardiac tissue engineering and highlights the effect of scaffold-based signals such as topographical and biochemical cues and stiffness. Then, conventional and novel micro-engineered bioreactors for the development of functional cardiac tissues will be explained. Bioreactor-based signals including mechanical and electrical cues to control cardiac cell behavior will also be elaborated in detail. Finally, the application of computational fluid dynamics to design suitable bioreactors will be discussed. This review presents the current state-of-the-art, emerging directions and future trends that critically appraise the concepts involved in various approaches to direct cells for building functional hearts and heart parts.
Collapse
Affiliation(s)
- Mahboubeh Jafarkhani
- School of Chemical Engineering, College of Engineering, University of Tehran, Iran.
| | | | | | | | | | | | | |
Collapse
|
40
|
Metabolic Reprogramming and the Recovery of Physiological Functionality in 3D Cultures in Micro-Bioreactors. Bioengineering (Basel) 2018. [PMID: 29518979 PMCID: PMC5874888 DOI: 10.3390/bioengineering5010022] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The recovery of physiological functionality, which is commonly seen in tissue mimetic three-dimensional (3D) cellular aggregates (organoids, spheroids, acini, etc.), has been observed in cells of many origins (primary tissues, embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and immortal cell lines). This plurality and plasticity suggest that probably several basic principles promote this recovery process. The aim of this study was to identify these basic principles and describe how they are regulated so that they can be taken in consideration when micro-bioreactors are designed. Here, we provide evidence that one of these basic principles is hypoxia, which is a natural consequence of multicellular structures grown in microgravity cultures. Hypoxia drives a partial metabolic reprogramming to aerobic glycolysis and an increased anabolic synthesis. A second principle is the activation of cytoplasmic glutaminolysis for lipogenesis. Glutaminolysis is activated in the presence of hypo- or normo-glycaemic conditions and in turn is geared to the hexosamine pathway. The reducing power needed is produced in the pentose phosphate pathway, a prime function of glucose metabolism. Cytoskeletal reconstruction, histone modification, and the recovery of the physiological phenotype can all be traced to adaptive changes in the underlying cellular metabolism. These changes are coordinated by mTOR/Akt, p53 and non-canonical Wnt signaling pathways, while myc and NF-kB appear to be relatively inactive. Partial metabolic reprogramming to aerobic glycolysis, originally described by Warburg, is independent of the cell’s rate of proliferation, but is interwoven with the cells abilities to execute advanced functionality needed for replicating the tissues physiological performance.
Collapse
|
41
|
Li Q, Lin H, Rauch J, Deleyrolle LP, Reynolds BA, Viljoen HJ, Zhang C, Zhang C, Gu L, Van Wyk E, Lei Y. Scalable Culturing of Primary Human Glioblastoma Tumor-Initiating Cells with a Cell-Friendly Culture System. Sci Rep 2018; 8:3531. [PMID: 29476107 PMCID: PMC5824878 DOI: 10.1038/s41598-018-21927-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/13/2018] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma is the most aggressive and deadly brain cancer. There is growing interest to develop drugs that specifically target to glioblastoma tumor-initiating cells (TICs). However, the cost-effective production of large numbers of high quality glioblastoma TICs for drug discovery with current cell culturing technologies remains very challenging. Here, we report a new method that cultures glioblastoma TICs in microscale alginate hydrogel tubes (or AlgTubes). The AlgTubes allowed long-term culturing (~50 days, 10 passages) of glioblastoma TICs with high growth rate (~700-fold expansion/14 days), high cell viability and high volumetric yield (~3.0 × 108 cells/mL) without losing the stem cell properties, all offered large advancements over current culturing methods. This method can be applied for the scalable production of glioblastoma TICs at affordable cost for drug discovery.
Collapse
Affiliation(s)
- Qiang Li
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, USA.,Biomedical Engineering Program, University of Nebraska, Lincoln, Nebraska, USA
| | - Haishuang Lin
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, USA
| | - Jack Rauch
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, USA
| | - Loic P Deleyrolle
- Department of Neurosurgery, University of Florida College of Medicine, McKnight Brain Institute, Gainesville, Florida, USA
| | - Brent A Reynolds
- Department of Neurosurgery, University of Florida College of Medicine, McKnight Brain Institute, Gainesville, Florida, USA
| | - Hendrik J Viljoen
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, USA
| | - Chi Zhang
- School of Biological Science, University of Nebraska, Lincoln, Nebraska, USA
| | - Chi Zhang
- Department of Radiation Oncology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Linxia Gu
- Department of Mechanical & Materials Engineering, University of Nebraska, Lincoln, Nebraska, USA
| | | | - Yuguo Lei
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, USA. .,Biomedical Engineering Program, University of Nebraska, Lincoln, Nebraska, USA. .,Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, USA. .,Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| |
Collapse
|
42
|
Li Q, Lin H, Du Q, Liu K, Wang O, Evans C, Christian H, Zhang C, Lei Y. Scalable and physiologically relevant microenvironments for human pluripotent stem cell expansion and differentiation. Biofabrication 2018; 10:025006. [PMID: 29319535 DOI: 10.1088/1758-5090/aaa6b5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Human pluripotent stem cells (hPSCs) are required in large numbers for various biomedical applications. However, the scalable and cost-effective culturing of high quality hPSCs and their derivatives remains very challenging. Here, we report a novel and physiologically relevant 3D culture system (called the AlgTube cell culture system) for hPSC expansion and differentiation. With this system, cells are processed into and cultured in microscale alginate hydrogel tubes that are suspended in the cell culture medium in a culture vessel. The hydrogel tubes protect cells from hydrodynamic stresses in the culture vessel and limit the cell mass smaller than 400 μm in diameter to ensure efficient mass transport, creating cell-friendly microenvironments for growing cells. This system is simple, scalable, highly efficient, defined and compatible with the current good manufacturing practices. Under optimized culture conditions, the AlgTubes enabled long-term culture of hPSCs (>10 passages, >50 days) with high cell viability, high growth rate (1000-fold expansion over 10 days per passage), high purity (>95% Oct4+) and high yield (5.0 × 108 cells ml-1), all of which offer considerable advantages compared to current approaches. Moreover, the AlgTubes enabled directed differentiation of hPSCs into various tissue cells. This system can be readily scaled to support research from basic biological study to clinical development and the future industry-scale production.
Collapse
Affiliation(s)
- Qiang Li
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, United States of America. Biomedical Engineering Program, University of Nebraska, Lincoln, Nebraska, United States of America
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Li J, Li Y, Gao B, Qin C, He Y, Xu F, Yang H, Lin M. Engineering mechanical microenvironment of macrophage and its biomedical applications. Nanomedicine (Lond) 2018; 13:555-576. [PMID: 29334336 DOI: 10.2217/nnm-2017-0324] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Macrophages are the most plastic cells in the hematopoietic system and can be widely found in almost all tissues. Recently studies have shown that mechanical cues (e.g., matrix stiffness and stress/strain) can significantly affect macrophage behaviors. Although existing reviews on the physical and mechanical cues that regulate the macrophage's phenotype are available, engineering mechanical microenvironment of macrophages in vitro as well as a comprehensive overview and prospects for their biomedical applications (e.g., tissue engineering and immunotherapy) has yet to be summarized. Thus, this review provides an overview on the existing methods for engineering mechanical microenvironment of macrophages in vitro and then a section on their biomedical applications and further perspectives are presented.
Collapse
Affiliation(s)
- Jing Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, PR China.,Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, PR China.,Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.,Key Laboratory on Space Physics and Chemistry of Ministry of Education and Key Laboratory on Macromolecular Science & Technology of Shanxi Province, Department of Applied Chemistry, School of Science, Northwestern Polytechnical University, 710072, P.R China
| | - Yuhui Li
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.,The Key Library of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
| | - Bin Gao
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.,The Key Library of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China.,Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, P.R. China
| | - Chuanguang Qin
- Key Laboratory on Space Physics and Chemistry of Ministry of Education and Key Laboratory on Macromolecular Science & Technology of Shanxi Province, Department of Applied Chemistry, School of Science, Northwestern Polytechnical University, 710072, P.R China
| | - Yining He
- College of Food Science and Engineering, Northwest A & F University Yangling Shaanxi 712100 China
| | - Feng Xu
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.,The Key Library of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, PR China.,Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, PR China
| | - Min Lin
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.,The Key Library of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
| |
Collapse
|
44
|
Li Q, Wang Q, Wang O, Shao K, Lin H, Lei Y. A simple and scalable hydrogel-based system for culturing protein-producing cells. PLoS One 2018; 13:e0190364. [PMID: 29293594 PMCID: PMC5749782 DOI: 10.1371/journal.pone.0190364] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 12/13/2017] [Indexed: 01/08/2023] Open
Abstract
Recombinant protein therapeutics have become important components of the modern medicine. Majority of them are produced with mammalian cells that are cultured either through adherent culturing, in which cells are cultured on substrates, or suspension culturing, in which cells are suspended and cultured in agitated cell culture medium in a culture vessel. The adherent cell culturing method is limited by its low yield. In suspension culturing, cells need extensive genetic manipulation to grow as single cells at high density, which is time- and labor-consuming. Here, we report a new method, which utilizes a thermoreversible hydrogel as the scaffold for culturing protein-expressing cells. The hydrogel scaffolds not only provide 3D spaces for the cells, but also act as physical barriers to prevent excessive cellular agglomeration and protect cells from the hydrodynamic stresses. As a result, cells can grow at high viability, high growth rate, and extremely high yield even without genetic manipulations. The cell yield in the hydrogels is around 20 times of the suspension culturing. In addition, the protein productivity per cell per day in the hydrogel is higher than the adherent culturing method. This new method is simple, scalable and defined. It will be of great value for both the research laboratories and pharmaceutical industry for producing proteins.
Collapse
Affiliation(s)
- Qiang Li
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, United States of America
- Biomedical Engineering Program, University of Nebraska, Lincoln, Nebraska, United States of America
| | - Qiaofeng Wang
- Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ou Wang
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, United States of America
- Biomedical Engineering Program, University of Nebraska, Lincoln, Nebraska, United States of America
| | - Kaifeng Shao
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Haishuang Lin
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, United States of America
| | - Yuguo Lei
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, United States of America
- Biomedical Engineering Program, University of Nebraska, Lincoln, Nebraska, United States of America
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
45
|
Rigamonti A, Repetti GG, Sun C, Price FD, Reny DC, Rapino F, Weisinger K, Benkler C, Peterson QP, Davidow LS, Hansson EM, Rubin LL. Large-Scale Production of Mature Neurons from Human Pluripotent Stem Cells in a Three-Dimensional Suspension Culture System. Stem Cell Reports 2017; 6:993-1008. [PMID: 27304920 PMCID: PMC4912437 DOI: 10.1016/j.stemcr.2016.05.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 05/19/2016] [Accepted: 05/19/2016] [Indexed: 01/11/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) offer a renewable source of cells that can be expanded indefinitely and differentiated into virtually any type of cell in the human body, including neurons. This opens up unprecedented possibilities to study neuronal cell and developmental biology and cellular pathology of the nervous system, provides a platform for the screening of chemical libraries that affect these processes, and offers a potential source of transplantable cells for regenerative approaches to neurological disease. However, defining protocols that permit a large number and high yield of neurons has proved difficult. We present differentiation protocols for the generation of distinct subtypes of neurons in a highly reproducible manner, with minimal experiment-to-experiment variation. These neurons form synapses with neighboring cells, exhibit spontaneous electrical activity, and respond appropriately to depolarization. hPSC-derived neurons exhibit a high degree of maturation and survive in culture for up to 4-5 months, even without astrocyte feeder layers.
Collapse
Affiliation(s)
- Alessandra Rigamonti
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Giuliana G Repetti
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Chicheng Sun
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Feodor D Price
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Danielle C Reny
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Francesca Rapino
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Karen Weisinger
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Chen Benkler
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Quinn P Peterson
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Lance S Davidow
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Emil M Hansson
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Medicine, KI-AZ Integrated Cardio Metabolic Centre, Karolinska Institutet, NOVUM, Hälsovägen 7, 141 57 Huddinge, Sweden
| | - Lee L Rubin
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA.
| |
Collapse
|
46
|
Yan Y, Song L, Madinya J, Ma T, Li Y. Derivation of Cortical Spheroids from Human Induced Pluripotent Stem Cells in a Suspension Bioreactor. Tissue Eng Part A 2017; 24:418-431. [PMID: 28825364 DOI: 10.1089/ten.tea.2016.0400] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) emerge as a promising source to construct human brain-like tissues, spheroids, or organoids in vitro for disease modeling and drug screening. A suspension bioreactor can be used to generate large size of brain organoids from hiPSCs through enhanced diffusion, but the influence of a dynamic bioreactor culture environment on neural tissue patterning from hiPSCs has not been well understood. The objective of this study is to assess the influence of a suspension bioreactor culture on cortical spheroid (i.e., forebrain-like aggregates) formation from hiPSCs. Single undifferentiated hiPSK3 cells or preformed embryoid bodies were inoculated into the bioreactor. Aggregate size distribution, neural marker expression (e.g., Nestin, PAX6, β-tubulin III, and MAP-2), and cortical tissue patterning markers (e.g., TBR1, BRN2, SATB2, and vGlut1) were evaluated with static control. Bioreactor culture was found to promote the expression of TBR1, a deep cortical layer VI marker, and temporally affect SATB2, a superficial cortical layer II-IV marker that appears later according to inside-out cortical tissue development. Prolonged culture after 70 days showed layer-specific cortical structure in the spheroids. Differential expression of matrix metalloproteinase-2 and -3 was also observed for bioreactor and static culture. The altered expression of cortical markers by a suspension bioreactor indicates the importance of culture environment on cortical tissue development from hiPSCs.
Collapse
Affiliation(s)
- Yuanwei Yan
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Liqing Song
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Jason Madinya
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| |
Collapse
|
47
|
Kropp C, Massai D, Zweigerdt R. Progress and challenges in large-scale expansion of human pluripotent stem cells. Process Biochem 2017. [DOI: 10.1016/j.procbio.2016.09.032] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
48
|
Kuo YC, Rajesh R. Guided differentiation and tissue regeneration of induced pluripotent stem cells using biomaterials. J Taiwan Inst Chem Eng 2017. [DOI: 10.1016/j.jtice.2017.04.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
49
|
McKee C, Chaudhry GR. Advances and challenges in stem cell culture. Colloids Surf B Biointerfaces 2017; 159:62-77. [PMID: 28780462 DOI: 10.1016/j.colsurfb.2017.07.051] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 07/04/2017] [Accepted: 07/22/2017] [Indexed: 12/12/2022]
Abstract
Stem cells (SCs) hold great promise for cell therapy, tissue engineering, and regenerative medicine as well as pharmaceutical and biotechnological applications. They have the capacity to self-renew and the ability to differentiate into specialized cell types depending upon their source of isolation. However, use of SCs for clinical applications requires a high quality and quantity of cells. This necessitates large-scale expansion of SCs followed by efficient and homogeneous differentiation into functional derivatives. Traditional methods for maintenance and expansion of cells rely on two-dimensional (2-D) culturing techniques using plastic culture plates and xenogenic media. These methods provide limited expansion and cells tend to lose clonal and differentiation capacity upon long-term passaging. Recently, new approaches for the expansion of SCs have emphasized three-dimensional (3-D) cell growth to mimic the in vivo environment. This review provides a comprehensive compendium of recent advancements in culturing SCs using 2-D and 3-D techniques involving spheroids, biomaterials, and bioreactors. In addition, potential challenges to achieve billion-fold expansion of cells are discussed.
Collapse
Affiliation(s)
- Christina McKee
- Department of Biological Sciences , Oakland University, Rochester, MI, 48309, USA; OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA
| | - G Rasul Chaudhry
- Department of Biological Sciences , Oakland University, Rochester, MI, 48309, USA; OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA.
| |
Collapse
|
50
|
Nampe D, Joshi R, Keller K, Zur Nieden NI, Tsutsui H. Impact of fluidic agitation on human pluripotent stem cells in stirred suspension culture. Biotechnol Bioeng 2017; 114:2109-2120. [PMID: 28480972 DOI: 10.1002/bit.26334] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 05/03/2017] [Accepted: 05/05/2017] [Indexed: 12/23/2022]
Abstract
The success of human pluripotent stem cells (hPSCs) as a source of future cell therapies hinges, in part, on the availability of a robust and scalable culture system that can readily produce a clinically relevant number of cells and their derivatives. Stirred suspension culture has been identified as one such promising platform due to its ease of use, scalability, and widespread use in the pharmaceutical industry (e.g., CHO cell-based production of therapeutic proteins) among others. However, culture of undifferentiated hPSCs in stirred suspension is a relatively new development within the past several years, and little is known beyond empirically optimized culture parameters. In particular, detailed characterizations of different agitation rates and their influence on the propagation of hPSCs are often not reported in the literature. In the current study, we systematically investigated various agitation rates to characterize their impact on cell yield, viability, and the maintenance of pluripotency. Additionally, we closely examined the distribution of cell aggregates and how the observed culture outcomes are attributed to their size distribution. Overall, our results showed that moderate agitation maximized the propagation of hPSCs to approximately 38-fold over 7 days by keeping the cell aggregates below the critical size, beyond which the cells are impacted by the diffusion limit, while limiting cell death caused by excessive fluidic forces. Furthermore, we observed that fluidic agitation could regulate not only cell aggregation, but also expression of some key signaling proteins in hPSCs. This indicates a new possibility to guide stem cell fate determination by fluidic agitation in stirred suspension cultures. Biotechnol. Bioeng. 2017;114: 2109-2120. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Daniel Nampe
- Department of Mechanical Engineering, University of California, Riverside, California 92521.,Department of Bioengineering, University of California, Riverside, California 92521.,Stem Cell Center, University of California, Riverside, California 92521
| | - Ronak Joshi
- Stem Cell Center, University of California, Riverside, California 92521.,Department of Cell Biology and Neuroscience, University of California, Riverside, California 92521
| | - Kevin Keller
- Stem Cell Center, University of California, Riverside, California 92521.,Department of Cell Biology and Neuroscience, University of California, Riverside, California 92521
| | - Nicole I Zur Nieden
- Stem Cell Center, University of California, Riverside, California 92521.,Department of Cell Biology and Neuroscience, University of California, Riverside, California 92521
| | - Hideaki Tsutsui
- Department of Mechanical Engineering, University of California, Riverside, California 92521.,Department of Bioengineering, University of California, Riverside, California 92521.,Stem Cell Center, University of California, Riverside, California 92521
| |
Collapse
|