1
|
Danev N, Poggi JM, Dewever EA, Bartlett AP, Oliveira L, Huntimer L, Harman RM, Van de Walle GR. Immortalized mammosphere-derived epithelial cells retain a bioactive secretome with antimicrobial, regenerative, and immunomodulatory properties. Stem Cell Res Ther 2024; 15:429. [PMID: 39543714 PMCID: PMC11566417 DOI: 10.1186/s13287-024-04019-1] [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: 08/08/2024] [Accepted: 10/25/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND The secretome of primary bovine mammosphere-derived epithelial cells (MDECs) has been shown to exert antimicrobial, regenerative, and immunomodulatory properties in vitro, which warrants its study as a potential biologic treatment with the potential to be translated to human medicine. Currently, the use of the MDEC secretome as a therapy is constrained by the limited life span of primary cell cultures and the decrease of secretome potency over cell passages. METHODS To address these limitations, early-passage bovine MDECs were immortalized using hTERT, a human telomerase reverse transcriptase. The primary and immortal MDECs were compared morphologically, transcriptomically, and phenotypically. The functional properties and proteomic profiles of the secretome of both cell lines were evaluated and compared. All experiments were performed with both low and high passage cell cultures. RESULTS We confirmed through in vitro experiments that the secretome of immortalized MDECs, unlike that of primary cells, maintained antimicrobial and pro-migratory properties over passages, while pro-angiogenic effects of the secretome from both primary and immortalized MDECs were lost when the cells reached high passage. The secretome from primary and immortalized MDECs, at low and high passages exerted immunomodulatory effects on neutrophils in vitro. CONCLUSIONS High passage immortalized MDECs retain a bioactive secretome with antimicrobial, regenerative, and immunomodulatory properties, suggesting they may serve as a consistent cell source for therapeutic use.
Collapse
Affiliation(s)
- Nikola Danev
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA
| | - Julia M Poggi
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA
| | - Emilie A Dewever
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA
| | - Arianna P Bartlett
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA
| | - Leane Oliveira
- Elanco Animal Health, 2500 Innovation Way, Indianapolis, IN, 46241, USA
| | - Lucas Huntimer
- Elanco Animal Health, 2500 Innovation Way, Indianapolis, IN, 46241, USA
| | - Rebecca M Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14853, USA.
- Department of Veterinary Pathobiology, Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Roslin, Midlothian, UK.
| |
Collapse
|
2
|
Guan Y, Yang B, Xu W, Li D, Wang S, Ren Z, Zhang J, Zhang T, Liu XZ, Li J, Li C, Meng F, Han F, Wu T, Wang Y, Peng J. Cell-derived extracellular matrix materials for tissue engineering. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:1007-1021. [PMID: 34641714 DOI: 10.1089/ten.teb.2021.0147] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The involvement of cell-derived extracellular matrix (CDM) in assembling tissue engineering scaffolds has yielded significant results. CDM possesses excellent characteristics, such as ideal cellular microenvironment mimicry and good biocompatibility, which make it a popular research direction in the field of bionanomaterials. CDM has significant advantages as an expansion culture substrate for stem cells, including stabilization of phenotype, reversal of senescence, and guidance of specific differentiation. In addition, the applications of CDM-assembled tissue engineering scaffolds for disease simulation and tissue organ repair are comprehensively summarized; the focus is mainly on bone and cartilage repair, skin defect or wound healing, engineered blood vessels, peripheral nerves, and periodontal tissue repair. We consider CDM a highly promising bionic biomaterial for tissue engineering applications and propose a vision for its comprehensive development.
Collapse
Affiliation(s)
- Yanjun Guan
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Boyao Yang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Wenjing Xu
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Dongdong Li
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Sidong Wang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Zhiqi Ren
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Jian Zhang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Tieyuan Zhang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Xiu-Zhi Liu
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Junyang Li
- Nankai University School of Medicine, 481107, Tianjin, Tianjin, China.,Chinese PLA General Hospital, 104607, Beijing, Beijing, China;
| | - Chaochao Li
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Fanqi Meng
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China.,Peking University People's Hospital, 71185, Department of spine surgery, Beijing, China;
| | - Feng Han
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Tong Wu
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Yu Wang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China.,Nantong University, 66479, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu, China;
| | - Jiang Peng
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China.,Nantong University, 66479, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu, China;
| |
Collapse
|
3
|
Borys BS, So T, Roberts EL, Ferrie L, Larijani L, Abraham B, Krawetz R, Rancourt DE, Kallos MS. Large-scale expansion of feeder-free mouse embryonic stem cells serially passaged in stirred suspension bioreactors at low inoculation densities directly from cryopreservation. Biotechnol Bioeng 2020; 117:1316-1328. [PMID: 31960947 DOI: 10.1002/bit.27279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/06/2020] [Accepted: 01/15/2020] [Indexed: 12/23/2022]
Abstract
Embryonic stem cells (ESCs) have almost unlimited proliferation capacity in vitro and can retain the ability to contribute to all cell lineages, making them an ideal platform material for cell-based therapies. ESCs are traditionally cultured in static flasks on a feeder layer of murine embryonic fibroblast cells. Although sufficient to generate cells for research purposes, this approach is impractical to achieve large quantities for clinical applications. In this study, we have developed protocols that address a variety of challenges that currently bottleneck clinical translation of ESCs expanded in stirred suspension bioreactors. We demonstrated that mouse ESCs (mESCs) cryopreserved in the absence of feeder cells could be thawed directly into stirred suspension bioreactors at extremely low inoculation densities (100 cells/ml). These cells sustained proliferative capacity through multiple passages and various reactor sizes and geometries, producing clinically relevant numbers (109 cells) and maintaining pluripotency phenotypic and functional properties. Passages were completed in stirred suspension bioreactors of increasing scale, under defined batch conditions which greatly improved resource efficiency. Output mESCs were analyzed for pluripotency marker expression (SSEA-1, SOX-2, and Nanog) through flow cytometry, and spontaneous differentiation and teratoma analysis was used to demonstrate functional maintenance of pluripotency.
Collapse
Affiliation(s)
- Breanna S Borys
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada
| | - Tania So
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - Erin L Roberts
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada
| | - Leah Ferrie
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada
| | - Leila Larijani
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Brett Abraham
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - Roman Krawetz
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Derrick E Rancourt
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michael S Kallos
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada.,Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
4
|
Gryshkov O, Müller M, Leal-Marin S, Mutsenko V, Suresh S, Kapralova VM, Glasmacher B. Advances in the application of electrohydrodynamic fabrication for tissue engineering. ACTA ACUST UNITED AC 2019. [DOI: 10.1088/1742-6596/1236/1/012024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
5
|
Chimenti I, Massai D, Morbiducci U, Beltrami AP, Pesce M, Messina E. Stem Cell Spheroids and Ex Vivo Niche Modeling: Rationalization and Scaling-Up. J Cardiovasc Transl Res 2017; 10:150-166. [PMID: 28289983 DOI: 10.1007/s12265-017-9741-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/27/2017] [Indexed: 02/08/2023]
Abstract
Improved protocols/devices for in vitro culture of 3D cell spheroids may provide essential cues for proper growth and differentiation of stem/progenitor cells (S/PCs) in their niche, allowing preservation of specific features, such as multi-lineage potential and paracrine activity. Several platforms have been employed to replicate these conditions and to generate S/PC spheroids for therapeutic applications. However, they incompletely reproduce the niche environment, with partial loss of its highly regulated network, with additional hurdles in the field of cardiac biology, due to debated resident S/PCs therapeutic potential and clinical translation. In this contribution, the essential niche conditions (metabolic, geometric, mechanical) that allow S/PCs maintenance/commitment will be discussed. In particular, we will focus on both existing bioreactor-based platforms for the culture of S/PC as spheroids, and on possible criteria for the scaling-up of niche-like spheroids, which could be envisaged as promising tools for personalized cardiac regenerative medicine, as well as for high-throughput drug screening.
Collapse
Affiliation(s)
- Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Rome, Italy
| | - Diana Massai
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Umberto Morbiducci
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | | | - Maurizio Pesce
- Tissue Engineering Research Unit, "Centro Cardiologico Monzino", IRCCS, Milan, Italy
| | - Elisa Messina
- Department of Pediatrics and Infant Neuropsychiatry, "Umberto I" Hospital, "La Sapienza" University, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
6
|
Galvanauskas V, Grincas V, Simutis R, Kagawa Y, Kino-oka M. Current state and perspectives in modeling and control of human pluripotent stem cell expansion processes in stirred-tank bioreactors. Biotechnol Prog 2017; 33:355-364. [DOI: 10.1002/btpr.2431] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 12/10/2016] [Indexed: 01/02/2023]
Affiliation(s)
| | - Vykantas Grincas
- Department of Automation; Kaunas University of Technology; Kaunas Lithuania
| | - Rimvydas Simutis
- Department of Automation; Kaunas University of Technology; Kaunas Lithuania
| | - Yuki Kagawa
- Department of Biotechnology; Osaka University; Osaka Japan
| | | |
Collapse
|
7
|
Leong MF, Lu HF, Lim TC, Du C, Ma NK, Wan AC. Electrospun polystyrene scaffolds as a synthetic substrate for xeno-free expansion and differentiation of human induced pluripotent stem cells. Acta Biomater 2016; 46:266-277. [PMID: 27667015 DOI: 10.1016/j.actbio.2016.09.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 09/16/2016] [Accepted: 09/22/2016] [Indexed: 01/01/2023]
Abstract
The use of human induced pluripotent stem cells (hiPSCs) for clinical tissue engineering applications requires expansion and differentiation of the cells using defined, xeno-free substrates. The screening and selection of suitable synthetic substrates however, is tedious, as their performance relies on the inherent material properties. In the present work, we demonstrate an alternative concept for xeno-free expansion and differentiation of hiPSCs using synthetic substrates, which hinges on the structure-function relationship between electrospun polystyrene scaffolds (ESPS) and pluripotent stem cell growth. ESPS of differential porosity was obtained by fusing the fibers at different temperatures. The more porous, loosely fused scaffolds were found to efficiently trap the cells, leading to a large number of three-dimensional (3D) aggregates which were shown to be pluripotent colonies. Immunostaining, PCR analyses, in vitro differentiation and in vivo teratoma formation studies demonstrated that these hiPSC aggregates could be cultured for up to 10 consecutive passages (P10) with maintenance of pluripotency. Flow cytometry showed that more than 80% of the cell population stained positive for the pluripotent marker OCT4 at P1, P5 and P10. P10 cells could be differentiated to neuronal-like cells and cultured within the ESPS for up to 18months. Our results suggest the usefulness of a generic class of synthetic substrates, exemplified by ESPS, for 'trapped aggregate culture' of hiPSCs. STATEMENT OF SIGNIFICANCE To realize the potential of human induced pluripotent stem cells (hiPSCs) in clinical medicine, robust, xeno-free substrates for expansion and differentiation of iPSCs are required. In the existing literature, synthetic materials have been reported that meet the requirement for non-xenogeneic substrates. However, the self-renewal and differentiation characteristics of hiPSCs are affected differently by the biocompatibility and physico-chemical properties of individual substrates. Although some rules based on chemical structure and substrate rigidity have been developed, most of these efforts are still empirical, and most synthetic substrates must still be rigorously screened for suitability. In this paper, we demonstrate an alternative concept for xeno-free expansion and differentiation of hiPSCs using synthetic substrates, which hinges on the structure-function relationship between electrospun polystyrene scaffolds (ESPS) and pluripotent stem cell growth. ESPS of differential porosity was obtained by fusing the fibers at different temperatures. The more porous, loosely fused scaffold was found to efficiently trap the cells, leading to a large number of three-dimensional (3D) aggregates. In the form of these trapped aggregates, we showed that hiPSCs could be cultured for up to 10 consecutive passages (P10) with maintenance of pluripotency, following which they could be differentiated to a chosen lineage. We believe that this novel, generic class of synthetic substrates that employs 'trapped aggregate culture' for expansion and differentiation of hiPSCs is an important conceptual advance, and would be of high interest to the readership of Acta Biomaterialia.
Collapse
|
8
|
Karimi M, Bahrami S, Mirshekari H, Basri SMM, Nik AB, Aref AR, Akbari M, Hamblin MR. Microfluidic systems for stem cell-based neural tissue engineering. LAB ON A CHIP 2016; 16:2551-71. [PMID: 27296463 PMCID: PMC4935609 DOI: 10.1039/c6lc00489j] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Neural tissue engineering aims at developing novel approaches for the treatment of diseases of the nervous system, by providing a permissive environment for the growth and differentiation of neural cells. Three-dimensional (3D) cell culture systems provide a closer biomimetic environment, and promote better cell differentiation and improved cell function, than could be achieved by conventional two-dimensional (2D) culture systems. With the recent advances in the discovery and introduction of different types of stem cells for tissue engineering, microfluidic platforms have provided an improved microenvironment for the 3D-culture of stem cells. Microfluidic systems can provide more precise control over the spatiotemporal distribution of chemical and physical cues at the cellular level compared to traditional systems. Various microsystems have been designed and fabricated for the purpose of neural tissue engineering. Enhanced neural migration and differentiation, and monitoring of these processes, as well as understanding the behavior of stem cells and their microenvironment have been obtained through application of different microfluidic-based stem cell culture and tissue engineering techniques. As the technology advances it may be possible to construct a "brain-on-a-chip". In this review, we describe the basics of stem cells and tissue engineering as well as microfluidics-based tissue engineering approaches. We review recent testing of various microfluidic approaches for stem cell-based neural tissue engineering.
Collapse
Affiliation(s)
- Mahdi Karimi
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Sajad Bahrami
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran. and Nanomedicine Research Association (NRA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hamed Mirshekari
- Advanced Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran, Iran.
| | - Seyed Masoud Moosavi Basri
- Bioenvironmental Research Center, Sharif University of Technology, Tehran, Iran. and Civil & Environmental Engineering Department, Shahid Beheshti University, Tehran, Iran
| | - Amirala Bakhshian Nik
- Department of Biomedical Engineering, Faculty of New Sciences and Technologies, University of Tehran, Iran.
| | - Amir R Aref
- Department of Cancer Biology, Center for Cancer Systems Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA and Department of Genetics, Harvard Medical School, Boston, MA 02215, USA.
| | - Mohsen Akbari
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA. and Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA. and Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA and Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| |
Collapse
|
9
|
Powers AD, Piras BA, Clark RK, Lockey TD, Meagher MM. Development and Optimization of AAV hFIX Particles by Transient Transfection in an iCELLis®Fixed-Bed Bioreactor. Hum Gene Ther Methods 2016; 27:112-21. [DOI: 10.1089/hgtb.2016.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Alicia D. Powers
- Department of Therapeutics Production & Quality, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Bryan A. Piras
- Department of Therapeutics Production & Quality, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Robert K. Clark
- Department of Therapeutics Production & Quality, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Timothy D. Lockey
- Department of Therapeutics Production & Quality, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Michael M. Meagher
- Department of Therapeutics Production & Quality, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
10
|
High-throughput screening approaches and combinatorial development of biomaterials using microfluidics. Acta Biomater 2016; 34:1-20. [PMID: 26361719 DOI: 10.1016/j.actbio.2015.09.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 09/07/2015] [Accepted: 09/08/2015] [Indexed: 12/11/2022]
Abstract
From the first microfluidic devices used for analysis of single metabolic by-products to highly complex multicompartmental co-culture organ-on-chip platforms, efforts of many multidisciplinary teams around the world have been invested in overcoming the limitations of conventional research methods in the biomedical field. Close spatial and temporal control over fluids and physical parameters, integration of sensors for direct read-out as well as the possibility to increase throughput of screening through parallelization, multiplexing and automation are some of the advantages of microfluidic over conventional, 2D tissue culture in vitro systems. Moreover, small volumes and relatively small cell numbers used in experimental set-ups involving microfluidics, can potentially decrease research cost. On the other hand, these small volumes and numbers of cells also mean that many of the conventional molecular biology or biochemistry assays cannot be directly applied to experiments that are performed in microfluidic platforms. Development of different types of assays and evidence that such assays are indeed a suitable alternative to conventional ones is a step that needs to be taken in order to have microfluidics-based platforms fully adopted in biomedical research. In this review, rather than providing a comprehensive overview of the literature on microfluidics, we aim to discuss developments in the field of microfluidics that can aid advancement of biomedical research, with emphasis on the field of biomaterials. Three important topics will be discussed, being: screening, in particular high-throughput and combinatorial screening; mimicking of natural microenvironment ranging from 3D hydrogel-based cellular niches to organ-on-chip devices; and production of biomaterials with closely controlled properties. While important technical aspects of various platforms will be discussed, the focus is mainly on their applications, including the state-of-the-art, future perspectives and challenges. STATEMENT OF SIGNIFICANCE Microfluidics, being a technology characterized by the engineered manipulation of fluids at the submillimeter scale, offers some interesting tools that can advance biomedical research and development. Screening platforms based on microfluidic technologies that allow high-throughput and combinatorial screening may lead to breakthrough discoveries not only in basic research but also relevant to clinical application. This is further strengthened by the fact that reliability of such screens may improve, since microfluidic systems allow close mimicking of physiological conditions. Finally, microfluidic systems are also very promising as micro factories of a new generation of natural or synthetic biomaterials and constructs, with finely controlled properties.
Collapse
|
11
|
Kumar A, Starly B. Large scale industrialized cell expansion: producing the critical raw material for biofabrication processes. Biofabrication 2015; 7:044103. [DOI: 10.1088/1758-5090/7/4/044103] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
12
|
Gelinsky M, Bernhardt A, Milan F. Bioreactors in tissue engineering: Advances in stem cell culture and three-dimensional tissue constructs. Eng Life Sci 2015. [DOI: 10.1002/elsc.201400216] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Michael Gelinsky
- Centre for Translational Bone; Joint and Soft Tissue Research; Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
| | - Anne Bernhardt
- Centre for Translational Bone; Joint and Soft Tissue Research; Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
| | - Falk Milan
- Centre for Translational Bone; Joint and Soft Tissue Research; Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden; Dresden Germany
| |
Collapse
|
13
|
Dai Y, Guo Y, Wang C, Liu Q, Yang Y, Li S, Guo X, Lian R, Yu R, Liu H, Chen J. Non-genetic direct reprogramming and biomimetic platforms in a preliminary study for adipose-derived stem cells into corneal endothelia-like cells. PLoS One 2014; 9:e109856. [PMID: 25333522 PMCID: PMC4198143 DOI: 10.1371/journal.pone.0109856] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 09/11/2014] [Indexed: 12/12/2022] Open
Abstract
Cell fate and function can be regulated and reprogrammed by intrinsic genetic program, extrinsic factors and niche microenvironment. Direct reprogramming has shown many advantages in the field of cellular reprogramming. Here we tried the possibility to generate corneal endothelia (CE) -like cells from human adipose-derived stem cells (ADSCs) by the non-genetic direct reprogramming of recombinant cell-penetrating proteins Oct4/Klf4/Sox2 (PTD-OKS) and small molecules (purmorphamine, RG108 and other reprogramming chemical reagents), as well as biomimetic platforms of simulate microgravity (SMG) bioreactor. Co-cultured with corneal cells and decellularized corneal ECM, Reprogrammed ADSCs revealed spherical growth and positively expressing Nanog for RT-PCR analysis and CD34 for immunofluorescence staining after 7 days-treatment of both purmorphamine and PTD-OKS (P-OKS) and in SMG culture. ADSCs changed to CEC polygonal morphology from spindle shape after the sequential non-genetic direct reprogramming and biomimetic platforms. At the same time, induced cells converted to weakly express CD31, AQP-1 and ZO-1. These findings demonstrated that the treatments were able to promote the stem-cell reprogramming for human ADSCs. Our study also indicates for the first time that SMG rotary cell culture system can be used as a non-genetic means to promote direct reprogramming. Our methods of reprogramming provide an alternative strategy for engineering patient-specific multipotent cells for cellular plasticity research and future autologous CEC replacement therapy that avoids complications associated with the use of human pluripotent stem cells.
Collapse
Affiliation(s)
- Ying Dai
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Yonglong Guo
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Chan Wang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Qing Liu
- Ophthalmology Department, First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Yan Yang
- Ophthalmology Department, First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Shanyi Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Xiaoling Guo
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Ruiling Lian
- Ophthalmology Department, First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Rongjie Yu
- Bioengineering Institute of Jinan University, Guangzhou, China
| | - Hongwei Liu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Jiansu Chen
- Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
- Ophthalmology Department, First Affiliated Hospital of Jinan University, Guangzhou, China
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
- * E-mail:
| |
Collapse
|
14
|
High throughput screening for biomaterials discovery. J Control Release 2014; 190:115-26. [DOI: 10.1016/j.jconrel.2014.06.045] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 06/23/2014] [Accepted: 06/23/2014] [Indexed: 01/29/2023]
|
15
|
Qi H, Huang G, Han YL, Lin W, Li X, Wang S, Lu TJ, Xu F. In vitro spatially organizing the differentiation in individual multicellular stem cell aggregates. Crit Rev Biotechnol 2014; 36:20-31. [PMID: 25025275 DOI: 10.3109/07388551.2014.922917] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
With significant potential as a robust source to produce specific somatic cells for regenerative medicine, stem cells have attracted increasing attention from both academia and government. In vivo, stem cell differentiation is a process under complicated regulations to precisely build tissue with unique spatial structures. Since multicellular spheroidal aggregates of stem cells, commonly called as embryoid bodies (EBs), are considered to be capable of recapitulating the events in early stage of embryonic development, a variety of methods have been developed to form EBs in vitro for studying differentiation of embryonic stem cells. The regulation of stem cell differentiation is crucial in directing stem cells to build tissue with the correct spatial architecture for specific functions. However, stem cells within the three-dimensional multicellular aggregates undergo differentiation in a less unpredictable and spatially controlled manner in vitro than in vivo. Recently, various microengineering technologies have been developed to manipulate stem cells in vitro in a spatially controlled manner. Herein, we take the spotlight on these technologies and researches that bring us the new potential for manipulation of stem cells for specific purposes.
Collapse
Affiliation(s)
- Hao Qi
- a MOE Key laboratory of Biomedical Information Engineering , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China .,b Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University , Xi'an , People's Republic of China .,c Department of Medical Genome Sciences , Graduate School of Frontier Sciences, University of Tokyo , Kashiwa , Chiba , Japan
| | - Guoyou Huang
- a MOE Key laboratory of Biomedical Information Engineering , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China .,b Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Yu Long Han
- a MOE Key laboratory of Biomedical Information Engineering , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China .,b Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Wang Lin
- a MOE Key laboratory of Biomedical Information Engineering , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China .,b Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Xiujun Li
- d Department of Chemistry , University of Texas at EI Paso , EI Paso , TX , USA , and
| | - Shuqi Wang
- e Brigham Women's Hospital, Harvard Medical School , Boston , MA , USA
| | - Tian Jian Lu
- b Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Feng Xu
- a MOE Key laboratory of Biomedical Information Engineering , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China .,b Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University , Xi'an , People's Republic of China
| |
Collapse
|
16
|
Lee JE, Lee DR. Human embryonic stem cells: derivation, maintenance and cryopreservation. Int J Stem Cells 2014; 4:9-17. [PMID: 24298329 DOI: 10.15283/ijsc.2011.4.1.9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2011] [Indexed: 12/29/2022] Open
Abstract
Human embryonic stem cells (hESCs) are the most powerful candidate for the treatment of incurable diseases through the replacement of damaged cells and/or tissues in patients, although there are some obstacles to overcome for the clinical application of hESCs such as the assurance of guided differentiation and control of the immune response following cell therapy or tissue grafting. To obtain genetically stable hESCs and use them clinically, it is important to develop appropriate culture conditions. Additionally, the establishment of a hESC bank with a large number of hESC lines will be required for their clinical application because each hESC line is directed to have a different differentiation ability and immune characteristics such as HLA type. In this review, we describe the derivation and culture conditions of hESCs based on recent advances. Then, we will introduce several cryopreservation methods for hESCs, which is important for the development of cell bank.
Collapse
|
17
|
A novel perfused rotary bioreactor for cardiomyogenesis of embryonic stem cells. Biotechnol Lett 2014; 36:947-60. [PMID: 24652542 DOI: 10.1007/s10529-014-1456-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 01/07/2014] [Indexed: 10/25/2022]
Abstract
Developments in bioprocessing technology play an important role for overcoming challenges in cardiac tissue engineering. To this end, our laboratory has developed a novel rotary perfused bioreactor for supporting three-dimensional cardiac tissue engineering. The dynamic culture environments provided by our novel perfused rotary bioreactor and/or the high-aspect rotating vessel produced constructs with higher viability and significantly higher cell numbers (up to 4 × 10(5) cells/bead) than static tissue culture flasks. Furthermore, cells in the perfused rotary bioreactor showed earlier gene expressions of cardiac troponin-T, α- and β-myosin heavy chains with higher percentages of cardiac troponin-I-positive cells and better uniformity of sacromeric α-actinin expression. A dynamic and perfused environment, as provided by this bioreactor, provides a superior culture performance in cardiac differentiation for embryonic stem cells particularly for larger 3D constructs.
Collapse
|
18
|
Sart S, Agathos SN, Li Y. Process engineering of stem cell metabolism for large scale expansion and differentiation in bioreactors. Biochem Eng J 2014. [DOI: 10.1016/j.bej.2014.01.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
19
|
Badenes SM, Fernandes TG, Rodrigues CAV, Diogo MM, Cabral JMS. Scalable expansion of human-induced pluripotent stem cells in xeno-free microcarriers. Methods Mol Biol 2014; 1283:23-9. [PMID: 25108454 DOI: 10.1007/7651_2014_106] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The expansion of human-induced pluripotent stem cells (hiPSCs) is commonly performed using feeder layers of mouse embryonic fibroblasts or in feeder-free conditions in two-dimensional culture platforms, which are associated with low production yields and lack of process control. Robust large-scale production of these cells under defined conditions has been one of the major challenges to fulfil the large cell number requirement for drug screening applications, toxicology assays, disease modeling and potential cellular therapies. Microcarrier-based systems, in particular, are a promising culture format since they provide a high surface-to-volume ratio and allow the scale-up of the process to stirred suspension bioreactors. In this context, this chapter describes a detailed methodology for the scalable expansion of hiPSCs in spinner flasks and using xeno-free microcarriers to allow further translation to Good Manufacturing Practice (GMP) conditions.
Collapse
Affiliation(s)
- Sara M Badenes
- Department of Bioengineering and IBB, Institute for Biotechnology and Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001, Lisbon, Portugal
| | | | | | | | | |
Collapse
|
20
|
Sart S, Agathos SN, Li Y. Engineering stem cell fate with biochemical and biomechanical properties of microcarriers. Biotechnol Prog 2013; 29:1354-66. [PMID: 24124017 DOI: 10.1002/btpr.1825] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 09/29/2013] [Indexed: 12/28/2022]
Abstract
Microcarriers have been widely used for various biotechnology applications because of their high scale-up potential, high reproducibility in regulating cellular behavior, and well-documented compliance with current Good Manufacturing Practices (cGMP). Recently, microcarriers have been emerging as a novel approach for stem cell expansion and differentiation, enabling potential scale-up of stem cell-derived products in large bioreactors. This review summarizes recent advances of using microcarriers in mesenchymal stem cell (MSC) and pluripotent stem cell (PSC) cultures. From the reported data, efficient expansion and differentiation of stem cells on microcarriers rely on their ability to modulate cell shape (i.e. round or spreading) and cell organization (i.e. aggregate size). Nonetheless, current screening of microcarriers remains empirical, and accurate understanding of how stem cells interact with microcarriers still remains unknown. This review suggests that accurate characterization of biochemical and biomechanical properties of microcarriers is required to fully exploit their potential in regulating stem cell fate decision. Due to the variety of microcarriers, such detailed analyses should lead to the rational design of application-specific microcarriers, enabling the exploitation of reproducible effects for large scale biomedical applications.
Collapse
Affiliation(s)
- Sébastien Sart
- Dept. of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL
| | | | | |
Collapse
|
21
|
Abbasalizadeh S, Baharvand H. Technological progress and challenges towards cGMP manufacturing of human pluripotent stem cells based therapeutic products for allogeneic and autologous cell therapies. Biotechnol Adv 2013; 31:1600-23. [PMID: 23962714 DOI: 10.1016/j.biotechadv.2013.08.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 06/20/2013] [Accepted: 08/12/2013] [Indexed: 12/16/2022]
Abstract
Recent technological advances in the generation, characterization, and bioprocessing of human pluripotent stem cells (hPSCs) have created new hope for their use as a source for production of cell-based therapeutic products. To date, a few clinical trials that have used therapeutic cells derived from hESCs have been approved by the Food and Drug Administration (FDA), but numerous new hPSC-based cell therapy products are under various stages of development in cell therapy-specialized companies and their future market is estimated to be very promising. However, the multitude of critical challenges regarding different aspects of hPSC-based therapeutic product manufacturing and their therapies have made progress for the introduction of new products and clinical applications very slow. These challenges include scientific, technological, clinical, policy, and financial aspects. The technological aspects of manufacturing hPSC-based therapeutic products for allogeneic and autologous cell therapies according to good manufacturing practice (cGMP) quality requirements is one of the most important challenging and emerging topics in the development of new hPSCs for clinical use. In this review, we describe main critical challenges and highlight a series of technological advances in all aspects of hPSC-based therapeutic product manufacturing including clinical grade cell line development, large-scale banking, upstream processing, downstream processing, and quality assessment of final cell therapeutic products that have brought hPSCs closer to clinical application and commercial cGMP manufacturing.
Collapse
Affiliation(s)
- Saeed Abbasalizadeh
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | |
Collapse
|
22
|
Selekman JA, Das A, Grundl NJ, Palecek SP. Improving efficiency of human pluripotent stem cell differentiation platforms using an integrated experimental and computational approach. Biotechnol Bioeng 2013; 110:3024-37. [PMID: 23740478 DOI: 10.1002/bit.24968] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 05/23/2013] [Accepted: 05/28/2013] [Indexed: 02/06/2023]
Abstract
Human pluripotent stem cells (hPSCs) have an unparalleled potential for tissue engineering applications including regenerative therapies and in vitro cell-based models for studying normal and diseased tissue morphogenesis, or drug and toxicological screens. While numerous hPSC differentiation methods have been developed to generate various somatic cell types, the potential of hPSC-based technologies is hinged on the ability to translate these established lab-scale differentiation systems to large-scale processes to meet the industrial and clinical demands for these somatic cell types. Here, we demonstrate a strategy for investigating the efficiency and scalability of hPSC differentiation platforms. Using two previously reported epithelial differentiation systems as models, we fit an ODE-based kinetic model to data representing dynamics of various cell subpopulations present in our culture. This fit was performed by estimating rate constants of each cell subpopulation's cell fate decisions (self-renewal, differentiation, death). Sensitivity analyses on predicted rate constants indicated which cell fate decisions had the greatest impact on overall epithelial cell yield in each differentiation process. In addition, we found that the final cell yield was limited by the self-renewal rate of either the progenitor state or the final differentiated state, depending on the differentiation protocol. Also, the relative impact of these cell fate decision rates was highly dependent on the maximum capacity of the cell culture system. Overall, we outline a novel approach for quantitative analysis of established laboratory-scale hPSC differentiation systems and this approach may ease development to produce large quantities of cells for tissue engineering applications.
Collapse
Affiliation(s)
- Joshua A Selekman
- Department of Chemical and Biological Engineering, University of Wisconsin, 3637 Engineering Hall, 1415 Engineering Drive, Madison, Wisconsin, 53706
| | | | | | | |
Collapse
|
23
|
Liu N, Li Y, Yang ST. Microfibrous carriers for integrated expansion and neural differentiation of embryonic stem cells in suspension bioreactor. Biochem Eng J 2013. [DOI: 10.1016/j.bej.2013.03.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
24
|
Abstract
INTRODUCTION Mesenchymal stem cells (MSC) and MSC-like cells hold great promise and offer many advantages for developing effective cellular therapeutics. Current trends indicate that the clinical application of MSC will continue to increase markedly. For clinical applications, large numbers of MSC are usually required, ideally in an off-the-shelf format, thus requiring extensive MSC expansion ex vivo and subsequent cryopreservation and banking. AREAS COVERED To exploit the full potential of MSC for cell-based therapies requires overcoming significant cell-manufacturing, banking and regulatory challenges. The current review will focus on the identification of optimal cell source for MSC, the techniques for production scale-up, cryopreservation and banking and the regulatory challenges involved. EXPERT OPINION There has been considerable success manufacturing and cryopreserving MSC at laboratory scale. Surprisingly little attention, however, has been given to translate these technologies to an industrial scale. The development of cost-effective advanced technologies for producing and cryopreserving commercial-scale MSC is important for successful clinical cell therapy.
Collapse
|
25
|
dos Santos FF, Andrade PZ, da Silva CL, Cabral JMS. Bioreactor design for clinical-grade expansion of stem cells. Biotechnol J 2013; 8:644-54. [PMID: 23625834 DOI: 10.1002/biot.201200373] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 03/25/2013] [Accepted: 04/02/2013] [Indexed: 01/24/2023]
Abstract
The many clinical trials currently in progress will likely lead to the widespread use of stem cell-based therapies for an extensive variety of diseases, either in autologous or allogeneic settings. With the current pace of progress, in a few years' time, the field of stem cell-based therapy should be able to respond to the market demand for safe, robust and clinically efficient stem cell-based therapeutics. Due to the limited number of stem cells that can be obtained from a single donor, one of the major challenges on the roadmap for regulatory approval of such medicinal products is the expansion of stem cells using Good Manufacturing Practices (GMP)-compliant culture systems. In fact, manufacturing costs, which include production and quality control procedures, may be the main hurdle for developing cost-effective stem cell therapies. Bioreactors provide a viable alternative to the traditional static culture systems in that bioreactors provide the required scalability, incorporate monitoring and control tools, and possess the operational flexibility to be adapted to the differing requirements imposed by various clinical applications. Bioreactor systems face a number of issues when incorporated into stem cell expansion protocols, both during development at the research level and when bioreactors are used in on-going clinical trials. This review provides an overview of the issues that must be confronted during the development of GMP-compliant bioreactors systems used to support the various clinical applications employing stem cells.
Collapse
Affiliation(s)
- Francisco F dos Santos
- Department of Bioengineering and IBB - Institute for Biotechnology and Bioengineering - Instituto Superior Técnico IST, Technical University of Lisbon, Lisboa, Portugal
| | | | | | | |
Collapse
|
26
|
Wolfe RP, Ahsan T. Shear stress during early embryonic stem cell differentiation promotes hematopoietic and endothelial phenotypes. Biotechnol Bioeng 2013; 110:1231-42. [PMID: 23138937 DOI: 10.1002/bit.24782] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 09/30/2012] [Accepted: 10/30/2012] [Indexed: 12/21/2022]
Abstract
Pluripotent embryonic stem cells (ESCs) are a potential source for cell-based tissue engineering and regenerative medicine applications, but their translation into clinical use will require efficient and robust methods for promoting differentiation. Fluid shear stress, which can be readily incorporated into scalable bioreactors, may be one solution for promoting endothelial and hematopoietic phenotypes from ESCs. Here we applied laminar shear stress to differentiating ESCs using a 2D adherent parallel plate configuration to systematically investigate the effects of several mechanical parameters. Treatment similarly promoted endothelial and hematopoietic differentiation for shear stress magnitudes ranging from 1.5 to 15 dyne/cm(2) and for cells seeded on collagen-, fibronectin- or laminin-coated surfaces. Extension of the treatment duration consistently induced an endothelial response, but application at later stages of differentiation was less effective at promoting hematopoietic phenotypes. Furthermore, inhibition of the FLK1 protein (a VEGF receptor) neutralized the effects of shear stress, implicating the membrane protein as a critical mediator of both endothelial and hematopoietic differentiation by applied shear. Using a systematic approach, studies such as these help elucidate the mechanisms involved in force-mediated stem cell differentiation and inform scalable bioprocesses for cellular therapies.
Collapse
Affiliation(s)
- Russell P Wolfe
- Department of Biomedical Engineering, Tulane University, 500 Lindy Boggs Center, New Orleans, LA 70118, USA
| | | |
Collapse
|
27
|
Mummery CL, Zhang J, Ng ES, Elliott DA, Elefanty AG, Kamp TJ. Differentiation of human embryonic stem cells and induced pluripotent stem cells to cardiomyocytes: a methods overview. Circ Res 2012; 111:344-58. [PMID: 22821908 DOI: 10.1161/circresaha.110.227512] [Citation(s) in RCA: 548] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Since human embryonic stem cells were first differentiated to beating cardiomyocytes a decade ago, interest in their potential applications has increased exponentially. This has been further enhanced over recent years by the discovery of methods to induce pluripotency in somatic cells, including those derived from patients with hereditary cardiac diseases. Human pluripotent stem cells have been among the most challenging cell types to grow stably in culture, but advances in reagent development now mean that most laboratories can expand both embryonic and induced pluripotent stem cells robustly using commercially available products. However, differentiation protocols have lagged behind and in many cases only produce the cell types required with low efficiency. Cardiomyocyte differentiation techniques were also initially inefficient and not readily transferable across cell lines, but there are now a number of more robust protocols available. Here, we review the basic biology underlying the differentiation of pluripotent cells to cardiac lineages and describe current state-of-the-art protocols, as well as ongoing refinements. This should provide a useful entry for laboratories new to this area to start their research. Ultimately, efficient and reliable differentiation methodologies are essential to generate desired cardiac lineages to realize the full promise of human pluripotent stem cells for biomedical research, drug development, and clinical applications.
Collapse
Affiliation(s)
- Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
28
|
Ting S, Lecina M, Reuveny S, Oh S. Differentiation of human embryonic stem cells to cardiomyocytes on microcarrier cultures. ACTA ACUST UNITED AC 2012; Chapter 1:Unit1D.7. [PMID: 22605644 DOI: 10.1002/9780470151808.sc01d07s21] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We have developed an improved cardiomyocyte differentiation protocol where we stabilized embryoid bodies (EB) in serum- and insulin-free medium (bSFS) supplemented with p38 MAP kinase inhibitor (SB203580) by addition of 10 µm laminin-coated positively charged (protamine sulfate derivatized TSKgel Tresyl-5PW) microcarriers. This protocol achieved a maximum 3-fold cell expansion, differentiation efficiency of 20%, and an overall cardiomyocyte yield of 3 × 10⁵ CM/ml in static conditions. In comparison, EB cultures achieved 1.5-fold cell expansion, differentiation efficiency of 15%, and an overall cardiomyocyte yield of 1.1 × 10⁵ CM/ml. The scalability of this platform was demonstrated in suspended spinner cultures, producing a maximum of 2.14 × 10⁵ CM/ml in 50-ml cultures. This yield is two-fold higher than the control static EB-based platform (1.1 × 10⁵ CM/ml), and seven-fold higher than yields reported in literature, 3.1-9 × 10⁴ CM/ml. The robustness of this protocol was tested with HES-3 and H1 cell lines.
Collapse
Affiliation(s)
- Sherwin Ting
- Bioprocessing Technology Institute, A*STAR-Agency for Science, Technology and Research, Singapore
| | | | | | | |
Collapse
|
29
|
|
30
|
Vickers DAL, Kulik M, Hincapie M, Hancock WS, Dalton S, Murthy SK. Lectin-functionalized microchannels for characterizing pluripotent cells and early differentiation. BIOMICROFLUIDICS 2012; 6:24122-2412210. [PMID: 22712033 PMCID: PMC3371070 DOI: 10.1063/1.4719979] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Accepted: 05/06/2012] [Indexed: 05/07/2023]
Abstract
Embryonic stem (ES) cells are capable of proliferating and differentiating to form cells of the three embryonic germ layers, namely, endoderm, mesoderm, and ectoderm. The utilization of human ES cell derivatives requires the ability to direct differentiation to specific lineages in defined, efficient, and scalable systems. Better markers are needed to identify early differentiation. Lectins have been reported as an attractive alternative to the common stem cell markers. They have been used to identify, characterize, and isolate various cell subpopulations on the basis of the presentation of specific carbohydrate groups on the cell surface. This article demonstrates how simple adhesion assays in lectin-coated microfluidic channels can provide key information on the interaction of lectins with ES and definitive endoderm cells and thereby track early differentiation. The microfluidic approach incorporates both binding strength and cell surface receptor density, whereas traditional flow cytometry only incorporates the latter. Both approaches are examined and shown to be complementary with the microfluidic approach providing more biologically relevant information.
Collapse
|
31
|
Serra M, Brito C, Correia C, Alves PM. Process engineering of human pluripotent stem cells for clinical application. Trends Biotechnol 2012; 30:350-9. [PMID: 22541338 DOI: 10.1016/j.tibtech.2012.03.003] [Citation(s) in RCA: 214] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 03/14/2012] [Accepted: 03/14/2012] [Indexed: 12/16/2022]
Abstract
Human pluripotent stem cells (hPSCs), including embryonic and induced pluripotent stem cells, constitute an extremely attractive tool for cell therapy. However, flexible platforms for the large-scale production and storage of hPSCs in tightly controlled conditions are necessary to deliver high-quality cells in relevant quantities to satisfy clinical demands. Here we discuss the main principles for the bioprocessing of hPSCs, highlighting the impact of environmental factors, novel 3D culturing approaches and integrated bioreactor strategies for controlling hPSC culture outcome. Knowledge on hPSC bioprocessing accumulated during recent years provides important insights for the establishment of more robust production platforms and should potentiate the implementation of novel hPSC-based therapies.
Collapse
Affiliation(s)
- Margarida Serra
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | | | | | | |
Collapse
|
32
|
Want AJ, Nienow AW, Hewitt CJ, Coopman K. Large-scale expansion and exploitation of pluripotent stem cells for regenerative medicine purposes: beyond the T flask. Regen Med 2012; 7:71-84. [DOI: 10.2217/rme.11.101] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Human pluripotent stem cells will likely be a significant part of the regenerative medicine-driven healthcare revolution. In order to realize this potential, culture processes must be standardized, scalable and able to produce clinically relevant cell numbers, whilst maintaining critical biological functionality. This review comprises a broad overview of important bioprocess considerations, referencing the development of biopharmaceutical processes in an effort to learn from current best practice in the field. Particular focus is given to the recent efforts to grow human pluripotent stem cells in microcarrier or aggregate suspension culture, which would allow geometric expansion of productive capacity were it to be fully realized. The potential of these approaches is compared with automation of traditional T-flask culture, which may provide a cost-effective platform for low-dose, low-incidence conditions or autologous therapies. This represents the first step in defining the full extent of the challenges facing bioprocess engineers in the exploitation of large-scale human pluripotent stem cell manufacture.
Collapse
Affiliation(s)
- Andrew J Want
- Centre for Biological Engineering, Department of Chemical Engineering, Loughborough University, Leicestershire, LE11 3TU, UK
| | - Alvin W Nienow
- Centre for Biological Engineering, Department of Chemical Engineering, Loughborough University, Leicestershire, LE11 3TU, UK
- Centre for Bioprocess Engineering, Department of Chemical Engineering, University of Birmingham, B15 2TT, UK
| | - Christopher J Hewitt
- Centre for Biological Engineering, Department of Chemical Engineering, Loughborough University, Leicestershire, LE11 3TU, UK
| | | |
Collapse
|
33
|
Azarin SM, Lian X, Larson EA, Popelka HM, de Pablo JJ, Palecek SP. Modulation of Wnt/β-catenin signaling in human embryonic stem cells using a 3-D microwell array. Biomaterials 2011; 33:2041-9. [PMID: 22177620 DOI: 10.1016/j.biomaterials.2011.11.070] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 11/25/2011] [Indexed: 12/27/2022]
Abstract
Intercellular interactions in the cell microenvironment play a critical role in determining cell fate, but the effects of these interactions on pathways governing human embryonic stem cell (hESC) behavior have not been fully elucidated. We and others have previously reported that 3-D culture of hESCs affects cell fates, including self-renewal and differentiation to a variety of lineages. Here we have used a microwell culture system that produces 3-D colonies of uniform size and shape to provide insight into the effect of modulating cell-cell contact on canonical Wnt/β-catenin signaling in hESCs. Canonical Wnt signaling has been implicated in both self-renewal and differentiation of hESCs, and competition for β-catenin between the Wnt pathway and cadherin-mediated cell-cell interactions impacts various developmental processes, including the epithelial-mesenchymal transition. Our results showed that hESCs cultured in 3-D microwells exhibited higher E-cadherin expression than cells on 2-D substrates. The increase in E-cadherin expression in microwells was accompanied by a downregulation of Wnt signaling, as evidenced by the lack of nuclear β-catenin and downregulation of Wnt target genes. Despite this reduction in Wnt signaling in microwell cultures, embryoid bodies (EBs) formed from hESCs cultured in microwells exhibited higher levels of Wnt signaling than EBs from hESCs cultured on 2-D substrates. Furthermore, the Wnt-positive cells within EBs showed upregulation of genes associated with cardiogenesis. These results demonstrate that modulation of intercellular interactions impacts Wnt/β-catenin signaling in hESCs.
Collapse
Affiliation(s)
- Samira M Azarin
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI 53706, USA
| | | | | | | | | | | |
Collapse
|
34
|
Ungrin MD, Clarke G, Yin T, Niebrugge S, Nostro MC, Sarangi F, Wood G, Keller G, Zandstra PW. Rational bioprocess design for human pluripotent stem cell expansion and endoderm differentiation based on cellular dynamics. Biotechnol Bioeng 2011; 109:853-66. [PMID: 22139975 DOI: 10.1002/bit.24375] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 11/01/2011] [Accepted: 11/07/2011] [Indexed: 12/23/2022]
Abstract
We present a predictive bioprocess design strategy employing cell- and molecular-level analysis of rate-limiting steps in human pluripotent stem cell (hPSC) expansion and differentiation, and apply it to produce definitive endoderm (DE) progenitors using a scalable directed-differentiation technology. We define a bioprocess optimization parameter (L; targeted cell Loss) and, with quantitative cell division tracking and fate monitoring, identify and overcome key suspension bioprocess bottlenecks. Adapting process operating conditions to pivotal parameters (single cell survival and growth rate) in a cell-line-specific manner enabled adherent-equivalent expansion of hPSCs in feeder- and matrix-free defined-medium suspension culture. Predominantly instructive differentiation mechanisms were found to underlie a subsequent 18-fold expansion, during directed differentiation, to high-purity DE competent for further commitment along pancreatic and hepatic lineages. This study demonstrates that iPSC expansion and differentiation conditions can be prospectively specified to guide the enhanced production of target cells in a scale-free directed differentiation system.
Collapse
Affiliation(s)
- Mark D Ungrin
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Healy L, Young L, Stacey GN. Stem cell banks: preserving cell lines, maintaining genetic integrity, and advancing research. Methods Mol Biol 2011; 767:15-27. [PMID: 21822864 DOI: 10.1007/978-1-61779-201-4_2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The ability to cryopreserve and successfully recover cell lines has been critical to the conservation of all cell lines, especially the preservation of pristine early-stage cultures and the preparation of well-characterized cell banks. Indeed, the systematic storage and establishment of cryopreserved banks of cells for the stem cell research community is fundamental to the promotion of standardisation in stem cell research and their use in clinical applications. In spite of the significant potential for the use of stem cells in research and therapy, they are challenging to maintain and have been shown to be unstable after prolonged culture that often results in permanent alterations in their genetic make-up, which ultimately alters the phenotype of the culture. This chapter will review the principles of cell bank production, techniques for the scale-up of human pluripotent stem cells, quality control, and characterisation methods for banked cell lines.
Collapse
Affiliation(s)
- Lyn Healy
- UK Stem Cell Bank, Cell Biology and Imaging, National Institute for Biological Standards and Control, South Mimms, Hertfordshire, UK
| | | | | |
Collapse
|
36
|
Serra M, Correia C, Malpique R, Brito C, Jensen J, Bjorquist P, Carrondo MJT, Alves PM. Microencapsulation technology: a powerful tool for integrating expansion and cryopreservation of human embryonic stem cells. PLoS One 2011; 6:e23212. [PMID: 21850261 PMCID: PMC3151290 DOI: 10.1371/journal.pone.0023212] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 07/14/2011] [Indexed: 12/30/2022] Open
Abstract
The successful implementation of human embryonic stem cells (hESCs)-based technologies requires the production of relevant numbers of well-characterized cells and their efficient long-term storage. In this study, cells were microencapsulated in alginate to develop an integrated bioprocess for expansion and cryopreservation of pluripotent hESCs. Different three-dimensional (3D) culture strategies were evaluated and compared, specifically, microencapsulation of hESCs as: i) single cells, ii) aggregates and iii) immobilized on microcarriers. In order to establish a scalable bioprocess, hESC-microcapsules were cultured in stirred tank bioreactors. The combination of microencapsulation and microcarrier technology resulted in a highly efficient protocol for the production and storage of pluripotent hESCs. This strategy ensured high expansion ratios (an approximately twenty-fold increase in cell concentration) and high cell recovery yields (>70%) after cryopreservation. When compared with non-encapsulated cells, cell survival post-thawing demonstrated a three-fold improvement without compromising hESC characteristics. Microencapsulation also improved the culture of hESC aggregates by protecting cells from hydrodynamic shear stress, controlling aggregate size and maintaining cell pluripotency for two weeks. This work establishes that microencapsulation technology may prove a powerful tool for integrating the expansion and cryopreservation of pluripotent hESCs. The 3D culture strategy developed herein represents a significant breakthrough towards the implementation of hESCs in clinical and industrial applications.
Collapse
Affiliation(s)
- Margarida Serra
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
- Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Cláudia Correia
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
- Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Rita Malpique
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
- Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Catarina Brito
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
- Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | | | | | - Manuel J. T. Carrondo
- Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Monte da Caparica, Portugal
| | - Paula M. Alves
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
- Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- * E-mail:
| |
Collapse
|
37
|
Serra M, Correia C, Malpique R, Brito C, Jensen J, Bjorquist P, Carrondo MJT, Alves PM. Microencapsulation technology: a powerful tool for integrating expansion and cryopreservation of human embryonic stem cells. PLoS One 2011. [PMID: 21850261 DOI: 10.1371/journal.pone.002321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The successful implementation of human embryonic stem cells (hESCs)-based technologies requires the production of relevant numbers of well-characterized cells and their efficient long-term storage. In this study, cells were microencapsulated in alginate to develop an integrated bioprocess for expansion and cryopreservation of pluripotent hESCs. Different three-dimensional (3D) culture strategies were evaluated and compared, specifically, microencapsulation of hESCs as: i) single cells, ii) aggregates and iii) immobilized on microcarriers. In order to establish a scalable bioprocess, hESC-microcapsules were cultured in stirred tank bioreactors.The combination of microencapsulation and microcarrier technology resulted in a highly efficient protocol for the production and storage of pluripotent hESCs. This strategy ensured high expansion ratios (an approximately twenty-fold increase in cell concentration) and high cell recovery yields (>70%) after cryopreservation. When compared with non-encapsulated cells, cell survival post-thawing demonstrated a three-fold improvement without compromising hESC characteristics.Microencapsulation also improved the culture of hESC aggregates by protecting cells from hydrodynamic shear stress, controlling aggregate size and maintaining cell pluripotency for two weeks.This work establishes that microencapsulation technology may prove a powerful tool for integrating the expansion and cryopreservation of pluripotent hESCs. The 3D culture strategy developed herein represents a significant breakthrough towards the implementation of hESCs in clinical and industrial applications.
Collapse
Affiliation(s)
- Margarida Serra
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Rodrigues CAV, Fernandes TG, Diogo MM, da Silva CL, Cabral JMS. Stem cell cultivation in bioreactors. Biotechnol Adv 2011; 29:815-29. [PMID: 21726624 DOI: 10.1016/j.biotechadv.2011.06.009] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 06/11/2011] [Accepted: 06/12/2011] [Indexed: 12/22/2022]
Abstract
Cell-based therapies have generated great interest in the scientific and medical communities, and stem cells in particular are very appealing for regenerative medicine, drug screening and other biomedical applications. These unspecialized cells have unlimited self-renewal capacity and the remarkable ability to produce mature cells with specialized functions, such as blood cells, nerve cells or cardiac muscle. However, the actual number of cells that can be obtained from available donors is very low. One possible solution for the generation of relevant numbers of cells for several applications is to scale-up the culture of these cells in vitro. This review describes recent developments in the cultivation of stem cells in bioreactors, particularly considerations regarding critical culture parameters, possible bioreactor configurations, and integration of novel technologies in the bioprocess development stage. We expect that this review will provide updated and detailed information focusing on the systematic production of stem cell products in compliance with regulatory guidelines, while using robust and cost-effective approaches.
Collapse
Affiliation(s)
- Carlos A V Rodrigues
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Centre for Biological and Chemical Engineering, Instituto Superior Técnico, Technical University of Lisbon, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | | | | | | | | |
Collapse
|
39
|
Chung BG, Choo J. Microfluidic gradient platforms for controlling cellular behavior. Electrophoresis 2010; 31:3014-27. [PMID: 20734372 DOI: 10.1002/elps.201000137] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Concentration gradients play an important role in controlling biological and pathological processes, such as metastasis, embryogenesis, axon guidance, and wound healing. Microfluidic devices fabricated by photo- and soft lithography techniques can manipulate the fluidic flow and diffusion profile to create biomolecular gradients in a temporal and spatial manner. Furthermore, microfluidic devices enable the control of cell-extracellular microenvironment interactions, including cell-cell, cell-matrix, and cell-soluble factor interaction. In this paper, we review the development of microfluidic-based gradient devices and highlight their biological applications.
Collapse
Affiliation(s)
- Bong Geun Chung
- Department of Bionano Engineering, Hanyang University, Ansan, Korea.
| | | |
Collapse
|
40
|
Lecina M, Ting S, Choo A, Reuveny S, Oh S. Scalable platform for human embryonic stem cell differentiation to cardiomyocytes in suspended microcarrier cultures. Tissue Eng Part C Methods 2010; 16:1609-19. [PMID: 20590381 DOI: 10.1089/ten.tec.2010.0104] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
A scalable platform for human embryonic stem cell (hESC)-derived cardiomyocyte (CM) production can provide a readily available source of CMs for cell therapy, drug screening, and cardiotoxicity tests. We have designed and optimized a scalable platform using microcarrier cultures in serum-free media supplemented with SB203580 mitogen-activated protein kinase-inhibitor. Different microcarriers (DE-53, Cytodex-1 and 3, FACT, and TOSOH-10) were used to investigate the effects of type, size, shape, and microcarrier concentrations on the differentiation efficiency. hESCs propagated on TOSOH-10 (protamine derivatized 10-μm beads) at the concentration of 0.125 mg/mL produced 80% beating aggregates, threefold cell expansion, and 20% of CMs (determined by fluorescence-activated cell sorting for myosin heavy chain and α-actinin expression). The ratio of CM/hESC seeded in this system was 0.62 compared to 0.22 in the embryoid body control cultures. The platform robustness has been tested with HES-3 and H1 cell lines, and its scalability was demonstrated in suspended spinner cultures. However, spinner culture yields dropped to 0.33 CM/hESC probably due to shear stress causing some cell death. Cells dissociated from differentiated aggregates showed positive staining for cardio-specific markers such as α-actinin, myosin heavy and light chain, troponin I, desmin, and emilin-2. Finally, CM functionality was also shown by QT-prolongation (QTempo) assay with/without Astemizole. This study represents a new scalable bioprocessing system for CM production using reagents that can comply with Good Manufacturing Practice.
Collapse
Affiliation(s)
- Marti Lecina
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.
| | | | | | | | | |
Collapse
|