1
|
Ding L, Oh S, Shrestha J, Lam A, Wang Y, Radfar P, Warkiani ME. Scaling up stem cell production: harnessing the potential of microfluidic devices. Biotechnol Adv 2023; 69:108271. [PMID: 37844769 DOI: 10.1016/j.biotechadv.2023.108271] [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: 04/29/2023] [Revised: 10/08/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
Stem cells are specialised cells characterised by their unique ability to both self-renew and transform into a wide array of specialised cell types. The widespread interest in stem cells for regenerative medicine and cultivated meat has led to a significant demand for these cells in both research and practical applications. Despite the growing need for stem cell manufacturing, the industry faces significant obstacles, including high costs for equipment and maintenance, complicated operation, and low product quality and yield. Microfluidic technology presents a promising solution to the abovementioned challenges. As an innovative approach for manipulating liquids and cells within microchannels, microfluidics offers a plethora of advantages at an industrial scale. These benefits encompass low setup costs, ease of operation and multiplexing, minimal energy consumption, and the added advantage of being labour-free. This review presents a thorough examination of the prominent microfluidic technologies employed in stem cell research and explores their promising applications in the burgeoning stem cell industry. It thoroughly examines how microfluidics can enhance cell harvesting from tissue samples, facilitate mixing and cryopreservation, streamline microcarrier production, and efficiently conduct cell separation, purification, washing, and final cell formulation post-culture.
Collapse
Affiliation(s)
- Lin Ding
- Smart MCs Pty Ltd, Ultimo, Sydney, 2007, Australia.
| | - Steve Oh
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Jesus Shrestha
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Alan Lam
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Yaqing Wang
- School of Biomedical Engineering, University of Science and Technology of China, Hefei 230026, China; Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| | - Payar Radfar
- Smart MCs Pty Ltd, Ultimo, Sydney, 2007, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia..
| |
Collapse
|
2
|
Zhang T, Cain AK, Semenec L, Liu L, Hosokawa Y, Inglis DW, Yalikun Y, Li M. Microfluidic Separation and Enrichment of Escherichia coli by Size Using Viscoelastic Flows. Anal Chem 2023; 95:2561-2569. [PMID: 36656064 DOI: 10.1021/acs.analchem.2c05084] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Here, we achieve the separation and enrichment of Escherichia coli clusters from its singlets in a viscoelastic microfluidic device. E. coli, an important prokaryotic model organism and a widely used microbial factory, can aggregate in clusters, leading to biofilm development that can be detrimental to human health and industrial processes. The ability to obtain high-purity populations of E. coli clusters is of significance for biological, biomedical, and industrial applications. In this study, polystyrene particles of two different sizes, 1 and 4.8 μm, are used to mimic E. coli singlets and clusters, respectively. Experimental results show that particles migrate toward the channel center in a size-dependent manner, due to the combined effects of inertial and elastic forces; 4.8 and 1 μm particles are found to have lateral equilibrium positions closer to the channel centerline and sidewalls, respectively. The size-dependent separation performance of the microdevice is demonstrated to be affected by three main factors: channel length, the ratio of sheath to sample flow rate, and poly(ethylene oxide) (PEO) concentration. Further, the separation of E. coli singlets and clusters is achieved at the outlets, and the separation efficiency is evaluated in terms of purity and enrichment factor.
Collapse
Affiliation(s)
- Tianlong Zhang
- School of Engineering, Macquarie University, Sydney, NSW 2109, Australia.,Division of Materials Science, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
| | - Amy K Cain
- ARC Centre of Excellence in Synthetic Biology, School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Lucie Semenec
- ARC Centre of Excellence in Synthetic Biology, School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Ling Liu
- School of Engineering, Macquarie University, Sydney, NSW 2109, Australia
| | - Yoichiroh Hosokawa
- Division of Materials Science, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
| | - David W Inglis
- School of Engineering, Macquarie University, Sydney, NSW 2109, Australia
| | - Yaxiaer Yalikun
- Division of Materials Science, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
| | - Ming Li
- School of Engineering, Macquarie University, Sydney, NSW 2109, Australia.,Biomolecular Discovery Research Centre, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
3
|
Sart S, Liu C, Zeng EZ, Xu C, Li Y. Downstream bioprocessing of human pluripotent stem cell-derived therapeutics. Eng Life Sci 2022; 22:667-680. [PMID: 36348655 PMCID: PMC9635003 DOI: 10.1002/elsc.202100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/08/2021] [Accepted: 08/16/2021] [Indexed: 11/30/2022] Open
Abstract
With the advancement in lineage-specific differentiation from human pluripotent stem cells (hPSCs), downstream cell separation has now become a critical step to produce hPSC-derived products. Since differentiation procedures usually result in a heterogeneous cell population, cell separation needs to be performed either to enrich the desired cell population or remove the undesired cell population. This article summarizes recent advances in separation processes for hPSC-derived cells, including the standard separation technologies, such as magnetic-activated cell sorting, as well as the novel separation strategies, such as those based on adhesion strength and metabolic flux. Specifically, the downstream bioprocessing flow and the identification of surface markers for various cell lineages are discussed. While challenges remain for large-scale downstream bioprocessing of hPSC-derived cells, the rational quality-by-design approach should be implemented to enhance the understanding of the relationship between process and the product and to ensure the safety of the produced cells.
Collapse
Affiliation(s)
- Sebastien Sart
- Laboratory of Physical Microfluidics and BioengineeringDepartment of Genome and GeneticsInstitut PasteurParisFrance
| | - Chang Liu
- Department of Chemical and Biomedical EngineeringFAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFLUSA
| | - Eric Z. Zeng
- Department of Chemical and Biomedical EngineeringFAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFLUSA
| | - Chunhui Xu
- Department of PediatricsEmory University School of Medicine and Children's Healthcare of AtlantaAtlantaGAUSA
| | - Yan Li
- Department of Chemical and Biomedical EngineeringFAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFLUSA
| |
Collapse
|
4
|
Fukunaga I, Oe Y, Chen C, Danzaki K, Ohta S, Koike A, Ikeda K, Kamiya K. Activin/Nodal/TGF-β Pathway Inhibitor Accelerates BMP4-Induced Cochlear Gap Junction Formation During in vitro Differentiation of Embryonic Stem Cells. Front Cell Dev Biol 2021; 9:602197. [PMID: 33968919 PMCID: PMC8097046 DOI: 10.3389/fcell.2021.602197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 03/30/2021] [Indexed: 11/13/2022] Open
Abstract
Mutations in gap junction beta-2 (GJB2), the gene that encodes connexin 26 (CX26), are the most frequent cause of hereditary deafness worldwide. We recently developed an in vitro model of GJB2-related deafness (induced CX26 gap junction-forming cells; iCX26GJCs) from mouse induced pluripotent stem cells (iPSCs) by using Bone morphogenetic protein 4 (BMP4) signaling-based floating cultures (serum-free culture of embryoid body-like aggregates with quick aggregation cultures; hereafter, SFEBq cultures) and adherent cultures. However, to use these cells as a disease model platform for high-throughput drug screening or regenerative therapy, cell yields must be substantially increased. In addition to BMP4, other factors may also induce CX26 gap junction formation. In the SFEBq cultures, the combination of BMP4 and the Activin/Nodal/TGF-β pathway inhibitor SB431542 (SB) resulted in greater production of isolatable CX26-expressing cell mass (CX26+ vesicles) and higher Gjb2 mRNA levels than BMP4 treatment alone, suggesting that SB may promote BMP4-mediated production of CX26+ vesicles in a dose-dependent manner, thereby increasing the yield of highly purified iCX26GJCs. This is the first study to demonstrate that SB accelerates BMP4-induced iCX26GJC differentiation during stem cell floating culture. By controlling the concentration of SB supplementation in combination with CX26+ vesicle purification, large-scale production of highly purified iCX26GJCs suitable for high-throughput drug screening or regenerative therapy for GJB2-related deafness may be possible.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kazusaku Kamiya
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
5
|
Faye PA, Poumeaud F, Chazelas P, Duchesne M, Rassat M, Miressi F, Lia AS, Sturtz F, Robert PY, Favreau F, Benayoun Y. Focus on cell therapy to treat corneal endothelial diseases. Exp Eye Res 2021; 204:108462. [PMID: 33493477 DOI: 10.1016/j.exer.2021.108462] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023]
Abstract
The cornea is a multi-layered structure which allows fine refraction and provides both resistance to external insults and adequate transparency. The corneal endothelium ensures stromal hydration, failure of which, such as in Fuchs endothelial corneal dystrophy, after trauma or in aging, may lead to loss of corneal transparency and induce blindness. Currently, no efficient therapeutic alternatives exist except for corneal grafting. Thus corneal tissue engineering represents a valuable alternative approach, which may overcome cornea donor shortage. Several studies describe protocols to isolate, differentiate, and cultivate corneal endothelial cells (CEnCs) in vitro. Two main in vitro strategies can be described: expansion of eye-native cell populations, such as CEnCs, or the production and expansion of CEnCs from non-eye native cell populations, such as induced Pluripotent Stem Cells (iPSCs). The challenge with these cells is to obtain a monolayer of CEnCs on a biocompatible carrier, with a specific morphology (flat hexagonal cells), and with specific functions such as programmed cell cycle arrest. Another issue for this cell culture methodology is to define the adapted protocol (media, trophic factors, timeframe) that can mimic physiological development. Additionally, contamination by other cell types still represents a huge problem. Thus, purification methods, such as Fluorescence Activated Cell Sorting (FACS), Magnetic Ativated Cell Sorting (MACS) or Sedimentation Field Flow Fractionation (SdFFF) are useful. Animal models are also crucial to provide a translational approach for these therapies, integrating macro- and microenvironment influences, systemic hormonal or immune responses, and exogenous interactions. Non-eye native cell graft protocols are constantly improving both in efficacy and safety, with the aim of being the most suitable candidate for corneal therapies in future routine practice. The aim of this work is to review these different aspects with a special focus on issues facing CEnC culture in vitro, and to highlight animal graft models adapted to screen the efficacy of these different protocols.
Collapse
Affiliation(s)
- Pierre Antoine Faye
- CHU de Limoges, Service de Biochimie et Génétique Moléculaire, F-87000, Limoges, France; Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France.
| | - François Poumeaud
- Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France
| | - Pauline Chazelas
- CHU de Limoges, Service de Biochimie et Génétique Moléculaire, F-87000, Limoges, France; Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France
| | - Mathilde Duchesne
- Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France; CHU de Limoges, Laboratoire de Neurologie, F-87000, Limoges, France; CHU de Limoges, Service d'Anatomie Pathologique, F-87000, Limoges, France
| | - Marion Rassat
- Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France
| | - Federica Miressi
- Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France
| | - Anne Sophie Lia
- CHU de Limoges, Service de Biochimie et Génétique Moléculaire, F-87000, Limoges, France; Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France; CHU Limoges, UF de Bioinformatique, F-87000, Limoges France
| | - Franck Sturtz
- CHU de Limoges, Service de Biochimie et Génétique Moléculaire, F-87000, Limoges, France; Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France
| | | | - Frédéric Favreau
- CHU de Limoges, Service de Biochimie et Génétique Moléculaire, F-87000, Limoges, France; Université de Limoges, Faculté de Médecine, Maintenance Myélinique et Neuropathies Périphériques, EA6309, F-87000, Limoges, France
| | - Yohan Benayoun
- Chénieux Ophtalmologie, Polyclinique de Limoges ELSAN, F-87000, Limoges, France
| |
Collapse
|
6
|
Gabr H, El Ghamrawy MK, Almaeen AH, Abdelhafiz AS, Hassan AOS, El Sissy MH. CRISPR-mediated gene modification of hematopoietic stem cells with beta-thalassemia IVS-1-110 mutation. Stem Cell Res Ther 2020; 11:390. [PMID: 32912325 PMCID: PMC7488347 DOI: 10.1186/s13287-020-01876-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/19/2020] [Accepted: 08/05/2020] [Indexed: 11/24/2022] Open
Abstract
Background β-Thalassemias represent a group of genetic disorders caused by human hemoglobin beta (HBB) gene mutations. The radical curative approach is to correct the mutations causing the disease. CRISPR-CAS9 is a novel gene-editing technology that can be used auspiciously for the treatment of these disorders. The study aimed to investigate the utility of CRISPR-CAS9 for gene modification of hematopoietic stem cells in β-thalassemia with IVS-1-110 mutation. Methods and results We successfully isolated CD34+ cells from peripheral blood of β-thalassemia patients with IVS-1-110 mutation. The cells were transfected with Cas9 endonuclease together with guide RNA to create double-strand breaks and knock out the mutation. The mutation-corrected CD34+ cells were subjected to erythroid differentiation by culturing in complete media containing erythropoietin. Conclusion CRISPR/Cas-9 is an effective tool for gene therapy that will broaden the spectrum of therapy and potentially improve the outcomes of β-thalassemia.
Collapse
Affiliation(s)
- Hala Gabr
- Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | | | | | - Aya Osama Saad Hassan
- Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Maha Hamdi El Sissy
- Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt.
| |
Collapse
|
7
|
Bacon K, Lavoie A, Rao BM, Daniele M, Menegatti S. Past, Present, and Future of Affinity-based Cell Separation Technologies. Acta Biomater 2020; 112:29-51. [PMID: 32442784 PMCID: PMC10364325 DOI: 10.1016/j.actbio.2020.05.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023]
Abstract
Progress in cell purification technology is critical to increase the availability of viable cells for therapeutic, diagnostic, and research applications. A variety of techniques are now available for cell separation, ranging from non-affinity methods such as density gradient centrifugation, dielectrophoresis, and filtration, to affinity methods such as chromatography, two-phase partitioning, and magnetic-/fluorescence-assisted cell sorting. For clinical and analytical procedures that require highly purified cells, the choice of cell purification method is crucial, since every method offers a different balance between yield, purity, and bioactivity of the cell product. For most applications, the requisite purity is only achievable through affinity methods, owing to the high target specificity that they grant. In this review, we discuss past and current methods for developing cell-targeting affinity ligands and their application in cell purification, along with the benefits and challenges associated with different purification formats. We further present new technologies, like stimuli-responsive ligands and parallelized microfluidic devices, towards improving the viability and throughput of cell products for tissue engineering and regenerative medicine. Our comparative analysis provides guidance in the multifarious landscape of cell separation techniques and highlights new technologies that are poised to play a key role in the future of cell purification in clinical settings and the biotech industry. STATEMENT OF SIGNIFICANCE: Technologies for cell purification have served science, medicine, and industrial biotechnology and biomanufacturing for decades. This review presents a comprehensive survey of this field by highlighting the scope and relevance of all known methods for cell isolation, old and new alike. The first section covers the main classes of target cells and compares traditional non-affinity and affinity-based purification techniques, focusing on established ligands and chromatographic formats. The second section presents an excursus of affinity-based pseudo-chromatographic and non-chromatographic technologies, especially focusing on magnetic-activated cell sorting (MACS) and fluorescence-activated cell sorting (FACS). Finally, the third section presents an overview of new technologies and emerging trends, highlighting how the progress in chemical, material, and microfluidic sciences has opened new exciting avenues towards high-throughput and high-purity cell isolation processes. This review is designed to guide scientists and engineers in their choice of suitable cell purification techniques for research or bioprocessing needs.
Collapse
Affiliation(s)
- Kaitlyn Bacon
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695-7905, USA
| | - Ashton Lavoie
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695-7905, USA
| | - Balaji M Rao
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695-7905, USA; Biomanufacturing Training and Education Center (BTEC), North Carolina State University, Raleigh, NC 27695-7928, USA
| | - Michael Daniele
- Joint Department of Biomedical Engineering, North Carolina State University - University of North Carolina Chapel Hill, North Carolina, United States
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695-7905, USA; Biomanufacturing Training and Education Center (BTEC), North Carolina State University, Raleigh, NC 27695-7928, USA.
| |
Collapse
|
8
|
Talaei-Khozani T, Aleahmad F, Bazrafshan A, Aliabadi E, Vojdani Z. Lectin Profile Variation in Mesenchymal Stem Cells Derived from Different Sources. Cells Tissues Organs 2020; 208:101-112. [PMID: 32464631 DOI: 10.1159/000505238] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 12/05/2019] [Indexed: 11/19/2022] Open
Abstract
Human mesenchymal stem cells (MSCs), a promising source of stem cells for regenerative medicine, have different morphological and functional characteristics. Carbohydrate moieties on the cell surface play an important role, including cell-cell interaction and cell recognition. The objective of this study was to determine possible differences in glycoconjugate distribution patterns of MSCs derived from various sources. MSCs were isolated from adipose tissue, bone marrow, Wharton's jelly, and cord blood. Then, they were stained with FITC-conjugated wheat germ agglutinin (WGA), peanut agglutinin (PNA), concanavalin A (ConA), Ulex europaeus (UEA), Dolichos biflorus (DBA), and Atto-488 conjugated Phytolacca americana (PWM) lectins. The intensity of the reactions was scored using ImageJ software. Flow cytometry was performed to detect the expression of the endothelial marker CD144. The obtained data were analyzed by ANOVA and LSD. Cord blood-derived MSCs showed the most significant staining intensities with all lectins. All MSCs were also moderately stained with PNA. Bone marrow-derived MSCs failed to react with UEA, DBA, and ConA. Wharton's jelly-derived MSCs could also not be stained with ConA. Cord blood-derived MSCs contained 2 subpopulations: osteoclast- and fibroblast-like cells. Both lectin staining intensity and distribution pattern were different in these 2 cell types; therefore, the central part of osteoclast-like cells stained more intensive with PNA and PWM, while that part in fibroblast-like cells stained more intensive with ConA. None of them expressed CD144. The glycoconjugate content of MSCs derived from various sources is different.
Collapse
Affiliation(s)
- Tahereh Talaei-Khozani
- Laboratory for Stem Cell Research, Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Aleahmad
- Laboratory for Stem Cell Research, Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ameneh Bazrafshan
- Department of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran, .,Maternal-Fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran,
| | - Elham Aliabadi
- Laboratory for Stem Cell Research, Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Vojdani
- Laboratory for Stem Cell Research, Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
9
|
Pereira Chilima TD, Moncaubeig F, Farid SS. Estimating capital investment and facility footprint in cell therapy facilities. Biochem Eng J 2020. [DOI: 10.1016/j.bej.2019.107439] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
10
|
Matos MJB, Pina AS, Roque ACA. Rational design of affinity ligands for bioseparation. J Chromatogr A 2020; 1619:460871. [PMID: 32044126 DOI: 10.1016/j.chroma.2020.460871] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/05/2020] [Accepted: 01/08/2020] [Indexed: 11/25/2022]
Abstract
Affinity adsorbents have been the cornerstone in protein purification. The selective nature of the molecular recognition interactions established between an affinity ligands and its target provide the basis for efficient capture and isolation of proteins. The plethora of affinity adsorbents available in the market reflects the importance of affinity chromatography in the bioseparation industry. Ligand discovery relies on the implementation of rational design techniques, which provides the foundation for the engineering of novel affinity ligands. The main goal for the design of affinity ligands is to discover or improve functionality, such as increased stability or selectivity. However, the methodologies must adapt to the current needs, namely to the number and diversity of biologicals being developed, and the availability of new tools for big data analysis and artificial intelligence. In this review, we offer an overview on the development of affinity ligands for bioseparation, including the evolution of rational design techniques, dating back to the years of early discovery up to the current and future trends in the field.
Collapse
Affiliation(s)
- Manuel J B Matos
- UCIBIO, Chemistry Department, School of Sciences and Technology, NOVA University of Lisbon, 2829-516 Caparica, Portugal
| | - Ana S Pina
- UCIBIO, Chemistry Department, School of Sciences and Technology, NOVA University of Lisbon, 2829-516 Caparica, Portugal
| | - A C A Roque
- UCIBIO, Chemistry Department, School of Sciences and Technology, NOVA University of Lisbon, 2829-516 Caparica, Portugal.
| |
Collapse
|
11
|
Liu N, Petchakup C, Tay HM, Li KHH, Hou HW. Spiral Inertial Microfluidics for Cell Separation and Biomedical Applications. Bioanalysis 2019. [DOI: 10.1007/978-981-13-6229-3_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
12
|
Human Pluripotent Stem Cells: Applications and Challenges for Regenerative Medicine and Disease Modeling. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2019; 171:189-224. [PMID: 31740987 DOI: 10.1007/10_2019_117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years, human pluripotent stem (hPS) cells have started to emerge as a potential tool with application in fields such as regenerative medicine, disease modeling, and drug screening. In particular, the ability to differentiate human-induced pluripotent stem (hiPS) cells into different cell types and to mimic structures and functions of a specific target organ, resourcing to organoid technology, has introduced novel model systems for disease recapitulation while offering a powerful tool to provide a faster and reproducible approach in the process of drug discovery. All these technologies are expected to improve the overall quality of life of the humankind. Here, we highlight the main applications of hiPS cells and the main challenges associated with the translation of hPS cell derivatives into clinical settings and other biomedical applications, such as the costs of the process and the ability to mimic the complexity of the in vivo systems. Moreover, we focus on the bioprocessing approaches that can be applied towards the production of high numbers of cells as well as their efficient differentiation into the final product and further purification.
Collapse
|
13
|
Matsuzaki T, Matsumoto S, Kasai T, Yoshizawa E, Okamoto S, Yoshikawa HY, Taniguchi H, Takebe T. Defining Lineage-Specific Membrane Fluidity Signatures that Regulate Adhesion Kinetics. Stem Cell Reports 2018; 11:852-860. [PMID: 30197117 PMCID: PMC6178887 DOI: 10.1016/j.stemcr.2018.08.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/10/2018] [Accepted: 08/11/2018] [Indexed: 12/13/2022] Open
Abstract
Cellular membrane fluidity is a critical modulator of cell adhesion and migration, prompting us to define the systematic landscape of lineage-specific cellular fluidity throughout differentiation. Here, we have unveiled membrane fluidity landscapes in various lineages ranging from human pluripotency to differentiated progeny: (1) membrane rigidification precedes the exit from pluripotency, (2) membrane composition modulates activin signaling transmission, and (3) signatures are relatively germ layer specific presumably due to unique lipid compositions. By modulating variable lineage-specific fluidity, we developed a label-free “adhesion sorting (AdSort)” method with simple cultural manipulation, effectively eliminating pluripotent stem cells and purifying target population as a result of the over 1,150 of screened conditions combining compounds and matrices. These results underscore the important role of tunable membrane fluidity in influencing stem cell maintenance and differentiation that can be translated into lineage-specific cell purification strategy. Membrane rigidification precedes the exit from pluripotency Germ layer-specific membrane fluidity signature exists Identification of polyphenols as a membrane fluidity modulator Fluidity-based adhesion sorting purify differentiated progeny from pluripotency
Collapse
Affiliation(s)
- Takahisa Matsuzaki
- Institute of Research, Tokyo Medical and Dental University (TMDU), 15-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Shinya Matsumoto
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Toshiharu Kasai
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Emi Yoshizawa
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Satoshi Okamoto
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Hiroshi Y Yoshikawa
- Department of Chemistry, Saitama University, Shimo-okubo 255, Sakura-ku, Saitama 338-8570, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan; Advanced Medical Research Center, Yokohama City University, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan
| | - Takanori Takebe
- Institute of Research, Tokyo Medical and Dental University (TMDU), 15-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan; Advanced Medical Research Center, Yokohama City University, Kanazawa-ku 3-9, Yokohama, Kanagawa 236-0004, Japan; Division of Gastroenterology, Hepatology & Nutrition, Developmental Biology, Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA.
| |
Collapse
|
14
|
Bongiorno T, Gura J, Talwar P, Chambers D, Young KM, Arafat D, Wang G, Jackson-Holmes EL, Qiu P, McDevitt TC, Sulchek T. Biophysical subsets of embryonic stem cells display distinct phenotypic and morphological signatures. PLoS One 2018. [PMID: 29518080 PMCID: PMC5843178 DOI: 10.1371/journal.pone.0192631] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The highly proliferative and pluripotent characteristics of embryonic stem cells engender great promise for tissue engineering and regenerative medicine, but the rapid identification and isolation of target cell phenotypes remains challenging. Therefore, the objectives of this study were to characterize cell mechanics as a function of differentiation and to employ differences in cell stiffness to select population subsets with distinct mechanical, morphological, and biological properties. Biomechanical analysis with atomic force microscopy revealed that embryonic stem cells stiffened within one day of differentiation induced by leukemia inhibitory factor removal, with a lagging but pronounced change from spherical to spindle-shaped cell morphology. A microfluidic device was then employed to sort a differentially labeled mixture of pluripotent and differentiating cells based on stiffness, resulting in pluripotent cell enrichment in the soft device outlet. Furthermore, sorting an unlabeled population of partially differentiated cells produced a subset of “soft” cells that was enriched for the pluripotent phenotype, as assessed by post-sort characterization of cell mechanics, morphology, and gene expression. The results of this study indicate that intrinsic cell mechanical properties might serve as a basis for efficient, high-throughput, and label-free isolation of pluripotent stem cells, which will facilitate a greater biological understanding of pluripotency and advance the potential of pluripotent stem cell differentiated progeny as cell sources for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Tom Bongiorno
- The G. W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Jeremy Gura
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, United States of America
| | - Priyanka Talwar
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, United States of America
| | - Dwight Chambers
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, United States of America
| | - Katherine M. Young
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, United States of America
| | - Dalia Arafat
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Gonghao Wang
- The G. W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Emily L. Jackson-Holmes
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Peng Qiu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, United States of America
| | - Todd C. McDevitt
- Gladstone Institute for Cardiovascular Disease, San Francisco, CA, United States of America
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, United States of America
| | - Todd Sulchek
- The G. W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, United States of America
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, United States of America
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States of America
- * E-mail:
| |
Collapse
|
15
|
KIMURA T, NAKAMURA N, HASHIMOTO Y, SAKAGUCHI S, KIMURA S, KISHIDA A. Selective Cell Capture and Release Using Antibody-Immobilized Polymer-Grafted Surface. KOBUNSHI RONBUNSHU 2018. [DOI: 10.1295/koron.2017-0074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Tsuyoshi KIMURA
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University
| | - Naoko NAKAMURA
- College of Systems Engineering and Science, Shibaura Institute of Technology
| | - Yoshihide HASHIMOTO
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University
| | | | | | - Akio KISHIDA
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University
| |
Collapse
|
16
|
Abbasalizadeh S, Pakzad M, Cabral JMS, Baharvand H. Allogeneic cell therapy manufacturing: process development technologies and facility design options. Expert Opin Biol Ther 2017; 17:1201-1219. [PMID: 28699788 DOI: 10.1080/14712598.2017.1354982] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Currently, promising outcomes from clinical trials of allogeneic cells, especially allogeneic mesenchymal stromal cells, fibroblasts, keratinocytes, and human cardiac stem cells, have encouraged research institutions, small and medium enterprises (SMEs), and big pharmaceutical companies to invest and focus on developing allogeneic cell therapy products. Commercial and large-scale production of allogeneic cell therapy products requires unique capabilities to develop technologies that generate safe and effective allogeneic cells/cell lines and their fully characterized master/working banks. In addition, it is necessary to design robust upstream and downstream manufacturing processes, and establish integrated, well-designed manufacturing facilities to produce high quality affordable products in accordance with current GMP regulations for the production of cell therapy products. Areas covered: The authors highlight: the recent advances in the development of allogeneic products, the available options to develop robust manufacturing processes, and facility design considerations. Expert opinion: Currently, there are multiple challenges in development of allogeneic cell therapy products. Indeed, the field is still in its infancy; with technologies and regulations still under development, as is our understanding of the mechanisms of action in the body and their interaction with the host immune system. Their characterization and testing is also an emerging and very complex area.
Collapse
Affiliation(s)
- Saeed Abbasalizadeh
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran.,b Department of Bioengineering and Institute for Bioengineering and Biosciences , Instituto Superior Técnico, Universidade de Lisboa , Lisboa , Portugal
| | - Mohammad Pakzad
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
| | - Joaquim M S Cabral
- b Department of Bioengineering and Institute for Bioengineering and Biosciences , Instituto Superior Técnico, Universidade de Lisboa , Lisboa , Portugal
| | - Hossein Baharvand
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran.,c Department of Developmental Biology , University of Science and Culture , Tehran , Iran
| |
Collapse
|
17
|
Rodrigues GMC, Gaj T, Adil MM, Wahba J, Rao AT, Lorbeer FK, Kulkarni RU, Diogo MM, Cabral JMS, Miller EW, Hockemeyer D, Schaffer DV. Defined and Scalable Differentiation of Human Oligodendrocyte Precursors from Pluripotent Stem Cells in a 3D Culture System. Stem Cell Reports 2017; 8:1770-1783. [PMID: 28552605 PMCID: PMC5470111 DOI: 10.1016/j.stemcr.2017.04.027] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/22/2017] [Accepted: 04/24/2017] [Indexed: 12/22/2022] Open
Abstract
Oligodendrocyte precursor cells (OPCs) offer considerable potential for the treatment of demyelinating diseases and injuries of the CNS. However, generating large quantities of high-quality OPCs remains a substantial challenge that impedes their therapeutic application. Here, we show that OPCs can be generated from human pluripotent stem cells (hPSCs) in a three-dimensional (3D), scalable, and fully defined thermoresponsive biomaterial system. We used CRISPR/Cas9 to create a NKX2.2-EGFP human embryonic stem cell reporter line that enabled fine-tuning of early OPC specification and identification of conditions that markedly increased the number of OLIG2+ and NKX2.2+ cells generated from hPSCs. Transplantation of 50-day-old OPCs into the brains of NOD/SCID mice revealed that progenitors generated in 3D without cell selection or purification subsequently engrafted, migrated, and matured into myelinating oligodendrocytes in vivo. These results demonstrate the potential of harnessing lineage reporter lines to develop 3D platforms for rapid and large-scale production of OPCs. A defined and scalable 3D system accelerates the differentiation of OPCs from hPSCs A NKX2.2-EGFP hESC reporter line enables optimization of OPC differentiation 3D-derived OPCs engraft, migrate, and mature after implantation into NOD/SCID mice
Collapse
Affiliation(s)
- Gonçalo M C Rodrigues
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720-1762, USA; Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisbon, Portugal
| | - Thomas Gaj
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720-1762, USA
| | - Maroof M Adil
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720-1462, USA
| | - Joyce Wahba
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720-1462, USA
| | - Antara T Rao
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720-1462, USA
| | - Franziska K Lorbeer
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Rishi U Kulkarni
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720-1462, USA
| | - Maria Margarida Diogo
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisbon, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisbon, Portugal
| | - Evan W Miller
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720-1462, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720-3370, USA; The Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720-3370, USA
| | - Dirk Hockemeyer
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - David V Schaffer
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720-1762, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720-1462, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720-3370, USA; The Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720-3370, USA.
| |
Collapse
|
18
|
Enomoto J, Kageyama T, Osaki T, Bonalumi F, Marchese F, Gautieri A, Bianchi E, Dubini G, Arrigoni C, Moretti M, Fukuda J. Catch-and-Release of Target Cells Using Aptamer-Conjugated Electroactive Zwitterionic Oligopeptide SAM. Sci Rep 2017; 7:43375. [PMID: 28266533 PMCID: PMC5339905 DOI: 10.1038/srep43375] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/23/2017] [Indexed: 11/18/2022] Open
Abstract
Nucleic acid aptamers possess attractive features such as specific molecular recognition, high-affinity binding, and rapid acquisition and replication, which could be feasible components for separating specific cells from other cell types. This study demonstrates that aptamers conjugated to an oligopeptide self-assembled monolayer (SAM) can be used to selectively trap human hepatic cancer cells from cell mixtures containing normal human hepatocytes or human fibroblasts. Molecular dynamics calculations have been performed to understand how the configurations of the aptamers are related to the experimental results of selective cell capture. We further demonstrate that the captured hepatic cancer cells can be detached and collected along with electrochemical desorption of the oligopeptide SAM, and by repeating these catch-and-release processes, target cells can be enriched. This combination of capture with aptamers and detachment with electrochemical reactions is a promising tool in various research fields ranging from basic cancer research to tissue engineering applications.
Collapse
Affiliation(s)
- Junko Enomoto
- Graduate School of Engineering, Yokohama National University, Japan
| | - Tatsuto Kageyama
- Graduate School of Engineering, Yokohama National University, Japan
| | - Tatsuya Osaki
- Graduate School of Engineering, Yokohama National University, Japan
| | - Flavia Bonalumi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Italy
| | - Francesca Marchese
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Italy
| | - Alfonso Gautieri
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Italy
| | - Elena Bianchi
- Department of Chemistry, Politecnico di Milan, Italy
| | | | - Chiara Arrigoni
- Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute, Italy
| | - Matteo Moretti
- Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute, Italy
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale, Switzerland
- Swiss Institute for Regenerative Medicine, Switzerland
- Cardiocentro Ticino, Switzerland
| | - Junji Fukuda
- Graduate School of Engineering, Yokohama National University, Japan
| |
Collapse
|
19
|
Lambrechts T, Sonnaert M, Schrooten J, Luyten FP, Aerts JM, Papantoniou I. Large-Scale Mesenchymal Stem/Stromal Cell Expansion: A Visualization Tool for Bioprocess Comparison. TISSUE ENGINEERING PART B-REVIEWS 2016; 22:485-498. [DOI: 10.1089/ten.teb.2016.0111] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Toon Lambrechts
- M3-BIORES: Measure, Model and Manage Bioresponses, KU Leuven, Leuven, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Maarten Sonnaert
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Department of Metallurgy and Materials Engineering, KU Leuven, Leuven, Belgium
| | - Jan Schrooten
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Antleron, Leuven, Belgium
| | - Frank P. Luyten
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Jean-Marie Aerts
- M3-BIORES: Measure, Model and Manage Bioresponses, KU Leuven, Leuven, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Ioannis Papantoniou
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| |
Collapse
|
20
|
Faye PA, Vedrenne N, De la Cruz-Morcillo MA, Barrot CC, Richard L, Bourthoumieu S, Sturtz F, Funalot B, Lia AS, Battu S. New Method for Sorting Endothelial and Neural Progenitors from Human Induced Pluripotent Stem Cells by Sedimentation Field Flow Fractionation. Anal Chem 2016; 88:6696-702. [PMID: 27263863 DOI: 10.1021/acs.analchem.6b00704] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Human induced pluripotent stem cells (hiPSc) are a very useful solution to create and observe the behavior of specific and usually inaccessible cells, such as human motor neurons. Obtained from a patient biopsy by reprograming dermal fibroblasts (DF), hiPSc present the same properties as embryonic stem cells and can generate any cell type after several weeks of differentiation. Today, there are numerus protocols which aim to control hiPSC differentiation. The principal challenge is to obtain a sufficiently enriched specific cell population to study disease pathophysiology and to provide a good model for further investigation and drug screening. The differentiation process is very costly and time-consuming, because many specific factors and different culture media must be used. In this study, we used Sedimentation Field Flow Fractionation (SdFFF) to prepare enriched populations derived from hiPSc after only 10 days of culture in a classical medium. Based on phenotypic and proteomic characterization, "hyperlayer" elution resulted in a fraction expressing markers of endothelial progenitors while another fraction expressed markers of neural progenitors. The isolation of subpopulations representing various differentiation lineages is of major interest for the production of specialized, cell-enriched fractions and in the preparation of increasingly complex models for the development of new therapeutic tools.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Benoît Funalot
- Département de Génétique, CHU Henri-Mondor , F-94000 Créteil, France.,Inserm U955-E10, Université Paris-Est-Créteil, F-94000 Créteil, France
| | | | | |
Collapse
|
21
|
Zimmermann S, Gretzinger S, Scheeder C, Schwab ML, Oelmeier SA, Osberghaus A, Gottwald E, Hubbuch J. High-throughput cell quantification assays for use in cell purification development - enabling technologies for cell production. Biotechnol J 2016; 11:676-86. [DOI: 10.1002/biot.201500577] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/24/2015] [Accepted: 01/22/2016] [Indexed: 01/17/2023]
Affiliation(s)
- Sarah Zimmermann
- Karlsruhe Institute of Technology (KIT), Institute of Process Engineering in Life Science, Section IV: Biomolecular Separation Engineering (MAB); Karlsruhe Germany
| | - Sarah Gretzinger
- Karlsruhe Institute of Technology (KIT), Institute of Process Engineering in Life Science, Section IV: Biomolecular Separation Engineering (MAB); Karlsruhe Germany
| | - Christian Scheeder
- Karlsruhe Institute of Technology (KIT), Institute of Process Engineering in Life Science, Section IV: Biomolecular Separation Engineering (MAB); Karlsruhe Germany
| | - Marie-Luise Schwab
- Karlsruhe Institute of Technology (KIT), Institute of Process Engineering in Life Science, Section IV: Biomolecular Separation Engineering (MAB); Karlsruhe Germany
- DIARECT AG, Department of Quality Assurance and Quality Control; Freiburg Germany
| | - Stefan A. Oelmeier
- Karlsruhe Institute of Technology (KIT), Institute of Process Engineering in Life Science, Section IV: Biomolecular Separation Engineering (MAB); Karlsruhe Germany
- Boehringer Ingelheim Pharma GmbH & Co. KG, Global Bioprocess & Pharmaceutical Development; Biberach Germany
| | - Anna Osberghaus
- Karlsruhe Institute of Technology (KIT), Institute of Process Engineering in Life Science, Section IV: Biomolecular Separation Engineering (MAB); Karlsruhe Germany
| | - Eric Gottwald
- Karlsruhe Institute of Technology (KIT), Institute for Biological Interfaces (IBG 5); Eggenstein-Leopoldshafen Germany
| | - Jürgen Hubbuch
- Karlsruhe Institute of Technology (KIT), Institute of Process Engineering in Life Science, Section IV: Biomolecular Separation Engineering (MAB); Karlsruhe Germany
| |
Collapse
|
22
|
Gu W, Sun W, Guo C, Yan Y, Liu M, Yao X, Yang B, Zheng J. Culture and Characterization of Circulating Endothelial Progenitor Cells in Patients with Renal Cell Carcinoma. J Urol 2015; 194:214-22. [PMID: 25659661 DOI: 10.1016/j.juro.2015.01.100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2015] [Indexed: 12/20/2022]
Affiliation(s)
- Wenyu Gu
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Wei Sun
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Changcheng Guo
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yang Yan
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Min Liu
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Bin Yang
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Junhua Zheng
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
23
|
Rodrigues GMC, Rodrigues CAV, Fernandes TG, Diogo MM, Cabral JMS. Clinical-scale purification of pluripotent stem cell derivatives for cell-based therapies. Biotechnol J 2015; 10:1103-14. [PMID: 25851544 DOI: 10.1002/biot.201400535] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/20/2015] [Accepted: 03/04/2015] [Indexed: 01/12/2023]
Abstract
Human pluripotent stem cells (hPSCs) have the potential to revolutionize cell-replacement therapies because of their ability to self renew and differentiate into nearly every cell type in the body. However, safety concerns have delayed the clinical translation of this technology. One cause for this is the capacity that hPSCs have to generate tumors after transplantation. Because of the challenges associated with achieving complete differentiation into clinically relevant cell types, the development of safe and efficient strategies for purifying committed cells is essential for advancing hPSC-based therapies. Several purification strategies have now succeeded in generating non-tumorigenic and homogeneous cell-populations. These techniques typically enrich for cells by either depleting early committed populations from teratoma-initiating hPSCs or by positively selecting cells after differentiation. Here we review the working principles behind separation methods that have facilitated the safe and controlled application of hPSC-derived cells in laboratory settings and pre-clinical research. We underscore the need for improving and integrating purification strategies within differentiation protocols in order to unlock the therapeutic potential of hPSCs.
Collapse
Affiliation(s)
- Gonçalo M C Rodrigues
- Department of Bioengineering and IBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Carlos A V Rodrigues
- Department of Bioengineering and IBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Tiago G Fernandes
- Department of Bioengineering and IBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Margarida Diogo
- Department of Bioengineering and IBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| | - Joaquim M S Cabral
- Department of Bioengineering and IBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
24
|
Abstract
Anchorage-dependent cells are of great interest for various biotechnological applications. (i) They represent a formidable production means of viruses for vaccination purposes at very large scales (in 1000-6000 l reactors) using microcarriers, and in the last decade many more novel viral vaccines have been developed using this production technology. (ii) With the advent of stem cells and their use/potential use in clinics for cell therapy and regenerative medicine purposes, the development of novel culture devices and technologies for adherent cells has accelerated greatly with a view to the large-scale expansion of these cells. Presently, the really scalable systems--microcarrier/microcarrier-clump cultures using stirred-tank reactors--for the expansion of stem cells are still in their infancy. Only laboratory scale reactors of maximally 2.5 l working volume have been evaluated because thorough knowledge and basic understanding of critical issues with respect to cell expansion while retaining pluripotency and differentiation potential, and the impact of the culture environment on stem cell fate, etc., are still lacking and require further studies. This article gives an overview on critical issues common to all cell culture systems for adherent cells as well as specifics for different types of stem cells in view of small- and large-scale cell expansion and production processes.
Collapse
|
25
|
Rodrigues GMC, Fernandes TG, Rodrigues CAV, Cabral JMS, Diogo MM. Purification of human induced pluripotent stem cell-derived neural precursors using magnetic activated cell sorting. Methods Mol Biol 2015; 1283:137-145. [PMID: 25537837 DOI: 10.1007/7651_2014_115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Neural precursor (NP) cells derived from human induced pluripotent stem cells (hiPSCs), and their neuronal progeny, will play an important role in disease modeling, drug screening tests, central nervous system development studies, and may even become valuable for regenerative medicine treatments. Nonetheless, it is challenging to obtain homogeneous and synchronously differentiated NP populations from hiPSCs, and after neural commitment many pluripotent stem cells remain in the differentiated cultures. Here, we describe an efficient and simple protocol to differentiate hiPSC-derived NPs in 12 days, and we include a final purification stage where Tra-1-60+ pluripotent stem cells (PSCs) are removed using magnetic activated cell sorting (MACS), leaving the NP population nearly free of PSCs.
Collapse
Affiliation(s)
- Gonçalo M C Rodrigues
- Department of Bioengineering and IBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001, Lisbon, Portugal
| | | | | | | | | |
Collapse
|
26
|
Integrated platform for production and purification of human pluripotent stem cell-derived neural precursors. Stem Cell Rev Rep 2014; 10:151-61. [PMID: 24221956 DOI: 10.1007/s12015-013-9482-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Human pluripotent stem cells (hPSCs) are a promising source of cells for clinical applications, such as transplantation of clinically engineered tissues and organs, and drug discovery programs due to their ability to self-renew and to be differentiated into cells from the three embryonic germ layers. In this study, the differentiation of two hPSC-lines into neural precursors (NPs) was accomplished with more than 80% efficiency, by means of the dual-SMAD inhibition protocol, based on the use of two small molecules (SB431542 and LDN193189) to generate Pax6 and Nestin-positive neural entities. One of the major hurdles related to the in vitro generation of PSC-derived populations is the tumorigenic potential of cells that remain undifferentiated. These remaining hPSCs have the potential to generate teratomas after being transplanted, and may interfere with the outcome of in vitro differentiation protocols. One strategy to tackle this problem is to deplete these "contaminating" cells during the differentiation process. Magnetic activated cell sorting (MACS) was used for the first time for purification of hPSC-derived NPs after the neural commitment stage using anti-Tra-1-60 micro beads for negative selection of the unwanted hPSCs. The depletion had an average efficiency of 80.4 ± 5% and less than 1.5% of Tra-1-60 positive cells were present in the purified populations. After re-plating, the purified neural precursors maintained their phenotype, and the success of the preparative purification with MACS was further confirmed with a decrease of 94.3% in the number of Oct4-positive proliferating hPSC colonies. Thus, the integration of the MACS depletion step with the neural commitment protocol paves the way towards the establishment of a novel bioprocess for production of purified populations of hPSC-derived neural cells for different applications.
Collapse
|
27
|
Relationship between preparation of cells for therapy and cell quality using artificial neural network analysis. Artif Intell Med 2014; 62:119-27. [DOI: 10.1016/j.artmed.2014.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/29/2014] [Accepted: 07/12/2014] [Indexed: 11/23/2022]
|
28
|
Loring JF, McDevitt TC, Palecek SP, Schaffer DV, Zandstra PW, Nerem RM. A global assessment of stem cell engineering. Tissue Eng Part A 2014; 20:2575-89. [PMID: 24428577 DOI: 10.1089/ten.tea.2013.0468] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Over the last 2 years a global assessment of stem cell engineering (SCE) was conducted with the sponsorship of the National Science Foundation, the National Cancer Institute at the National Institutes of Health, and the National Institute of Standards and Technology. The purpose was to gather information on the worldwide status and trends in SCE, that is, the involvement of engineers and engineering approaches in the stem cell field, both in basic research and in the translation of research into clinical applications and commercial products. The study was facilitated and managed by the World Technology Evaluation Center. The process involved site visits in both Asia and Europe, and it also included several different workshops. From this assessment, the panel concluded that there needs to be an increased role for engineers and the engineering approach. This will provide a foundation for the generation of new markets and future economic growth. To do this will require an increased investment in engineering, applied research, and commercialization as it relates to stem cell research and technology. It also will require programs that support interdisciplinary teams, new innovative mechanisms for academic-industry partnerships, and unique translational models. In addition, the global community would benefit from forming strategic partnerships between countries that can leverage existing and emerging strengths in different institutions. To implement such partnerships will require multinational grant programs with appropriate review mechanisms.
Collapse
Affiliation(s)
- Jeanne F Loring
- 1 Director, Center for Regenerative Medicine, the Scripps Research Institute , LaJolla, California
| | | | | | | | | | | |
Collapse
|
29
|
Sart S, Schneider YJ, Li Y, Agathos SN. Stem cell bioprocess engineering towards cGMP production and clinical applications. Cytotechnology 2014; 66:709-22. [PMID: 24500393 DOI: 10.1007/s10616-013-9687-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 12/31/2013] [Indexed: 12/17/2022] Open
Abstract
Stem cells, including mesenchymal stem cells and pluripotent stem cells, are becoming an indispensable tool for various biomedical applications including drug discovery, disease modeling, and tissue engineering. Bioprocess engineering, targeting large scale production, provides a platform to generate a controlled microenvironment that could potentially recreate the stem cell niche to promote stem cell proliferation or lineage-specific differentiation. This survey aims at defining the characteristics of stem cell populations currently in use and the present-day limits in their applications for therapeutic purposes. Furthermore, a bioprocess engineering strategy based on bioreactors and 3-D cultures is discussed in order to achieve the improved stem cell yield, function, and safety required for production under current good manufacturing practices.
Collapse
Affiliation(s)
- Sébastien Sart
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, 2525 Pottsdamer St, Tallahassee, FL, 32310, USA
| | | | | | | |
Collapse
|
30
|
Li Y, Liu M, Yang ST. Dendritic cells derived from pluripotent stem cells: Potential of large scale production. World J Stem Cells 2014; 6:1-10. [PMID: 24567783 PMCID: PMC3927009 DOI: 10.4252/wjsc.v6.i1.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 10/23/2013] [Accepted: 11/05/2013] [Indexed: 02/06/2023] Open
Abstract
Human pluripotent stem cells (hPSCs), including human embryonic stem cells and human induced pluripotent stem cells, are promising sources for hematopoietic cells due to their unlimited growth capacity and the pluripotency. Dendritic cells (DCs), the unique immune cells in the hematopoietic system, can be loaded with tumor specific antigen and used as vaccine for cancer immunotherapy. While autologous DCs from peripheral blood are limited in cell number, hPSC-derived DCs provide a novel alternative cell source which has the potential for large scale production. This review summarizes recent advances in differentiating hPSCs to DCs through the intermediate stage of hematopoietic stem cells. Step-wise growth factor induction has been used to derive DCs from hPSCs either in suspension culture of embryoid bodies (EBs) or in co-culture with stromal cells. To fulfill the clinical potential of the DCs derived from hPSCs, the bioprocess needs to be scaled up to produce a large number of cells economically under tight quality control. This requires the development of novel bioreactor systems combining guided EB-based differentiation with engineered culture environment. Hence, recent progress in using bioreactors for hPSC lineage-specific differentiation is reviewed. In particular, the potential scale up strategies for the multistage DC differentiation and the effect of shear stress on hPSC differentiation in bioreactors are discussed in detail.
Collapse
|
31
|
Diogo MM, da Silva CL, Cabral JMS. Separation Technologies for Stem Cell Bioprocessing. CELL ENGINEERING 2014. [DOI: 10.1007/978-94-007-7196-3_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
32
|
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
|
33
|
Liu N, Ouyang A, Li Y, Yang ST. Three-dimensional neural differentiation of embryonic stem cells with ACM induction in microfibrous matrices in bioreactors. Biotechnol Prog 2013; 29:1013-22. [PMID: 23657995 DOI: 10.1002/btpr.1742] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Revised: 03/17/2013] [Indexed: 12/21/2022]
Abstract
The clinical use of pluripotent stem cell (PSC)-derived neural cells requires an efficient differentiation process for mass production in a bioreactor. Toward this goal, neural differentiation of murine embryonic stem cells (ESCs) in three-dimensional (3D) polyethylene terephthalate microfibrous matrices was investigated in this study. To streamline the process and provide a platform for process integration, the neural differentiation of ESCs was induced with astrocyte-conditioned medium without the formation of embryoid bodies, starting from undifferentiated ESC aggregates expanded in a suspension bioreactor. The 3D neural differentiation was able to generate a complex neural network in the matrices. When compared to 2D differentiation, 3D differentiation in microfibrous matrices resulted in a higher percentage of nestin-positive cells (68% vs. 54%) and upregulated gene expressions of nestin, Nurr1, and tyrosine hydroxylase. High purity of neural differentiation in 3D microfibrous matrix was also demonstrated in a spinner bioreactor with 74% nestin + cells. This study demonstrated the feasibility of a scalable process based on 3D differentiation in microfibrous matrices for the production of ESC-derived neural cells.
Collapse
Affiliation(s)
- Ning Liu
- William G. Lowrie Dept. of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | |
Collapse
|
34
|
Turaç G, Hindley CJ, Thomas R, Davis JA, Deleidi M, Gasser T, Karaöz E, Pruszak J. Combined flow cytometric analysis of surface and intracellular antigens reveals surface molecule markers of human neuropoiesis. PLoS One 2013; 8:e68519. [PMID: 23826393 PMCID: PMC3691147 DOI: 10.1371/journal.pone.0068519] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 05/30/2013] [Indexed: 02/03/2023] Open
Abstract
Surface molecule profiles undergo dynamic changes in physiology and pathology, serve as markers of cellular state and phenotype and can be exploited for cell selection strategies and diagnostics. The isolation of well-defined cell subsets is needed for in vivo and in vitro applications in stem cell biology. In this technical report, we present an approach for defining a subset of interest in a mixed cell population by flow cytometric detection of intracellular antigens. We have developed a fully validated protocol that enables the co-detection of cluster of differentiation (CD) surface antigens on fixed, permeabilized neural cell populations defined by intracellular staining. Determining the degree of co-expression of surface marker candidates with intracellular target population markers (nestin, MAP2, doublecortin, TUJ1) on neuroblastoma cell lines (SH-SY5Y, BE(2)-M17) yielded a combinatorial CD49f-/CD200high surface marker panel. Its application in fluorescence-activated cell sorting (FACS) generated enriched neuronal cultures from differentiated cell suspensions derived from human induced pluripotent stem cells. Our data underlines the feasibility of using the described co-labeling protocol and co-expression analysis for quantitative assays in mammalian neurobiology and for screening approaches to identify much needed surface markers in stem cell biology.
Collapse
Affiliation(s)
- Gizem Turaç
- Emmy Noether-Group for Stem Cell Biology, Department of Molecular Embryology, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey
| | - Christopher J. Hindley
- Emmy Noether-Group for Stem Cell Biology, Department of Molecular Embryology, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
| | - Ria Thomas
- Emmy Noether-Group for Stem Cell Biology, Department of Molecular Embryology, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Jason A. Davis
- Emmy Noether-Group for Stem Cell Biology, Department of Molecular Embryology, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
| | - Michela Deleidi
- 4 Hertie, Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), University of Tübingen, Tübingen, Germany
| | - Thomas Gasser
- 4 Hertie, Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), University of Tübingen, Tübingen, Germany
| | - Erdal Karaöz
- Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey
| | - Jan Pruszak
- Emmy Noether-Group for Stem Cell Biology, Department of Molecular Embryology, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
- Center for Biological Signaling Studies (BIOSS), University of Freiburg, Freiburg, Germany
- *
| |
Collapse
|
35
|
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: 76] [Impact Index Per Article: 6.3] [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
|
36
|
Andrade PZ, dos Santos F, Cabral JMS, da Silva CL. Stem cell bioengineering strategies to widen the therapeutic applications of haematopoietic stem/progenitor cells from umbilical cord blood. J Tissue Eng Regen Med 2013; 9:988-1003. [PMID: 23564692 DOI: 10.1002/term.1741] [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: 07/30/2012] [Revised: 01/18/2013] [Accepted: 02/05/2013] [Indexed: 12/11/2022]
Abstract
Umbilical cord blood (UCB) transplantation has observed a significant increase in recent years, due to the unique features of UCB haematopoietic stem/progenitor cells (HSCs) for the treatment of blood-related disorders. However, the low cell numbers available per UCB unit significantly impairs the widespread use of this source for transplantation of adult patients, resulting in graft failure, delayed engraftment and delayed immune reconstitution. In order to overcome this issue, distinct approaches are now being considered in clinical trials, such as double-UCB transplantation, intrabone injection or ex vivo expansion. In this article the authors review the current state of the art, future trends and challenges on the ex vivo expansion of UCB HSCs, focusing on culture parameters affecting the yield and quality of the expanded HSC grafts: novel HSC selection schemes prior to cell culture, cytokine/growth factor cocktails, the impact of biochemical factors (e.g. O2 ) or the addition of supportive cells, e.g. mesenchymal stem/stromal cell (MSC)-based feeder layers) were addressed. Importantly, a critical challenge in cellular therapy is still the scalability, reproducibility and control of the expansion process, in order to meet the clinical requirements for therapeutic applications. Efficient design of bioreactor systems and operation modes are now the focus of many bioengineers, integrating the increasing 'know-how' on HSC biology and physiology, while complying with the GMP standards for the production of cellular products, i.e. through the use of commercially available, highly controlled, disposable technologies.
Collapse
Affiliation(s)
- Pedro Z Andrade
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Instituto Superior Técnico, Lisboa, Portugal.,Cell2b, Advanced Therapeutics, Biocant Park, Cantanhede, Portugal
| | - Francisco dos Santos
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Instituto Superior Técnico, Lisboa, Portugal.,Cell2b, Advanced Therapeutics, Biocant Park, Cantanhede, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Instituto Superior Técnico, Lisboa, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Instituto Superior Técnico, Lisboa, Portugal
| |
Collapse
|