1
|
Ahn Y, Jeong J, Choi KH, Lee DK, Lee M, Lee NY, Kim DY, Lee CK. Development of Reproducible and Scalable Culture Conditions for In Vitro Maintenance of Pig Embryonic Stem Cells Using the Sandoz Inbred Swiss Mouse Thioguanine-Resistant Ouabain-Resistant Cell Line as a Feeder Layer. Stem Cells Dev 2023; 32:747-757. [PMID: 37756363 DOI: 10.1089/scd.2023.0171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023] Open
Abstract
Feeder cells play a crucial role in maintaining the pluripotency of embryonic stem cells (ESCs) by secreting various extrinsic regulators, such as extracellular matrix (ECM) proteins and growth factors. Although primary mouse embryonic fibroblasts (MEFs) are the most widely used feeder cell type for the culture of ESCs, they have inevitable disadvantages such as batch-to-batch variation and labor-intensive isolation processes. Here, we revealed that the Sandoz inbred Swiss Mouse (SIM) thioguanine-resistant ouabain-resistant (STO) cell line, an immortalized cell line established from mouse SIM embryonic fibroblasts, can be used as a feeder layer for in vitro culture of authentic pig ESCs instead of primary MEFs. First, the expression of genes encoding ECM proteins and growth factors was analyzed to compare their secretory functions as feeder cells. Quantitative real-time polymerase chain reaction (qPCR) showed that the gene expression of these pluripotency-associated factors was downregulated in STO cells compared to primary MEFs of similar density. Therefore, subsequent optimization of the culture conditions was attempted using higher STO cell densities. Notably, pig ESCs cultured on STO cell density of 3 × (187,500 cells/cm2) exhibited the most similar pluripotent state to pig ESCs cultured on primary MEF density of 1 × (62,500 cells/cm2), as determined by alkaline phosphatase staining, qPCR, and immunocytochemistry. In addition, pig ESCs cultured on STO cell density of 3 × formed complex teratoma containing multiple types of tissues derived from all three germ layers. Our culture conditions using optimal STO cell density can be applied to fields requiring reproducible and scalable production of pig ESCs, such as preclinical research and cellular agriculture.
Collapse
Affiliation(s)
- Yelim Ahn
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute for Agriculture and Life Science; College of Veterinary Medicine; Seoul National University, Seoul, South Korea
| | - Jinsol Jeong
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute for Agriculture and Life Science; College of Veterinary Medicine; Seoul National University, Seoul, South Korea
| | - Kwang-Hwan Choi
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute for Agriculture and Life Science; College of Veterinary Medicine; Seoul National University, Seoul, South Korea
- Research and Development Center, Space F Corporation, Hwasung, South Korea
| | - Dong-Kyung Lee
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute for Agriculture and Life Science; College of Veterinary Medicine; Seoul National University, Seoul, South Korea
- Research and Development Center, Space F Corporation, Hwasung, South Korea
| | - Mingyun Lee
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute for Agriculture and Life Science; College of Veterinary Medicine; Seoul National University, Seoul, South Korea
| | - Na-Young Lee
- Department of Veterinary Pathology, College of Veterinary Medicine; Seoul National University, Seoul, South Korea
| | - Dae-Yong Kim
- Department of Veterinary Pathology, College of Veterinary Medicine; Seoul National University, Seoul, South Korea
| | - Chang-Kyu Lee
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute for Agriculture and Life Science; College of Veterinary Medicine; Seoul National University, Seoul, South Korea
- Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, South Korea
| |
Collapse
|
2
|
Ghorbani S, Christine Füchtbauer A, Møllebjerg A, Møller Martensen P, Hvidbjerg Laursen S, Christian Evar Kraft D, Kjems J, Meyer RL, Rahimi K, Foss M, Füchtbauer EM, Sutherland DS. Protein ligand and nanotopography separately drive the phenotype of mouse embryonic stem cells. Biomaterials 2023; 301:122244. [PMID: 37459700 DOI: 10.1016/j.biomaterials.2023.122244] [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: 01/02/2023] [Revised: 07/09/2023] [Accepted: 07/12/2023] [Indexed: 09/06/2023]
Abstract
Biochemical and biomechanical signals regulate stem cell function in the niche environments in vivo. Current in vitro culture of mouse embryonic stem cells (mESC) uses laminin (LN-511) to provide mimetic biochemical signaling (LN-521 for human systems) to maintain stemness. Alternative approaches propose topographical cues to provide biomechanical cues, however combined biochemical and topographic cues may better mimic the in vivo environment, but are largely unexplored for in vitro stem cell expansion. In this study, we directly compare in vitro signals from LN-511 and/or topographic cues to maintain stemness, using systematically-varied submicron pillar patterns or flat surfaces with or without preadsorbed LN-511. The adhesion of cells, colony formation, expression of the pluripotency marker,octamer-binding transcription factor 4 (Oct4), and transcriptome profiling were characterized. We observed that either biochemical or topographic signals could maintain stemness of mESCs in feeder-free conditions, indicated by high-level Oct4 and gene profiling by RNAseq. The combination of LN-511 with nanotopography reduced colony growth, while maintaining stemness markers, shifted the cellular phenotype indicating that the integration of biochemical and topographic signals is antagonistic. Overall, significantly faster (up to 2.5 times) colony growth was observed at nanotopographies without LN-511, suggesting for improved ESC expansion.
Collapse
Affiliation(s)
- Sadegh Ghorbani
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; The Centre for Cellular Signal Patterns (CELLPAT), Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
| | | | - Andreas Møllebjerg
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| | | | - Sara Hvidbjerg Laursen
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| | - David Christian Evar Kraft
- Department of Dentistry and Oral Health, Faculty of Health, University of Aarhus, Aarhus C, 8000, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; The Centre for Cellular Signal Patterns (CELLPAT), Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; Department of Molecular Biology, University of Aarhus, Aarhus C, 8000, Denmark
| | - Rikke Louise Meyer
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| | - Karim Rahimi
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; Department of Molecular Biology, University of Aarhus, Aarhus C, 8000, Denmark
| | - Morten Foss
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| | | | - Duncan S Sutherland
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; The Centre for Cellular Signal Patterns (CELLPAT), Gustav Wieds Vej 14, Aarhus C, 8000, Denmark.
| |
Collapse
|
3
|
Watanabe M, Buth JE, Haney JR, Vishlaghi N, Turcios F, Elahi LS, Gu W, Pearson CA, Kurdian A, Baliaouri NV, Collier AJ, Miranda OA, Dunn N, Chen D, Sabri S, Torre-Ubieta LDL, Clark AT, Plath K, Christofk HR, Kornblum HI, Gandal MJ, Novitch BG. TGFβ superfamily signaling regulates the state of human stem cell pluripotency and capacity to create well-structured telencephalic organoids. Stem Cell Reports 2022; 17:2220-2238. [PMID: 36179695 PMCID: PMC9561534 DOI: 10.1016/j.stemcr.2022.08.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 12/25/2022] Open
Abstract
Telencephalic organoids generated from human pluripotent stem cells (hPSCs) are a promising system for studying the distinct features of the developing human brain and the underlying causes of many neurological disorders. While organoid technology is steadily advancing, many challenges remain, including potential batch-to-batch and cell-line-to-cell-line variability, and structural inconsistency. Here, we demonstrate that a major contributor to cortical organoid quality is the way hPSCs are maintained prior to differentiation. Optimal results were achieved using particular fibroblast-feeder-supported hPSCs rather than feeder-independent cells, differences that were reflected in their transcriptomic states at the outset. Feeder-supported hPSCs displayed activation of diverse transforming growth factor β (TGFβ) superfamily signaling pathways and increased expression of genes connected to naive pluripotency. We further identified combinations of TGFβ-related growth factors that are necessary and together sufficient to impart broad telencephalic organoid competency to feeder-free hPSCs and enhance the formation of well-structured brain tissues suitable for disease modeling.
Collapse
Affiliation(s)
- Momoko Watanabe
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Jessie E Buth
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jillian R Haney
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Neda Vishlaghi
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Felix Turcios
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lubayna S Elahi
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Wen Gu
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Caroline A Pearson
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Arinnae Kurdian
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Natella V Baliaouri
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Amanda J Collier
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Osvaldo A Miranda
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Natassia Dunn
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Di Chen
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shan Sabri
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Luis de la Torre-Ubieta
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Amander T Clark
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kathrin Plath
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Heather R Christofk
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Harley I Kornblum
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michael J Gandal
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Bennett G Novitch
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
4
|
Thin films of functionalized carbon nanotubes support long-term maintenance and cardio-neuronal differentiation of canine induced pluripotent stem cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 40:102487. [PMID: 34740869 DOI: 10.1016/j.nano.2021.102487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 03/25/2021] [Indexed: 01/24/2023]
Abstract
Induced pluripotent stem cells (iPSCs) are a promising cell source for regenerative medicine. However, their feeder-free maintenance in undifferentiated states remains challenging. In recent past extensive studies have been directed using pristine or functionalized carbon nanotube in tissue engineering. Here we proposed thin films of functionalized carbon nanotubes (OH-single-walled CNTs [SWCNTs] and OH-multiwalled CNTs [MWCNTs]), as alternatives for the feeder-free in vitro culture of canine iPSCs (ciPSCs), considered as the cellular model. The ciPSC colonies could maintain their dome-shaped compactness and other characteristics when propagated on CNT films. Concomitantly, high cell viability and upregulation of pluripotency-associated genes and cell adhesion molecules were observed, further supported by molecular docking. Moreover, CNTs did not have profound toxic effects compared to feeder cultures as evident by cytocompatibility studies. Further, cardiac and neuronal differentiation of ciPSCs was induced on these films to determine their influence on the differentiation process. The cells retained differentiation potential and the nanotopographical features of the substrates provided positive cues to enhance differentiation to both lineages as evident by immunocytochemical staining and marker gene expression. Overall, OH-SWCNT provided better cues, maintained pluripotency, and induced the differentiation of ciPSCs. These results indicate that OH-functionalized CNT films could be used as alternatives for the feeder-free maintenance of ciPSCs towards prospective utilization in regenerative medicine.
Collapse
|
5
|
Natarajan M, Singh P, Mondal T, Kumar K, Das K, Dutt T, Bag S. In vitro propagation and cardiac differentiation of canine induced pluripotent stem cells on carbon nanotube substrates. Tissue Cell 2021; 71:101571. [PMID: 34139604 DOI: 10.1016/j.tice.2021.101571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 02/07/2023]
Abstract
Induced pluripotent stem cells (iPSCs) have attracted an interest for personalized cell based therapy along with various other applications. There have been few studies that effective nanomaterial based scaffolds act as alternative to the commonly used feeder dependent in vitro maintenance of iPSCs. The present study provides the fundamental information on ex vivo behavior of canine iPSC (ciPSCs) maintained on carboxylic acid (COOH) functionalized single-walled carbon nanotubes (COOH-SWCNTs) and multi-walled carbon nanotubes (COOH-MWCNTs) substrates. Here in we evaluated the comparative colony morphology, propagation, characterization, cytocompatibility and differentiation capability of ciPSC cultured on MEF feeder taken as control, and COOH-SWCNTs and COOH-MWCNTs substrates. We observed a healthy growth of ciPSCs on both the types of carbon nanotubes (CNTs) similar to feeder. The ciPSC colonies grown on both CNTs were positive for alkaline phosphatase staining and expressed pluripotent markers with notable significance. Further, the ciPSC colonies grew on these CNTs retained the in vitro differentiation ability into three germ layers as well as cardiac cell. Cytotoxicity analysis revealed that (COOH) functionalized CNTs provided a culture condition of low cytotoxicity. The results of the present study indicated that (COOH) functionalized CNTs could be used as xeno-free substrate to support the maintenance of iPSCs.
Collapse
Affiliation(s)
- Mahalakshmi Natarajan
- Division of Physiology and Climatology, ICAR - Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Purnima Singh
- Division of Physiology and Climatology, ICAR - Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Tanmay Mondal
- Division of Physiology and Climatology, ICAR - Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Kuldeep Kumar
- Division of Physiology and Climatology, ICAR - Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Kinsuk Das
- Department of Veterinary Physiology, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| | - Triveni Dutt
- Division of Livestock Production and Management, ICAR - Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Sadhan Bag
- Division of Physiology and Climatology, ICAR - Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India.
| |
Collapse
|
6
|
Chan SW, Rizwan M, Yim EKF. Emerging Methods for Enhancing Pluripotent Stem Cell Expansion. Front Cell Dev Biol 2020; 8:70. [PMID: 32117992 PMCID: PMC7033584 DOI: 10.3389/fcell.2020.00070] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
Pluripotent stem cells (PSCs) have great potential to revolutionize the fields of tissue engineering and regenerative medicine as well as stem cell therapeutics. However, the end goal of using PSCs for therapeutic use remains distant due to limitations in current PSC production. Conventional methods for PSC expansion have limited potential to be scaled up to produce the number of cells required for the end-goal of therapeutic use due to xenogenic components, high cost or low efficiency. In this mini review, we explore novel methods and emerging technologies of improving PSC expansion: the use of the two-dimensional mechanobiological strategies of topography and stiffness and the use of three-dimensional (3D) expansion methods including encapsulation, microcarrier-based culture, and suspension culture. Additionally, we discuss the limitations of conventional PSC expansion methods as well as the challenges in implementing non-conventional methods.
Collapse
Affiliation(s)
- Sarah W. Chan
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Muhammad Rizwan
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Evelyn K. F. Yim
- Department of Chemical Engineering, Faculty of Engineering, University of Waterloo, Waterloo, ON, Canada
- Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, Canada
- Centre for Biotechnology and Bioengineering, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
7
|
Chang J, Kim MH, Agung E, Senda S, Kino-Oka M. Effect of migratory behaviors on human induced pluripotent stem cell colony formation on different extracellular matrix proteins. Regen Ther 2018; 10:27-35. [PMID: 30525068 PMCID: PMC6260426 DOI: 10.1016/j.reth.2018.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/10/2018] [Accepted: 10/23/2018] [Indexed: 01/10/2023] Open
Abstract
Introduction Understanding how extracellular matrix (ECM) protein composition regulates the process of human induced pluripotent stem cell (hiPSC) colony formation may facilitate the design of optimal cell culture environments. In this study, we investigated the effect of migratory behaviors on hiPSC colony formation on various ECM-coated surfaces. Methods To quantify how different ECM proteins affect migratory behavior during the colony formation process, single cells were seeded onto surfaces coated with varying concentrations of different ECM proteins. Cell behavior was monitored by time-lapse observation, and quantitative analysis of migration rates in relation to colony formation patterns was performed. Actin cytoskeleton, focal adhesions, and cell–cell interactions were detected by fluorescence microscopy. Results Time-lapse observations revealed that different mechanisms of colony formation were dependent upon the migratory behavior of cells on different ECM surfaces. HiPSCs formed tight colonies on concentrated ECM substrates, while coating with dilute concentrations of ECM yielded more motile cells and colonies capable of splitting into single cells or small clusters. Enhanced migration caused a reduction of cell–cell contacts that enabled splitting or merging between cells and cell clusters, consequently reducing the efficiency of clonal colony formation. High cell-to-cell variability in migration responses to ECM surfaces elicited differential focal adhesion formation and E-cadherin expression within cells and colonies. This resulted in variability within focal adhesions and further loss of E-cadherin expression by hiPSCs. Conclusions Migration is an important factor affecting hiPSC colony-forming patterns. Regulation of migratory behavior can be an effective way to improve the expansion of hiPSCs while improving the process of clonal colony formation. We believe that this investigation provides a valuable method for understanding cell phenotypes and heterogeneity during colony formation in culture. hiPSC colony-forming patterns were dependent on migratory behavior on different ECM surfaces. Colony formation without splitting during migration improved efficiency of clonal colony formation. Variability in migration behavior elicited differential cytoskeletal formation and E-cadherin expression. Our method is valuable for understanding cell phenotypes and heterogeneity during colony formation.
Collapse
Affiliation(s)
- Jessica Chang
- Institute for Innovation, Ajinomoto Co., Inc., Kawasaki, 210-8681 Japan
| | - Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Eviryanti Agung
- Institute for Innovation, Ajinomoto Co., Inc., Kawasaki, 210-8681 Japan
| | - Sho Senda
- Institute for Innovation, Ajinomoto Co., Inc., Kawasaki, 210-8681 Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
8
|
Peterson SE, Garitaonandia I, Loring JF. The tumorigenic potential of pluripotent stem cells: What can we do to minimize it? Bioessays 2017; 38 Suppl 1:S86-95. [PMID: 27417126 DOI: 10.1002/bies.201670915] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 02/05/2016] [Accepted: 02/09/2016] [Indexed: 12/14/2022]
Abstract
Human pluripotent stem cells (hPSCs) have the potential to fundamentally change the way that we go about treating and understanding human disease. Despite this extraordinary potential, these cells also have an innate capability to form tumors in immunocompromised individuals when they are introduced in their pluripotent state. Although current therapeutic strategies involve transplantation of only differentiated hPSC derivatives, there is still a concern that transplanted cell populations could contain a small percentage of cells that are not fully differentiated. In addition, these cells have been frequently reported to acquire genetic alterations that, in some cases, are associated with certain types of human cancers. Here, we try to separate the panic from reality and rationally evaluate the true tumorigenic potential of these cells. We also discuss a recent study examining the effect of culture conditions on the genetic integrity of hPSCs. Finally, we present a set of sensible guidelines for minimizing the tumorigenic potential of hPSC-derived cells. © 2016 The Authors. Inside the Cell published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Suzanne E Peterson
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.,Center for Regenerative Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Ibon Garitaonandia
- Department of Neurogenetics, International Stem Cell Corporation, Oceanside, CA, USA
| | - Jeanne F Loring
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA.,Center for Regenerative Medicine, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
9
|
Chugh RM, Chaturvedi M, Yerneni LK. An optimization protocol for Swiss 3T3 feeder cell growth-arrest by Mitomycin C dose-to-volume derivation strategy. Cytotechnology 2017; 69:391-404. [PMID: 28110386 PMCID: PMC5366971 DOI: 10.1007/s10616-017-0064-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 01/04/2017] [Indexed: 10/20/2022] Open
Abstract
Feeder cell functionality following growth-arrest with the cost-effective Mitomycin C vis-à-vis irradiation is controversial due to several methodological variables reported. Earlier, we demonstrated variability in growth arrested Swiss 3T3 feeder cell life-span following titration of feeder cell densities with Mitomycin C concentrations which led to the derivation of doses per cell. Alternatively, to counter the unexpected feeder regrowth at high exposure cell density, we proposed titration of a fixed density with arithmetically derived volumes of Mitomycin C solution that corresponded to permutations of specific concentrations and doses per cell. We now describe an experimental procedure of inducing differential feeder cell growth-arrest by titrating with such volumes and validating the best feeder batch through target cell growth assessment. A safe cell density of Swiss 3T3 tested for the exclusion of Mitomycin C resistant variants was titrated with a range of volumes of a Mitomycin C solution. The differentially growth-arrested feeder batches generated were tested for short-term and long-term viability and human epidermal keratinocyte growth supporting ability. The feeder cell extinction rate was directly proportional to the volume of Mitomycin C solution within a given concentration per se. The keratinocyte colony forming efficiency and the overall growth in mass cultures were maximal with a median extinction rate produced by an intermediate volume, while the faster and slower extinction rates by high and low volumes, respectively, were suboptimal. The described method could counter the inadequacies of growth-arrest with Mitomycin C.
Collapse
Affiliation(s)
- Rishi Man Chugh
- Cell Biology Laboratory, National Institute of Pathology (ICMR), New Delhi, India
| | - Madhusudan Chaturvedi
- Cell Biology Laboratory, National Institute of Pathology (ICMR), New Delhi, India
- Department of Medical Elementology and Toxicology, Jamia Hamdard, New Delhi, India
| | | |
Collapse
|
10
|
Lee H, Nam D, Choi JK, Araúzo-Bravo MJ, Kwon SY, Zaehres H, Lee T, Park CY, Kang HW, Schöler HR, Kim JB. Establishment of feeder-free culture system for human induced pluripotent stem cell on DAS nanocrystalline graphene. Sci Rep 2016; 6:20708. [PMID: 26846167 PMCID: PMC4742916 DOI: 10.1038/srep20708] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 01/11/2016] [Indexed: 12/12/2022] Open
Abstract
The maintenance of undifferentiated human pluripotent stem cells (hPSC) under
xeno-free condition requires the use of human feeder cells or extracellular matrix
(ECM) coating. However, human-derived sources may cause human pathogen contamination
by viral or non-viral agents to the patients. Here we demonstrate feeder-free and
xeno-free culture system for hPSC expansion using diffusion assisted synthesis-grown
nanocrystalline graphene (DAS-NG), a synthetic non-biological nanomaterial which
completely rule out the concern of human pathogen contamination. DAS-NG exhibited
advanced biocompatibilities including surface nanoroughness, oxygen containing
functional groups and hydrophilicity. hPSC cultured on DAS-NG could maintain
pluripotency in vitro and in vivo, and especially cell
adhesion-related gene expression profile was comparable to those of cultured on
feeders, while hPSC cultured without DAS-NG differentiated spontaneously with high
expression of somatic cell-enriched adhesion genes. This feeder-free and xeno-free
culture method using DAS-NG will facilitate the generation of clinical-grade
hPSC.
Collapse
Affiliation(s)
- Hyunah Lee
- Hans Schöler Stem Cell Research Center (HSSCRC), School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), 44919 Ulsan, South Korea
| | - Donggyu Nam
- Hans Schöler Stem Cell Research Center (HSSCRC), School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), 44919 Ulsan, South Korea
| | - Jae-Kyung Choi
- SMEs Support Center, Korea Institute of Science and Technology Information, 48058 Busan, South Korea
| | - Marcos J Araúzo-Bravo
- Group of Computational Biology and Bioinformatics, Biodonostia Health Research Institute, 20014 San Sebastián, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Soon-Yong Kwon
- Hans Schöler Stem Cell Research Center (HSSCRC), School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), 44919 Ulsan, South Korea.,School of Materials Science and Engineering, Ulsan National Institute of Science and Technology (UNIST), 44919 Ulsan, South Korea
| | - Holm Zaehres
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149 Münster, Germany
| | - Taehee Lee
- Hans Schöler Stem Cell Research Center (HSSCRC), School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), 44919 Ulsan, South Korea
| | - Chan Young Park
- Hans Schöler Stem Cell Research Center (HSSCRC), School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), 44919 Ulsan, South Korea
| | - Hyun-Wook Kang
- Hans Schöler Stem Cell Research Center (HSSCRC), School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), 44919 Ulsan, South Korea
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149 Münster, Germany
| | - Jeong Beom Kim
- Hans Schöler Stem Cell Research Center (HSSCRC), School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), 44919 Ulsan, South Korea
| |
Collapse
|
11
|
Soong YK, Huang SY, Yeh CH, Wang TH, Chang KH, Cheng PJ, Shaw SWS. The use of human amniotic fluid mesenchymal stem cells as the feeder layer to establish human embryonic stem cell lines. J Tissue Eng Regen Med 2015; 9:E302-E307. [PMID: 23460275 DOI: 10.1002/term.1702] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 12/04/2012] [Accepted: 12/20/2012] [Indexed: 02/06/2023]
Abstract
Human embryonic stem cells (hESCs) are pluripotent cells that have the potential to differentiate into the three germ layers and possibly all tissues of the human body. To fulfil the clinical potentials for cell-based therapy, banks of hESC lines that express different combinations of the major histocompatibility genes should be established, preferably without exposing such cells to animal cells and proteins. In this study, we tested human amniotic fluid mesenchymal stem cells (AFMSCs) as feeder cells to support the growth of hESCs. Our results indicated that mitomycin-treated AFMSCs were able to support the newly established hESC lines CGLK-1 and CGLK-2. The hESC colonies cultured on AFMSCs expressed alkaline phosphatase (ALK-P), SSEA-4, TRA-1-60, TRA-1-81, Oct-4, Nanog and Sox-2, which are markers for undifferentiated hESCs. Chromosomal analyses of both hESC lines, CGLK-1 and CGLK-2, which were cultured on AFMSC feeders for 22 and 14 passages, respectively, were confirmed to be normal karyotypes (46, XX). The ability of AFMSCs as feeder cells to maintain the undifferentiated growth and pluripotency of hESCs was confirmed by in vivo formation of teratomas derived on AFMSC hESCs in severe combined immune-compromised mice. The use of AFMSCs for feeder cells to culture hESCs has several advantages, in that AFMSCs are not tumourigenic and can be expanded extensively with a short doubling time.
Collapse
Affiliation(s)
- Yung-Kwei Soong
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Lin-Kou Medical Centre, Chang Gung University, Tao-Yuan, Taiwan, Republic of China
| | - Shang-Yu Huang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Lin-Kou Medical Centre, Chang Gung University, Tao-Yuan, Taiwan, Republic of China
| | - Chiu-Hsiang Yeh
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Lin-Kou Medical Centre, Chang Gung University, Tao-Yuan, Taiwan, Republic of China
| | - Tzu-Hao Wang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Lin-Kou Medical Centre, Chang Gung University, Tao-Yuan, Taiwan, Republic of China
| | - Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Lin-Kou Medical Centre, Chang Gung University, Tao-Yuan, Taiwan, Republic of China
| | - Po-Jen Cheng
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Lin-Kou Medical Centre, Chang Gung University, Tao-Yuan, Taiwan, Republic of China
| | - S W Steven Shaw
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Lin-Kou Medical Centre, Chang Gung University, Tao-Yuan, Taiwan, Republic of China
| |
Collapse
|
12
|
Freude K, Pires C, Hyttel P, Hall VJ. Induced Pluripotent Stem Cells Derived from Alzheimer's Disease Patients: The Promise, the Hope and the Path Ahead. J Clin Med 2014; 3:1402-36. [PMID: 26237610 PMCID: PMC4470192 DOI: 10.3390/jcm3041402] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 11/12/2014] [Accepted: 11/14/2014] [Indexed: 02/07/2023] Open
Abstract
The future hope of generated induced pluripotent stem cells (iPS cells) from Alzheimer’s disease patients is multifold. Firstly, they may help to uncover novel mechanisms of the disease, which could lead to the development of new and unprecedented drugs for patients and secondly, they could also be directly used for screening and testing of potential new compounds for drug discovery. In addition, in the case of familial known mutations, these cells could be targeted by use of advanced gene-editing techniques to correct the mutation and be used for future cell transplantation therapies. This review summarizes the work so far in regards to production and characterization of iPS cell lines from both sporadic and familial Alzheimer’s patients and from other iPS cell lines that may help to model the disease. It provides a detailed comparison between published reports and states the present hurdles we face with this new technology. The promise of new gene-editing techniques and accelerated aging models also aim to move this field further by providing better control cell lines for comparisons and potentially better phenotypes, respectively.
Collapse
Affiliation(s)
- Kristine Freude
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Gronnegaardsvej 7, Frederiksberg C DK-1870, Denmark.
| | - Carlota Pires
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Gronnegaardsvej 7, Frederiksberg C DK-1870, Denmark.
| | - Poul Hyttel
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Gronnegaardsvej 7, Frederiksberg C DK-1870, Denmark.
| | - Vanessa Jane Hall
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Gronnegaardsvej 7, Frederiksberg C DK-1870, Denmark.
| |
Collapse
|
13
|
Yang K, Lee J, Cho SW. Engineering biomaterials for feeder-free maintenance of human pluripotent stem cells. Int J Stem Cells 2014; 5:1-5. [PMID: 24298348 DOI: 10.15283/ijsc.2012.5.1.1] [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] [Accepted: 01/05/2012] [Indexed: 02/03/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) are capable of differentiating into any type of somatic cell, a characteristic that imparts significant therapeutic potential. Human embryonic stem cells and induced pluripotent stem cells are types of hPSCs. Although hPSCs have high therapeutic potential, their clinical relevance is limited by the requirement for animal feeder layers, which maintain their pluripotency and self-renewal. hPSCs grown on animal feeder cells are at high risk for pathogen contamination and can be affected by the immunogenicity of the feeder layer. The presence of animal feeder cells also limits the scalability of hPSCs in culture because of the high cost of culturing and batch-to-batch variations. Therefore, development of feeder-free systems is imperative for robust, lower-cost, xeno-free, scalable culture of hPSCs. Biomaterials engineered with bioactive molecules such as adhesion proteins and extracellular matrix proteins, or synthetic materials such as peptides and polymers, may provide alternative substrates to animal feeder cells. This article reviews biomaterial-based, feeder-free systems for hPSC growth and maintenance, which provide clinically relevant alternatives to feeder cell systems.
Collapse
Affiliation(s)
- Kisuk Yang
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | | | | |
Collapse
|
14
|
Wei J, Han J, Zhao Y, Cui Y, Wang B, Xiao Z, Chen B, Dai J. The importance of three-dimensional scaffold structure on stemness maintenance of mouse embryonic stem cells. Biomaterials 2014; 35:7724-33. [PMID: 24930853 DOI: 10.1016/j.biomaterials.2014.05.060] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 05/21/2014] [Indexed: 12/26/2022]
Abstract
Revealing the mechanisms of cell fate regulation is important for scientific research and stem cell-based therapy. The traditional two-dimensional (2D) cultured mES cells are in a very different 2D niche from the in vivo equivalent-inner cell mass (ICM). Because the cell fate decision could be regulated by many cues which could be impacted by geometry, the traditional 2D culture system would hamper us from understanding the in vivo situations correctly. Three-dimensional (3D) scaffold was believed to provide a 3D environment closed to the in vivo one. In this work, three different scaffolds were prepared for cell culture. Several characters of mES cells were changed under 3D scaffolds culture compared to 2D, and these changes were mainly due to the alteration in geometry but not the matrix. The self-renewal of mES cells was promoted by the introducing of dimensionality. The stemness maintenance of mES was supported by all three 3D scaffolds without feeder cells in the long-time culture. Our findings demonstrated that the stemness maintenance of mES cells was promoted by the 3D geometry of scaffolds and this would provide a promising platform for ES cell research.
Collapse
Affiliation(s)
- Jianshu Wei
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Graduate School, ChineseAcademy of Sciences, Beijing 100190, China
| | - Jin Han
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Cui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bin Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
15
|
Rubashkin M, Ou G, Weaver VM. Deconstructing signaling in three dimensions. Biochemistry 2014; 53:2078-90. [PMID: 24649923 PMCID: PMC3985742 DOI: 10.1021/bi401710d] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 03/17/2014] [Indexed: 01/13/2023]
Abstract
Cells in vivo exist within the context of a multicellular tissue, where their behavior is governed by homo- and heterotypic cell-cell interactions, the material properties of the extracellular matrix, and the distribution of various soluble and physical factors. Most methods currently used to study and manipulate cellular behavior in vitro, however, sacrifice physiological relevance for experimental expediency. The fallacy of such approaches has been highlighted by the recent development and application of three-dimensional culture models to cell biology, which has revealed striking phenotypic differences in cell survival, migration, and differentiation in genetically identical cells simply by varying culture conditions. These perplexing findings beg the question of what constitutes a three-dimensional culture and why cells behave so differently in two- and three-dimensional culture formats. In the following review, we dissect the fundamental differences between two- and three-dimensional culture conditions. We begin by establishing a basic definition of what "three dimensions" means at different biological scales and discuss how dimensionality influences cell signaling across different length scales. We identify which three-dimensional features most potently influence intracellular signaling and distinguish between conserved biological principles that are maintained across culture conditions and cellular behaviors that are sensitive to microenvironmental context. Finally, we highlight state-of-the-art molecular tools amenable to the study of signaling in three dimensions under conditions that facilitate deconstruction of signaling in a more physiologically relevant manner.
Collapse
Affiliation(s)
- Matthew
G. Rubashkin
- Joint
Bioengineering Program, UC-Berkeley/UCSF, Center for Bioengineering
and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, California 94143, United States
| | - Guanqing Ou
- Joint
Bioengineering Program, UC-Berkeley/UCSF, Center for Bioengineering
and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, California 94143, United States
| | - Valerie M. Weaver
- Joint
Bioengineering Program, UC-Berkeley/UCSF, Center for Bioengineering
and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, California 94143, United States
- Departments
of Anatomy and Bioengineering and Therapeutic Sciences, Eli and Edythe
Broad Center of Regeneration Medicine and Stem Cell Research, and
UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94143, United States
| |
Collapse
|
16
|
Lim JM, Ahn JY, Lee ST. Stem cell maintenance in a different niche. Clin Exp Reprod Med 2013; 40:47-54. [PMID: 23875159 PMCID: PMC3714428 DOI: 10.5653/cerm.2013.40.2.47] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 05/30/2013] [Accepted: 06/03/2013] [Indexed: 01/22/2023] Open
Abstract
To overcome the difficulty of controlling stem cell fate and function in applications to regenerative medicine, a number of alternative approaches have been made. Recent reports demonstrate that a non-cellular niche modulating the biophysical microenvironment with chemical factors can support stem cell self-renewal. In our previous studies, early establishment was executed to optimize biophysical factors and it was subsequently found that the microgeometry of the extracellular matrix made huge differences in stem cell behavior and phenotype. We review here a three-dimensional, non-cellular niche designed to support stem cell self-renewal. The characteristics of stem cells under the designed system are further discussed.
Collapse
Affiliation(s)
- Jeong Mook Lim
- WCU Biomodulation Program, Seoul National University, Seoul, Korea. ; Department of Agricultural Biotechnology, Seoul National University, Seoul, Korea
| | | | | |
Collapse
|
17
|
Autogenic feeder free system from differentiated mesenchymal progenitor cells, maintains pluripotency of the MEL-1 human embryonic stem cells. Differentiation 2013; 85:110-8. [PMID: 23722082 DOI: 10.1016/j.diff.2013.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 12/21/2012] [Accepted: 01/23/2013] [Indexed: 12/24/2022]
Abstract
Human embryonic stem cells (hESc) are known for its pluripotency and self renewal capability, thus possess great potential in regenerative medicine. However, the lack of suitable xenofree extracellular matrix substrate inhibits further applications or the use of hESc in cell-based therapy. In this study, we described a new differentiation method, which generates a homogeneous population of mesenchymal progenitor cells (hESc-MPC) from hESc via epithelial-mesenchymal transition. The extracellular matrix (ECM) proteins from hESc-MPC had in turn supported the undifferentiated expansion of hESc. Immunocytochemistry and flow cytometry characterization of hESc-MPC revealed the presence of early mesenchymal markers. Tandem mass spectometry analysis of ECM produced by hESc-MPC revealed the presence of a mixture of extracellular proteins which includes tenascin C, fibronectin, and vitronectin. The pluripotency of hESc (MEL-1) cultured on the ECM was maintained as shown by the expression of pluripotent genes (FoxD3, Oct-4, Tdgf1, Sox-2, Nanog, hTERT, Rex1), protein markers (SSEA-3, SSEA-4, TRA-1-81, TRA-1-60, Oct-4) and the ability to differentiate into cells representative of ectoderm, endoderm and mesoderm. In summary, we have established a xeno-free autogenic feeder free system to support undifferentiated expansion of hESc, which could be of clinical relevance.
Collapse
|
18
|
Mikkola M, Toivonen S, Tamminen K, Alfthan K, Tuuri T, Satomaa T, Natunen J, Saarinen J, Tiittanen M, Lampinen M, Valmu L, Partanen J, Otonkoski T. Lectin from Erythrina cristagalli supports undifferentiated growth and differentiation of human pluripotent stem cells. Stem Cells Dev 2012; 22:707-16. [PMID: 23106381 DOI: 10.1089/scd.2012.0365] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Lectins are carbohydrate-binding proteins, which occur ubiquitously in nature and are abundant in all living organisms from bacteria to mammals. They have several biological functions among which cell adhesion is well known and characterized. Based on the characterization of the glycome of human embryonic stem cells (hESCs), we have investigated the properties of glycan-binding lectins as a novel class of culture support matrices supporting hESC culture. We report that an Erythrina cristagalli lectin (agglutinin) (ECA) matrix supported the undifferentiated growth and significantly increased the plating efficiency of both hESC and human induced pluripotent stem cells when used in conjunction with pinacidil, an antihypertensive drug with ROCK inhibition activity. As a matrix, ECA maintained pluripotency, robust proliferation with a normal karyotype, and the ability to differentiate both in vitro and in vivo. Therefore, our findings indicate that lectins are potential candidates for design of culture and differentiation methods, and that ECA is a potent simple defined matrix for human pluripotent stem cells.
Collapse
Affiliation(s)
- Milla Mikkola
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Center, University of Helsinki, Helsinki, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Olmer R, Lange A, Selzer S, Kasper C, Haverich A, Martin U, Zweigerdt R. Suspension culture of human pluripotent stem cells in controlled, stirred bioreactors. Tissue Eng Part C Methods 2012; 18:772-84. [PMID: 22519745 DOI: 10.1089/ten.tec.2011.0717] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Therapeutic and industrial applications of pluripotent stem cells and their derivatives require large cell quantities generated in defined conditions. To this end, we have translated single cell-inoculated suspension cultures of human pluripotent stem cells (hPSCs; including human induced pluripotent stem cells [hiPS] and human embryonic stem cells [hESC]) to stirred tank bioreactors. These systems that are widely used in biopharmaceutical industry allow straightforward scale up and detailed online monitoring of key process parameters. To ensure minimum medium consumption, but in parallel functional integration of all probes mandatory for process monitoring, that is, for pO₂ and pH, experiments were performed in 100 mL culture volume in a "mini reactor platform" consisting of four independently controlled vessels. By establishing defined parameters for tightly controlled cell inoculation and aggregate formation up to 2×10⁸ hiPSCs/100 mL were generated in a single process run in 7 days. Expression of pluripotency markers and ability of cells to differentiate into derivates of all three germ layers in vitro was maintained, underlining practical utility of this new process. The presented data provide key steps toward scalable mass expansion of human iPS and ES cells thereby enabling translation of stem cell research to (pre)clinical application in relevant large animal models and valuable in vitro assays for drug development and validation as well.
Collapse
Affiliation(s)
- Ruth Olmer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs-LEBAO, Department of Cardiac, Thoracic, Transplantation, and Vascular Surgery, Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
20
|
Transcriptional expression profile of cultured human embryonic stem cells in vitro and in vivo. In Vitro Cell Dev Biol Anim 2012; 48:165-74. [PMID: 22311374 DOI: 10.1007/s11626-012-9487-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 01/13/2012] [Indexed: 01/23/2023]
Abstract
The aims of this study were to analyze the spontaneous differentiation of human embryonic stem cells in vitro and in vivo and to investigate the influence of in vitro partial differentiation on in vivo teratoma formation in immunodeficient mice. Standardized methods are needed for long-term cultivation of undifferentiated stem cells and the multilineage cells that spontaneously differentiate from them. Accordingly, SA002 human embryonic stem cells were cultured on irradiated mouse embryonic fibroblasts cells, on irradiated human foreskin fibroblasts, or were cultured feeder-free using matrigel. Expression of marker protein transcripts was analyzed in undifferentiated and differentiated stem cells using real-time PCR, and both types of stem cells were transplanted subcutaneously into immunodeficient NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice to test for teratoma formation. Teratoma histology and expression profiles were subsequently characterized. Cells cultured using different conditions and morphologically undifferentiated cells had comparable marker expression profiles, showing high expression levels of markers for pluripotency and low-to-moderate expression levels of germ layer markers. Cells showing spontaneous differentiation that were cultured in feeder-free conditions in the absence of basic fibroblast growth factor demonstrated slight upregulation of sex determining region Y-box 17, connexin 32, and albumin expression at early time points, as well as expression of octamer-binding transcription factor 4, proteoglycan epitopes on podocalyxin (Trafalgar), and alkaline phosphatase. At later time points, expression of hepatocyte nuclear factor-3-beta, and hepatocyte nuclear factor-4-alpha and alpha fetoprotein was upregulated, whereas beta-3-tubulin, chemokine receptor, nestin, sex-determining region Y-box 17, and connexin 32 were downregulated. Expression of pluripotency markers remained high, and hematopoetic markers were not expressed. SA002 cells that showed spontaneous partial differentiation in vitro had a low teratoma formation capacity in vivo. Cells that were partially differentiated led to slower growing teratomas with more uniform histology.
Collapse
|
21
|
Ausubel LJ, Lopez PM, Couture LA. GMP scale-up and banking of pluripotent stem cells for cellular therapy applications. Methods Mol Biol 2011; 767:147-59. [PMID: 21822873 DOI: 10.1007/978-1-61779-201-4_11] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human pluripotent stem cells (PSCs), which include human embryonic stem cells (ESCs) as well as induced pluripotent stem cells (iPSCs), represent an important source of cellular therapies in regenerative medicine and the study of early human development. As such, it is becoming increasingly important to develop methods for the large-scale banking of human PSC lines. There are several well-established methods for the propagation of human PSCs. The key to development of a good manufacturing practice (GMP) bank is to determine a manufacturing method that is amenable to large-scale production using materials that are fully documented. We have developed several banks of hESCs using animal feeder cells, animal-based matrices, or animal-free matrices. Protocols for growing hESCs on mouse embryonic fibroblasts (MEFs) are well established and are very helpful for producing research grade banks of cells. As most human ESCs cultured by research laboratories have been exposed to xenogeneic reagents, it is not imperative that all materials used in the production of a master cell bank be animal-free in origin. Nevertheless, as the field develops, it will no doubt become increasingly important to produce a bank of cells for clinical use without xenogeneic reagents, particularly nonhuman feeder cells which might harbor viruses with potential risk to human health or cell product integrity. Thus, even for cell lines previously exposed to xenogeneic reagents, it is important to minimize any subsequent exposure of the cell lines to additional adventitious agents. We have specifically described procedures for the growth of hESCs on Matrigel, an animal-matrix, and CELLstart, an animal-free matrix, and these can be used to produce hESCs as part of a clinical manufacturing process.
Collapse
Affiliation(s)
- Lara J Ausubel
- Center for Biomedicine and Genetics, Center for Applied Technology Development, Beckman Research Institute of City of Hope, Duarte, CA, USA.
| | | | | |
Collapse
|
22
|
T'joen V, Declercq H, Cornelissen M. Expansion of human embryonic stem cells: a comparative study. Cell Prolif 2011; 44:462-76. [PMID: 21951289 DOI: 10.1111/j.1365-2184.2011.00773.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES Human embryonic stem cells (hESC) are promising for tissue engineering (TE) purposes due to their unique properties. However, current standard mechanical passaging techniques limit rates of possible TE experiments, as it is difficult to obtain high enough numbers of the cells for experimentation. In this study, several dissociative solutions and application methods are tested for their applicability to, and influence on, hESC culture and expansion. MATERIALS AND METHODS Expansion of two hESC lines, H1 and VUB01, subjected to different passaging techniques, was evaluated. Four dissociative solutions - TrypLE™ Express, Trypsin-EDTA, Cell Dissociation Solution and Accutase™- were combined with two application protocols. As reference conditions, manual and bead-based passaging techniques were used. RESULTS Results showed that use of Cell Dissociation Solution in combination with a slow adaptation protocol, generated the best expansion profile for both cell lines. The hESC single cell lines remained pluripotent, had good expansion profiles and were capable of differentiation into representatives of all three germ layers. Reproducibility of the results was confirmed by adaptation for three other hESC lines. CONCLUSION Use of Cell Dissociation Solution, combined with slow adaptation protocol, allows a fast switch from the mechanical passaging technique to a single-cell split technique, generating stable and robust hESC cell lines, which allow for large scale expansion of hESC for TE purposes.
Collapse
Affiliation(s)
- V T'joen
- Department of Basic Medical Science - Tissue Engineering Group, Faculty of Medicine and Health Science, Ghent University - UGent, Gent, Belgium.
| | | | | |
Collapse
|
23
|
Hongisto H, Vuoristo S, Mikhailova A, Suuronen R, Virtanen I, Otonkoski T, Skottman H. Laminin-511 expression is associated with the functionality of feeder cells in human embryonic stem cell culture. Stem Cell Res 2011; 8:97-108. [PMID: 22099024 DOI: 10.1016/j.scr.2011.08.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 08/19/2011] [Accepted: 08/22/2011] [Indexed: 11/30/2022] Open
Abstract
Fibroblast feeder cells play an important role in supporting the derivation and long term culture of undifferentiated, pluripotent human embryonic stem cells (hESCs). The feeder cells secrete various growth factors and extracellular matrix (ECM) proteins into extracellular milieu. However, the roles of the feeder cell-secreted factors are largely unclear. Animal feeder cells and use of animal serum also make current feeder cell culture conditions unsuitable for derivation of clinical grade hESCs. We established xeno-free feeder cell lines using human serum (HS) and studied their function in hESC culture. While human foreskin fibroblast (hFF) feeder cells were clearly hESC supportive, none of the established xeno-free human dermal fibroblast (hDF) feeder cells were able to maintain undifferentiated hESC growth. The two fibroblast types were compared for their ECM protein synthesis, integrin receptor expression profiles and key growth factor secretion. We show that hESC supportive feeder cells produce laminin-511 and express laminin-binding integrins α3ß1, α6ß1 and α7ß1. These results indicate specific laminin isoforms and integrins in maintenance of hESC pluripotency in feeder-dependent cultures. In addition, several genes with a known or possible role for hESC pluripotency were differentially expressed in distinct feeder cells.
Collapse
Affiliation(s)
- Heidi Hongisto
- Regea - Institute for Regenerative Medicine, University of Tampere, Tampere, Finland.
| | | | | | | | | | | | | |
Collapse
|
24
|
Fu X, Toh WS, Liu H, Lu K, Li M, Cao T. Establishment of clinically compliant human embryonic stem cells in an autologous feeder-free system. Tissue Eng Part C Methods 2011; 17:927-37. [PMID: 21561302 DOI: 10.1089/ten.tec.2010.0735] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Applications of human embryonic stem cells (hESCs) are limited by the use of mouse embryonic fibroblasts feeder and animal-derived components during culture. In this study, we demonstrated the potential use of extracellular matrix (ECM) derived from the autologous feeders to support long-term undifferentiated growth of hESCs in xeno-free, serum-free, and feeder-free conditions. Autologous H9 ebF (feeder cells derived from outgrowth of embryoid body [EB] predifferentiated from H9 hESCs) was derived from EBs predifferentiated from H9 hESCs through a direct-plating outgrowth system. The ECM comprising collagen VI, laminin, and fibronectin was extracted from H9 ebF through a freeze-thaw procedure. The autologous ECM together with animal component-free TeSR™2 medium was used to support long-term growth of H1 and H9 hESC lines for up to 20 passages. The maintenance of hESC undifferentiated state by autologous ECM was confirmed by the positive staining of hESC-specific markers (Oct4, SSEA-4, and Tra-1-60) and the expression of pluripotency marker genes (Oct4, Nanog, and Sox2). Flow cytometry further showed that more than 99% of hESCs retained the expression of SSEA-3/4 after long-term culture on autologous ECM. Pluripotency of hESCs on ECM was further proven by in vitro EB formation and in vivo teratoma assay. Overall, this study suggested a strategy for efficient propagation of clinically compliant hESCs in an autologous feeder-free culture system.
Collapse
Affiliation(s)
- Xin Fu
- Stem Cell Laboratory, Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | | | | | | | | | | |
Collapse
|
25
|
Sneddon SF, DeSousa PA, Arnesen RE, Lieberman BA, Kimber SJ, Brison DR. Gene expression analysis of a new source of human oocytes and embryos for research and human embryonic stem cell derivation. Fertil Steril 2011; 95:1410-5. [DOI: 10.1016/j.fertnstert.2010.08.050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 07/29/2010] [Accepted: 08/20/2010] [Indexed: 10/19/2022]
|
26
|
Higuchi A, Ling QD, Ko YA, Chang Y, Umezawa A. Biomaterials for the feeder-free culture of human embryonic stem cells and induced pluripotent stem cells. Chem Rev 2011; 111:3021-35. [PMID: 21344932 DOI: 10.1021/cr1003612] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, 32001 Taiwan.
| | | | | | | | | |
Collapse
|
27
|
Simple autogeneic feeder cell preparation for pluripotent stem cells. Stem Cell Res 2011; 6:83-9. [DOI: 10.1016/j.scr.2010.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2010] [Revised: 09/21/2010] [Accepted: 09/21/2010] [Indexed: 11/18/2022] Open
|
28
|
Abstract
Human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) reprogrammed from somatic cells can self-renew while maintaining their pluripotency to differentiate into virtually all cell types. In addition to their potential for regenerative medicine, hESCs and iPSCs can also serve as excellent in vitro models for the study of human organogenesis and disease models, as well as drug toxicity screening. MicroRNAs (miRNAs) are nonencoding RNAs of ∼22 nucleotides that function as negative transcriptional regulators via degradation or inhibition by RNA interference (RNAi). MiRNAs play essential roles in developmental pathways. This chapter provides a description of how miRNAs can be introduced into hESCs/iPSCs or their derivatives for experiments via lentivirus-mediated gene transfer.
Collapse
|
29
|
Englund MCO, Sartipy P, Hyllner J. Human Embryonic Stem Cells. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
30
|
Fleischmann G, Müller T, Blasczyk R, Sasaki E, Horn PA. Growth characteristics of the nonhuman primate embryonic stem cell line cjes001 depending on feeder cell treatment. CLONING AND STEM CELLS 2009; 11:225-33. [PMID: 19522672 DOI: 10.1089/clo.2008.0064] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Embryonic stem cells (ESC) hold tremendous potential for therapeutic applications, including regenerative medicine, as well as for understanding basic mechanisms in stem cell biology. Since numerous experiments cannot be conducted in human ESC because of ethical or practical limitations, nonhuman primate ESC serve as invaluable clinically relevant models. The novel marmoset (Callithrix jacchus) ESC line cjes001 was characterized using different stem cell markers. The cells were stained positively with Oct4, SSEA-3, SSEA-4, Tra-1-60, Tra-1-81, and Sox-2 underscoring their status as undifferentiated ESC. ESC are typically grown on mouse embryonic fibroblasts (MEF) as feeder cells whose proliferation is arrested either by treatment with Mitomycin C or by gamma-irradiation. To assess the impact of these treatments on the ability of MEF to support the growth of undifferentiated ESC, we used an MTT assay to evaluate the cellular metabolic activity of growth arrested feeder cells. There was a significant (p < 0.02) difference in gamma-irradiated cells displaying a higher metabolic activity compared to Mitomycin C inactivation. Also we quantified 69 soluble factors in the supernatant of both Mitomycin-treated and gamma-irradiated MEF by bead-based multiplex analysis, and thus established a profile of MEF-secreted factors. The time course of secretion was analyzed by monitoring the supernatant at 0, 6, 12, and 24 h after changing the medium. Comparing gamma-irradiated and Mitomycin-treated MEF suggested higher amounts of some cytokines including FGF or SCF by the former. We also assessed whether the method of inactivation had an effect on growth kinetics and differentiation of primate ESC. There appeared to be a trend to a lower number of differentiated ESC colonies on the gamma-irradiated feeder cells, suggesting that this may be the preferable method of growth arrest.
Collapse
Affiliation(s)
- Gesine Fleischmann
- Institute for Transfusion Medicine, University Hospital Essen , Essen, Germany
| | | | | | | | | |
Collapse
|
31
|
Hakala H, Rajala K, Ojala M, Panula S, Areva S, Kellomäki M, Suuronen R, Skottman H. Comparison of Biomaterials and Extracellular Matrices as a Culture Platform for Multiple, Independently Derived Human Embryonic Stem Cell Lines. Tissue Eng Part A 2009; 15:1775-85. [DOI: 10.1089/ten.tea.2008.0316] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Heidi Hakala
- REGEA, Institute for Regenerative Medicine, University of Tampere, Tampere, Finland
| | - Kristiina Rajala
- REGEA, Institute for Regenerative Medicine, University of Tampere, Tampere, Finland
| | - Marisa Ojala
- REGEA, Institute for Regenerative Medicine, University of Tampere, Tampere, Finland
| | - Sarita Panula
- REGEA, Institute for Regenerative Medicine, University of Tampere, Tampere, Finland
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Palo Alto, California
| | - Sami Areva
- Turku Biomaterials Centre, University of Turku, Turku, Finland
| | - Minna Kellomäki
- Department of Biomedical Engineering, Tampere University of Technology, Tampere, Finland
| | - Riitta Suuronen
- REGEA, Institute for Regenerative Medicine, University of Tampere, Tampere, Finland
- Department of Biomedical Engineering, Tampere University of Technology, Tampere, Finland
- Department of Eye, Ear, and Oral Diseases, Tampere University Hospital, Tampere, Finland
| | - Heli Skottman
- REGEA, Institute for Regenerative Medicine, University of Tampere, Tampere, Finland
| |
Collapse
|
32
|
Desbordes SC, Placantonakis DG, Ciro A, Socci ND, Lee G, Djaballah H, Studer L. High-throughput screening assay for the identification of compounds regulating self-renewal and differentiation in human embryonic stem cells. Cell Stem Cell 2009; 2:602-12. [PMID: 18522853 DOI: 10.1016/j.stem.2008.05.010] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Revised: 04/01/2008] [Accepted: 05/13/2008] [Indexed: 12/24/2022]
Abstract
High-throughput screening (HTS) of chemical libraries has become a critical tool in basic biology and drug discovery. However, its implementation and the adaptation of high-content assays to human embryonic stem cells (hESCs) have been hampered by multiple technical challenges. Here we present a strategy to adapt hESCs to HTS conditions, resulting in an assay suitable for the discovery of small molecules that drive hESC self-renewal or differentiation. Use of this new assay has led to the identification of several marketed drugs and natural compounds promoting short-term hESC maintenance and compounds directing early lineage choice during differentiation. Global gene expression analysis upon drug treatment defines known and novel pathways correlated to hESC self-renewal and differentiation. Our results demonstrate feasibility of hESC-based HTS and enhance the repertoire of chemical compounds for manipulating hESC fate. The availability of high-content assays should accelerate progress in basic and translational hESC biology.
Collapse
Affiliation(s)
- Sabrina C Desbordes
- Developmental Biology Program, Department of Neurosurgery, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Farin A, Liu CY, Elder JB, Langmoen IA, Apuzzo MLJ. The biological restoration of central nervous system architecture and function: part 1-foundations and historical landmarks in contemporary stem cell biology. Neurosurgery 2009; 64:15-39; discussion 34. [PMID: 19145154 DOI: 10.1227/01.neu.0000337580.02706.dc] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Since their discovery, stem cells have fascinated scientists with their ultimate potential: the ability to cure disease, repair altered physiology, and reverse neurological deficit. Stem cell science unquestionably promises to eliminate many of the tragic limitations contemporary medicine must acknowledge, and cloning may provide young cells for an aging population. Although it is widely believed that stem cells will transform the way medicine is practiced, therapeutic interventions using stem cell technology are still in their infancy. The 3 most common stem cell sources studied today are umbilical cord blood, bone marrow, and human embryos. Although cord blood is currently used to treat dozens of disorders and bone marrow stem cells have been used clinically since the 1960s, human embryonic stem cells have yet to be successfully applied to any disease. Undeniably, stem cell therapy has the potential to be one of the most powerful therapeutic options available. In this introductory article of a 5-part series on stem cells, we narrate the evolution of modern stem cell science, delineating major landmarks that will prove responsible for taking stem cell technology from the laboratory into revolutionary clinical applications: from the first milestone of identifying the mouse hematopoietic stem cell to the latest feats of producing pluripotent stem cells without embryos at all. In Part 2, we present the evidence demonstrating the certainty of adult mammalian neurogenesis; in Parts 3 and 4, we describe neurosurgical applications of stem cell technology; and in Part 5, we discuss the philosophical and ethical issues surrounding stem cell therapy, as well as future areas of exploration.
Collapse
Affiliation(s)
- Azadeh Farin
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | | | | | | | | |
Collapse
|
34
|
Zhang F, Pasumarthi KBS. Embryonic stem cell transplantation: promise and progress in the treatment of heart disease. BioDrugs 2009; 22:361-74. [PMID: 18998754 DOI: 10.2165/0063030-200822060-00003] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cardiovascular diseases remain the leading cause of death worldwide, and the burden is equally shared between men and women around the globe. Cardiomyocytes that die in response to disease processes or aging are replaced by scar tissue instead of new muscle cells. Although recent reports suggest an intrinsic capacity for the mammalian myocardium to regenerate via endogenous stem/progenitor cells, the magnitude of such a response appears to be minimal and has yet to be realized fully in cardiovascular patients. Despite the advances in pharmacotherapy and new biomedical technologies, the prognosis for patients diagnosed with end-stage heart failure appears to be grave. While heart transplantation is a viable option, this life-saving intervention suffers from an acute shortage of cardiac organ donors. In view of these existing issues, donor cell transplantation is emerging as a promising strategy to regenerate diseased myocardium. Studies from multiple laboratories have shown that transplantation of donor cells (e.g. fetal cardiomyocytes, skeletal myoblasts, smooth muscle cells, and adult stem cells) can improve the function of diseased hearts over a short period of time (1-4 weeks). While long-term follow-up studies are warranted, it is generally perceived that the beneficial effects of transplanted cells are mainly due to increased angiogenesis or favorable scar remodeling in the engrafted myocardium. Although skeletal myoblasts and bone marrow stem cells hold the highest potential for implementation of autologous therapies, initial results from phase I trials are not promising. In contrast, transplantation of fetal cardiomyocytes has been shown to confer protection against the induction of ventricular tachycardia in experimental myocardial injury models. Furthermore, results from multiple laboratories suggest that fetal cardiomyocytes can couple functionally with host myocytes, stimulate formation of new blood vessels, and improve myocardial function. While it is neither practical nor ethical to test the potential of fetal cardiomyocytes in clinical trials, embryonic stem (ES) cells serve as a novel source for generation of unlimited quantities of cardiomyocytes for myocardial repair. The initial success in the application of ES cells to partially repair and improve myocardial function in experimental models of heart disease has been quite promising. However, multiple hurdles need to be crossed before the potential benefits of ES cells can be translated to the clinic. In this review, we summarize the current knowledge of cardiomyocyte derivation and enrichment from ES-cell cultures and provide a brief survey of factors increasing cardiomyogenic induction in both mouse and human ES cultures. Subsequently, we summarize the current state of research using mouse and human ES cells for the treatment of heart disease in various experimental models. Furthermore, we discuss the challenges that need to be overcome prior to the successful clinical utilization of ES-derived cardiomyocytes for the treatment of end-stage heart disease. While we are optimistic that the researchers in this field will sail across the hurdles, we also suggest that a more cautious approach to the validation of ES cardiomyocytes in experimental models would certainly prevent future disappointments, as seen with skeletal myoblast studies.
Collapse
Affiliation(s)
- Feixiong Zhang
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | |
Collapse
|
35
|
Bosio A, Huppert V, Donath S, Hennemann P, Malchow M, Heinlein UAO. Isolation and enrichment of stem cells. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2009; 114:23-72. [PMID: 19347268 DOI: 10.1007/10_2008_38] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Stem cells have the potential to revolutionize tissue regeneration and engineering. Both general types of stem cells, those with pluripotent differentiation potential as well as those with multipotent differentiation potential, are of equal interest. They are important tools to further understanding of general cellular processes, to refine industrial applications for drug target discovery and predictive toxicology, and to gain more insights into their potential for tissue regeneration. This chapter provides an overview of existing sorting technologies and protocols, outlines the phenotypic characteristics of a number of different stem cells, and summarizes their potential clinical applications.
Collapse
Affiliation(s)
- Andreas Bosio
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429, Bergisch Gladbach, Germany
| | | | | | | | | | | |
Collapse
|
36
|
Schneider A, Spitkovsky D, Riess P, Molcanyi M, Kamisetti N, Maegele M, Hescheler J, Schaefer U. "The good into the pot, the bad into the crop!"--a new technology to free stem cells from feeder cells. PLoS One 2008; 3:e3788. [PMID: 19023443 PMCID: PMC2582950 DOI: 10.1371/journal.pone.0003788] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Accepted: 10/22/2008] [Indexed: 01/11/2023] Open
Abstract
A variety of embryonic and adult stem cell lines require an initial co-culturing with feeder cells for non-differentiated growth, self renewal and maintenance of pluripotency. However for many downstream ES cell applications the feeder cells have to be considered contaminations that might interfere not just with the analysis of experimental data but also with clinical application and tissue engineering approaches. Here we introduce a novel technique that allows for the selection of pure feeder-freed stem cells, following stem cell proliferation on feeder cell layers. Complete and reproducible separation of feeder and embryonic stem cells was accomplished by adaptation of an automated cell selection system that resulted in the aspiration of distinct cell colonies or fraction of colonies according to predefined physical parameters. Analyzing neuronal differentiation we demonstrated feeder-freed stem cells to exhibit differentiation potentials comparable to embryonic stem cells differentiated under standard conditions. However, embryoid body growth as well as differentiation of stem cells into cardiomyocytes was significantly enhanced in feeder-freed cells, indicating a feeder cell dependent modulation of lineage differentiation during early embryoid body development. These findings underline the necessity to separate stem and feeder cells before the initiation of in vitro differentiation. The complete separation of stem and feeder cells by this new technology results in pure stem cell populations for translational approaches. Furthermore, a more detailed analysis of the effect of feeder cells on stem cell differentiation is now possible, that might facilitate the identification and development of new optimized human or genetically modified feeder cell lines.
Collapse
Affiliation(s)
- Annette Schneider
- Institute for Research in Operative Medicine (IFOM), Faculty of Medicine, University of Witten/Herdecke, Witten, Germany
| | - Dimitry Spitkovsky
- Center of Physiology, Institute of Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Peter Riess
- Department of Trauma and Orthopedic Surgery, Faculty of Medicine, University of Witten-Herdecke at the Hospital Merheim, Cologne, Germany
| | - Marek Molcanyi
- Clinic of Neurosurgery and 2nd Department of Neurophysiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Naidu Kamisetti
- Center of Physiology, Institute of Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Marc Maegele
- Department of Trauma and Orthopedic Surgery, Faculty of Medicine, University of Witten-Herdecke at the Hospital Merheim, Cologne, Germany
| | - Jürgen Hescheler
- Center of Physiology, Institute of Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Ute Schaefer
- Department of Experimental Neurotraumatology, Medical University Graz, Graz, Austria
- * E-mail:
| |
Collapse
|
37
|
Enrichment of undifferentiated mouse embryonic stem cells on a culture surface with a glucose-displaying dendrimer. Biomaterials 2008; 29:4236-43. [DOI: 10.1016/j.biomaterials.2008.07.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Accepted: 07/18/2008] [Indexed: 12/20/2022]
|
38
|
Rebuzzini P, Neri T, Zuccotti M, Redi CA, Garagna S. Chromosome number variation in three mouse embryonic stem cell lines during culture. Cytotechnology 2008; 58:17-23. [PMID: 19002773 DOI: 10.1007/s10616-008-9164-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2008] [Accepted: 09/12/2008] [Indexed: 12/30/2022] Open
Abstract
Although mouse embryonic stem cell lines (mESCs) have been established since 1981, systematic studies about chromosomal changes during culture are lacking. In this study, we report the results of a cytogenetic analysis performed on three mESC lines (named UPV02, UPV06 and UPV08) cultured for a period of 3 months. At time intervals, the variation of the chromosome number together with the expression of markers of the undifferentiated status, i.e., OCT-4, SSEA-1, FOM-1 and alkaline phosphatase activity, were determined. The three mESC lines showed a progressive loss of euploid metaphases during the 3 months period of culture. Chromosome abnormalities were accumulated at the latest passages analysed. Metacentric chromosomes were the most frequent chromosome abnormality observed throughout the period of culture. Interestingly, in coincidence with, or few passages after, the drop of euploidy, the alkaline phosphatase activity was partially or totally lost, whereas the OCT-4, SSEA-1 and FOM-1 stem markers were always positive throughout the period of culture. Our results remark the necessity to perform the karyotype analysis during culture in order to develop new culture conditions to maintain the correct chromosome complement in long-term culture of mESC lines.
Collapse
Affiliation(s)
- Paola Rebuzzini
- Dipartimento di Biologia Animale, Laboratorio di Biologia dello Sviluppo, Universita' degli Studi di Pavia, Piazza Botta 9, 27100, Pavia, Italy
| | | | | | | | | |
Collapse
|
39
|
Sheridan SD, Gil S, Wilgo M, Pitt A. Microporous membrane growth substrates for embryonic stem cell culture and differentiation. Methods Cell Biol 2008; 86:29-57. [PMID: 18442643 DOI: 10.1016/s0091-679x(08)00003-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
As the field of embryonic stem cell culture and differentiation advances, many diverse culturing techniques will ultimately be necessary in order to fully reproduce the various environments these cells normally encounter during development. Although most of the work to date has been performed on solid plastic supports, this growth support has several limitations in its representation of the in vivo environment. Impermeable substrates force the cells to exchange their gas and nutrients exclusively through the top side of the cultured cells. In contrast, cells growing in vivo are exposed from several directions to factors from the blood, other cells, soluble factors, and liquid-air interfaces. Additionally, solid plastic presents a smooth two-dimensional surface that is not experienced in vivo. Therefore, the use of traditional plastic presents limitations upon normal cellular morphology, function, and differentiation. An important alternative to growth on solid plastic is the growth of cells on microporous membranes. One of the many advantages to cell growth on porous membrane substrates is their ability to provide a surface that better mimics a three-dimensional in vivo setting. A porous membrane allows multidirectional exposure to nutrients and waste products. In addition, the membrane separation of dual chambers allows for the coculture of cells of different origin to study how cells interact through indirect signaling or through providing a conditioned niche for the proper growth and differentiation of cell types.
Collapse
Affiliation(s)
- Steven D Sheridan
- Millipore Corporation, Bioscience Division, Danvers, Massachusetts 01923, USA
| | | | | | | |
Collapse
|
40
|
Coulombel L, la relecture critique A, John De Vos et Michel Pucéat D. FISH-ESC. Med Sci (Paris) 2008; 24:419-26. [DOI: 10.1051/medsci/2008244419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
41
|
Three-dimensional extracellular matrix-directed cardioprogenitor differentiation: systematic modulation of a synthetic cell-responsive PEG-hydrogel. Biomaterials 2008; 29:2757-66. [PMID: 18396331 DOI: 10.1016/j.biomaterials.2008.03.016] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Accepted: 03/16/2008] [Indexed: 12/31/2022]
Abstract
We show that synthetic three-dimensional (3D) matrix metalloproteinase (MMP)-sensitive poly(ethylene glycol) (PEG)-based hydrogels can direct differentiation of pluripotent cardioprogenitors, using P19 embryonal carcinoma (EC) cells as a model, along a cardiac lineage in vitro. In order to systematically probe 3D matrix effects on P19 EC differentiation, matrix elasticity, MMP-sensitivity and the concentration of a matrix-bound RGDSP peptide were modulated. Soft matrices (E=322+/-64.2 Pa, stoichiometric ratio: 0.8), mimicking the elasticity of embryonic cardiac tissue, increased the fraction of cells expressing the early cardiac transcription factor Nkx2.5 around 2-fold compared to embryoid bodies (EB) in suspension. In contrast, stiffer matrices (E=4,036+/-419.6 Pa, stoichiometric ratio: 1.2) decreased the number of Nkx2.5-positive cells significantly. Further indicators of cardiac maturation were promoted by ligation of integrins relevant in early cardiac development (alpha(5)beta(1,) alpha(v)beta(3)) by the RGDSP ligand in combination with the MMP-sensitivity of the matrix, with a 6-fold increased amount of myosin heavy chain (MHC)-positive cells as compared to EB in suspension. This precisely controlled 3D culture system thus may serve as a potential alternative to natural matrices for engineering cardiac tissue structures for cell culture and potentially therapeutic applications.
Collapse
|
42
|
Li Z, Barron MR, Lough J, Zhao M. Rapid Single-Step Separation of Pluripotent Mouse Embryonic Stem Cells from Mouse Feeder Fibroblasts. Stem Cells Dev 2008; 17:383-7. [DOI: 10.1089/scd.2007.0138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Zhixin Li
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Matthew R. Barron
- Department of Cell Biology, Neurobiology & Anatomyz, Medical College of Wisconsin, Milwaukee, WI 53226
| | - John Lough
- Department of Cell Biology, Neurobiology & Anatomyz, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Ming Zhao
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI 53226
| |
Collapse
|
43
|
Derive and conquer: sourcing and differentiating stem cells for therapeutic applications. Nat Rev Drug Discov 2008; 7:131-42. [PMID: 18079756 DOI: 10.1038/nrd2403] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although great progress has been made in the isolation and culture of stem cells, the future of stem-cell-based therapies and their productive use in drug discovery and regenerative medicine depends on two key factors: finding reliable sources of multipotent and pluripotent cells and the ability to control their differentiation to generate desired derivatives. It is essential for clinical applications to establish reliable sources of pathogen-free human embryonic stem cells (ESCs) and develop suitable differentiation techniques. Here, we address some of the problems associated with the sourcing of human ESCs and discuss the current status of stem-cell differentiation technology.
Collapse
|
44
|
Hunt CJ. The Banking and Cryopreservation of Human Embryonic Stem Cells. Transfus Med Hemother 2007. [DOI: 10.1159/000104458] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
45
|
Yap DYL, Smith DK, Zhang XW, Hill J. Using biomarker signature patterns for an mRNA molecular diagnostic of mouse embryonic stem cell differentiation state. BMC Genomics 2007; 8:210. [PMID: 17605829 PMCID: PMC1931595 DOI: 10.1186/1471-2164-8-210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2006] [Accepted: 07/03/2007] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The pluripotency and self-renewal capabilities, which define the "stemness" state, of mouse embryonic stem (ES) cells, are usually investigated by functional assays or quantitative measurements of the expression levels of known ES cell markers. Strong correlations between these expression levels and functional assays, particularly at the early stage of cell differentiation, have usually not been observed. An effective molecular diagnostic to properly identify the differentiation state of mouse ES cells, prior to further experimentation, is needed. RESULTS A novel molecular pattern recognition procedure has been developed to diagnose the differentiation state of ES cells. This is based on mRNA transcript levels of genes differentially expressed between ES cells and their differentiating progeny. Large publicly available ES cell data sets from various platforms were used to develop and test the diagnostic model. Signature patterns consisting of five gene expression levels achieved high accuracy at determining the cell state (sensitivity and specificity > 97%). CONCLUSION The effective ES cell state diagnostic scheme described here can be implemented easily to assist researchers in identifying the differentiation state of their cultures. It also provides a step towards standardization of experiments relying on cells being in the stem cell or differentiating state.
Collapse
Affiliation(s)
- Daniel YL Yap
- Bioinformatics Institute, 30 Biopolis Street, #07-01, Matrix, 138671, Singapore
| | - David K Smith
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Xue W Zhang
- College of Light Industry and Food Sciences, South China University of Technology, Guangzhou, Guangdong, China
| | - Jeffrey Hill
- Bioinformatics Institute, 30 Biopolis Street, #07-01, Matrix, 138671, Singapore
| |
Collapse
|
46
|
Mannello F, Tonti GA. Concise Review: No Breakthroughs for Human Mesenchymal and Embryonic Stem Cell Culture: Conditioned Medium, Feeder Layer, or Feeder-Free; Medium with Fetal Calf Serum, Human Serum, or Enriched Plasma; Serum-Free, Serum Replacement Nonconditioned Medium, or Ad Hoc Formula? All That Glitters Is Not Gold! Stem Cells 2007; 25:1603-9. [PMID: 17395775 DOI: 10.1634/stemcells.2007-0127] [Citation(s) in RCA: 233] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The choice of an optimal strategy of stem cell culture is at the moment an impossible task, and the elaboration of a culture medium adapted to the production of embryonic and adult mesenchymal stem cells for the clinical application of cell therapy remains a crucial matter. To make an informed choice, it is crucial to not underestimate the theoretical health risk of using xenogenic compounds, to limit the immunological reactions once stem cells are transplanted, to not overestimate the controversial results obtained with human serum, plasma, and blood derivatives, as well as to carefully examine the pros and cons of serum-free and ad hoc formulation strategies; besides that, to also maintain multipotentiality, self-renewal, and transplantability. The extent to which we are able to achieve effective cell therapies will depend on assimilating a rapidly developing base of scientific knowledge with the practical considerations of design, delivery, and host response. Although clinical studies have already started, many questions remain unsolved, and concomitantly even more evidence on suitable and safe off-the-shelf products (mainly xeno-free) for embryonic and mesenchymal stem cells is cropping up, even though there should be no rush to enter the clinical stage while the underlying basic research is still not so solid; this solely will lead to high-quality translational research, without making blunders stemming from the assumption that all that glitters is not gold. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Ferdinando Mannello
- Institute of Histology and Laboratory Analysis, Faculty of Sciences and Technologies, University of Urbino Carlo Bo, Via O. Ubaldini 7, 61029 Urbino (PU), Italy.
| | | |
Collapse
|
47
|
Stephenson EL, Braude PR, Mason C. International community consensus standard for reporting derivation of human embryonic stem cell lines. Regen Med 2007; 2:349-62. [PMID: 17635044 DOI: 10.2217/17460751.2.4.349] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The development and implementation of new methods in human embryonic stem cell (hESC) research is fraught with difficulties, not least of which is the highly variable reporting of the number and quality of embryos used to derive hESC lines. Without a clear minimum information convention among the derivation teams, understanding and comparing derivation methods and their potential impact on the resulting stem cell line will continue to be extremely difficult. In order to address this issue, we consulted international teams regarding the implementation of a minimum information convention for derivation with the aim of universal use, data collection and central analysis, followed by a multi-author publication. The responses demonstrated overwhelming support for such a system; over 90% of the respondents agreed that a universal standard for reporting the derivation of hESC lines was essential as part of the international effort to advance the field efficiently, and over 87% plan to use this standard and share collected data in Spring 2008 for central analysis and public dissemination. A number of future steps are planned in order to ensure that this standard evolves with the field and remains relevant and up-to-date. Our aim is to incorporate these data within existing international initiatives aimed at improving derivation standards. This article is an open-access publication in order to make the convention freely available to the international community and encourage universal participation.
Collapse
Affiliation(s)
- Emma L Stephenson
- Advanced Center for Biochemical Engineering, University College London, London, UK
| | | | | |
Collapse
|