1
|
Nakashima Y, Tsukahara M. Laminin-511 activates the human induced pluripotent stem cell survival via α6β1 integrin-Fyn-RhoA-ROCK signaling. Stem Cells Dev 2022; 31:706-719. [PMID: 35726387 DOI: 10.1089/scd.2022.0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
In human induced pluripotent stem cells (hiPSCs), laminin-511/α6β1 integrin interacts with E-cadherin, an intercellular adhesion molecule, to induce the activation of the PI3K-dependent signaling pathway. The interaction between laminin-511/α6β1 integrin and E-cadherin, an intercellular adhesion molecule, results in protection against apoptosis via the proto-oncogene tyrosine-protein kinase Fyn(Fyn)-RhoA-ROCK signaling pathway and the Ras homolog gene family member A (RhoA)/Rho kinase (ROCK) signaling pathway (the major pathway for cell death). In this paper, the impact of laminin-511 on hiPSC on α6β1 integrin-Fyn-RhoA-ROCK signaling is discussed and explored along with validation experiments. PIK3CA mRNA (mean [standard deviation {SD}]: iMatrix-511, 1.00 [0. 61]; collagen+MFGE8, 0.023 [0.02]; **P<0.01; n=6) and PIK3R1 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 79]; collagen+MFGE8, 0.040 [0.06]; *P<0.05; n=6) were upregulated by iMatrix-511 resulting from an increased expression of Integrin α6 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 42]; collagen+MFGE8, 0.23 [0.05]; **P<0.01; n=6). iMatrix-511 increased the expression of p120-Catenin mRNA (mean [SD]: iMatrix-511, 1.00 [0. 71]; collagen+MFGE8, 0.025 [0.03]; **P<0.01; n=6) and RAC1 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 28]; collagen+MFGE8, 0.39 [0.15]; **P<0.01; n=6) by increasing the expression of E-cadherin mRNA (mean [SD]: iMatrix-511, 1.00 [0. 38]; collagen+MFGE8, 0.16 [0.11]; **P<0.01; n=6). As a result, iMatrix-511 increased the expression of P190 RhoGAP (GTPase-activating proteins) mRNA, such as ARHGAP1 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 57]; collagen+MFGE8, 0.032 [0.03]; **P<0.01; n=6), ARHGAP4 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 56]; collagen+MFGE8, 0.039 [0.049]; **P<0.01; n=6), and ARHGAP5 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 39]; collagen+MFGE8, 0.063 [0.043]; **P<0.01; n=6). Western blotting showed that phospho-Rac1 remained in the cytoplasm and phospho-Fyn showed nuclear transition in iPSCs cultured on iMatrix-511. Proteome analysis showed that PI3K signaling was enhanced and cytoskeletal actin was activated in iPSCs cultured on iMatrix-511. In conclusion, laminin-511 strongly activated the cell survival by promoting α6β1 integrin-Fyn-RhoA-ROCK signaling in hiPSCs.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto Research Park KISTIC Building Room 501, 5th floor, KISTIC building,, Kyoto Research Park KISTIC Building Room 501, 5th floor, KISTIC building, 134 Chudoji Minami-cho,, Shimogyo-ku,, Kyoto, Kyoto, Japan, 600-8813;
| | - Masayoshi Tsukahara
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), kyoto, Kyoto, Japan;
| |
Collapse
|
2
|
Ornelas-González A, González-González M, Rito-Palomares M. Microcarrier-based stem cell bioprocessing: GMP-grade culture challenges and future trends for regenerative medicine. Crit Rev Biotechnol 2021; 41:1081-1095. [PMID: 33730936 DOI: 10.1080/07388551.2021.1898328] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Recently, stem cell-based therapies have been proposed as an alternative for the treatment of many diseases. Stem cells (SCs) are well known for their capacity to preserve themselves, proliferate, and differentiate into multiple lineages. These characteristics allow stem cells to be a viable option for the treatment of diverse diseases. Traditional methodologies based on 2-dimensional culture techniques (T-flasks and Petri dishes) are simple and well standardized; however, they present disadvantages that limit the production of the cell yield required for regenerative medicine applications. Lately, microcarrier (MC)-based culture techniques have emerged as an attractive platform for expanding stem cells in suspension systems. Although the use of stem cell expansion on MCs has recently shown significant increase, their implementation for medical purposes is been hampered by bottlenecks in upstream and downstream processing. Therefore, there is an urgent need in the development of bioprocesses that simplify stem cell cultures under xeno-free conditions and detachment from MCs without diminishing their pluripotency and viability. A critical analysis of the factors that impact the up and downstream bioprocessing on MC-based stem cell cultures is presented in this review. This analysis aims to raise the awareness of the current drawbacks that limit MC-based stem cell bioprocessing in regenerative medicine and propose alternatives to overcome them.
Collapse
Affiliation(s)
| | | | - Marco Rito-Palomares
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Mexico
| |
Collapse
|
3
|
Hanna M, Sahito RGA, Rateb M, Kachiwal AB, Seddiek HA, Bhutto B, Hescheler J. Generation of transgene-free induced pluripotent stem cells from cardiac fibroblasts of goat embryos. J Stem Cells Regen Med 2020; 16:34-43. [PMID: 33414579 DOI: 10.46582/jsrm.1602007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/11/2020] [Indexed: 11/19/2022]
Abstract
Induced pluripotent stem cells (iPSCs) hold a great potential for therapeutic regenerative medicine. The aim of this study was to generate induced pluripotent stem cells from goat embryonic cardiac tissue derived fibroblasts. The isolated cardiac fibroblasts from the cardiac tissue of goat embryos were positive for alfa smooth muscle actin, vimentin and discoidin domain receptor2. From these cells, we generated transgene free iPSCs using piggyBac transposons / transposase using five transcription factors (Oct4, Sox2, Klf, Myc and Lin 28). The generated iPSCs were SSEA1, SSEA4 and Oct4 positive. They were cultured on neofeeders using 20% Serum replacement - IMDM with bFGF. They could form cystic and compact embryoid bodies that showed differentiated ectodermal and mesodermal like cells when cultured using 20% FBS-IMDM without bFGF. The iPSCs, generated in the frame of this approach were produced without the use of integrating virus and the reprogramming transgenes were removed at the end of the process. Though there were limitations in the approach used, a substantial sign of reprogramming was obtained.
Collapse
Affiliation(s)
- Mira Hanna
- Institute of Neurophysiology, University of Cologne, Robert-Koch-Strasse 39, 50931 Cologne, Germany.,Department of physiology, Faculty of medicine (Kasr El-Aini) Cairo University, El-Maniel, Cairo 11451, Egypt
| | | | - Moshira Rateb
- Department of physiology, Faculty of medicine (Kasr El-Aini) Cairo University, El-Maniel, Cairo 11451, Egypt
| | - Allah Bux Kachiwal
- Department of Veterinary Physiology and Biochemistry, Sindh Agriculture University Tandojam, Pakistan
| | - Hanan A Seddiek
- Department of physiology, Faculty of medicine (Kasr El-Aini) Cairo University, El-Maniel, Cairo 11451, Egypt
| | - Bachal Bhutto
- Department of Veterinary Parasitology, Sindh Agriculture University Tandojam, Pakistan
| | - Jürgen Hescheler
- Institute of Neurophysiology, University of Cologne, Robert-Koch-Strasse 39, 50931 Cologne, Germany
| |
Collapse
|
4
|
Välimäki H, Hyvärinen T, Leivo J, Iftikhar H, Pekkanen-Mattila M, Rajan DK, Verho J, Kreutzer J, Ryynänen T, Pirhonen J, Aalto-Setälä K, Kallio P, Narkilahti S, Lekkala J. Covalent immobilization of luminescent oxygen indicators reduces cytotoxicity. Biomed Microdevices 2020; 22:41. [PMID: 32494857 PMCID: PMC7270993 DOI: 10.1007/s10544-020-00495-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Luminescence-based oxygen sensing is a widely used tool in cell culture applications. In a typical configuration, the luminescent oxygen indicators are embedded in a solid, oxygen-permeable matrix in contact with the culture medium. However, in sensitive cell cultures even minimal leaching of the potentially cytotoxic indicators can become an issue. One way to prevent the leaching is to immobilize the indicators covalently into the supporting matrix. In this paper, we report on a method where platinum(II)-5,10,15,20-tetrakis-(2,3,4,5,6-pentafluorphenyl)-porphyrin (PtTFPP) oxygen indicators are covalently immobilized into a polymer matrix consisting of polystyrene and poly(pentafluorostyrene). We study how the covalent immobilization influences the sensing material’s cytotoxicity to human induced pluripotent stem cell-derived (hiPSC-derived) neurons and cardiomyocytes (CMs) through 7–13 days culturing experiments and various viability analyses. Furthermore, we study the effect of the covalent immobilization on the indicator leaching and the oxygen sensing properties of the material. In addition, we demonstrate the use of the covalently linked oxygen sensing material in real time oxygen tension monitoring in functional hypoxia studies of the hiPSC-derived CMs. The results show that the covalently immobilized indicators substantially reduce indicator leaching and the cytotoxicity of the oxygen sensing material, while the influence on the oxygen sensing properties remains small or nonexistent.
Collapse
Affiliation(s)
- Hannu Välimäki
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, 33720, Tampere, Finland.
| | - Tanja Hyvärinen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Joni Leivo
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, 33720, Tampere, Finland
| | - Haider Iftikhar
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, 33720, Tampere, Finland
| | - Mari Pekkanen-Mattila
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | | | - Jarmo Verho
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, 33720, Tampere, Finland
| | - Joose Kreutzer
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, 33720, Tampere, Finland
| | - Tomi Ryynänen
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, 33720, Tampere, Finland
| | - Jonatan Pirhonen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Katriina Aalto-Setälä
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Pasi Kallio
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, 33720, Tampere, Finland
| | - Susanna Narkilahti
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Jukka Lekkala
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, 33720, Tampere, Finland
| |
Collapse
|
5
|
Bovine serum albumin affects N-glycoforms of murine IgG monoclonal antibody purified from hybridoma supernatants. Appl Microbiol Biotechnol 2020; 104:1583-1594. [PMID: 31915902 DOI: 10.1007/s00253-019-10309-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/25/2019] [Accepted: 12/08/2019] [Indexed: 12/11/2022]
Abstract
Immunoglobulin G (IgG) is a class of monoclonal antibodies (mAbs) commonly produced in mammalian cell lines. These cell lines are grown in finely adjusted culture media, which contain components that may impact glycoforms. As variation of N-glycoforms can impact the biological properties of IgGs, medium composition should be controlled. Here, we studied the effects on IgG N-glycoforms of different components in hybridoma culture media, specifically compared bovine serum albumin (BSA) with other small molecules, using a matrix-assisted laser desorption/ionization quadrupole ion trap time-of-flight multistage mass spectrometry (MALDI-QIT-TOF MSn)-based approach. We show that small molecular additives caused little change in glycan species, though a number of these reagents, especially glutamine, affected levels of glycosylation. In comparison, the addition of macromolecular protein BSA significantly changed IgG N-glycan patterns, not only in species but also in glycosylation levels. Together, our finding suggests that BSA increases the complexity of IgG N-glycoforms, thus raising the difficulty in maintaining glycoforms consistency during antibody production. Therefore, the effect of BSA on IgG N-glycans should be considered when designing optimal medium formulations for IgG production. KEY POINTS: • Small molecular medium additives only affect glycosylation levels of IgG N-glycans. • BSA significantly changes IgG N-glycoforms as a medium additive. • BSA's skewing of IgG N-glycoforms should be considered in IgG production.
Collapse
|
6
|
Hyvärinen T, Hyysalo A, Kapucu FE, Aarnos L, Vinogradov A, Eglen SJ, Ylä-Outinen L, Narkilahti S. Functional characterization of human pluripotent stem cell-derived cortical networks differentiated on laminin-521 substrate: comparison to rat cortical cultures. Sci Rep 2019; 9:17125. [PMID: 31748598 PMCID: PMC6868015 DOI: 10.1038/s41598-019-53647-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/01/2019] [Indexed: 12/15/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-derived neurons provide exciting opportunities for in vitro modeling of neurological diseases and for advancing drug development and neurotoxicological studies. However, generating electrophysiologically mature neuronal networks from hPSCs has been challenging. Here, we report the differentiation of functionally active hPSC-derived cortical networks on defined laminin-521 substrate. We apply microelectrode array (MEA) measurements to assess network events and compare the activity development of hPSC-derived networks to that of widely used rat embryonic cortical cultures. In both of these networks, activity developed through a similar sequence of stages and time frames; however, the hPSC-derived networks showed unique patterns of bursting activity. The hPSC-derived networks developed synchronous activity, which involved glutamatergic and GABAergic inputs, recapitulating the classical cortical activity also observed in rodent counterparts. Principal component analysis (PCA) based on spike rates, network synchronization and burst features revealed the segregation of hPSC-derived and rat network recordings into different clusters, reflecting the species-specific and maturation state differences between the two networks. Overall, hPSC-derived neural cultures produced with a defined protocol generate cortical type network activity, which validates their applicability as a human-specific model for pharmacological studies and modeling network dysfunctions.
Collapse
Affiliation(s)
- Tanja Hyvärinen
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere, Finland
| | - Anu Hyysalo
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere, Finland
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Fikret Emre Kapucu
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Laura Aarnos
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere, Finland
| | - Andrey Vinogradov
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere, Finland
| | - Stephen J Eglen
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, United Kingdom
| | - Laura Ylä-Outinen
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere, Finland
| | - Susanna Narkilahti
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere, Finland.
| |
Collapse
|
7
|
Doornaert M, De Maere E, Colle J, Declercq H, Taminau J, Lemeire K, Berx G, Blondeel P. Xenogen-free isolation and culture of human adipose mesenchymal stem cells. Stem Cell Res 2019; 40:101532. [PMID: 31421383 DOI: 10.1016/j.scr.2019.101532] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 07/22/2019] [Accepted: 08/07/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Adipose-derived Stem Cells (ASCs) present great potential for reconstructive procedures. Currently, isolation by enzyme digestion and culturing using xenogenic substances remain the gold standard, impairing clinical use. METHODS Abdominal lipo-aspirate and blood samples were obtained from healthy patients. A novel mechanical isolation method for ASCs was compared to (the standard) collagenase digestion. ASCs are examined by flowcytometry and multilineage differentiation assays. Cell cultures were performed without xenogenic or toxic substances, using autologous plasma extracted from peripheral blood. After eGFP-transfection, an in vivo differentiation assay was performed. RESULTS Mechanical isolation is more successful in isolating CD34+/CD31-/CD45-/CD13+/CD73+/CD146- ASCs from lipo-aspirate than isolation via collagenase digestion (p <0 .05). ASCs display multilineage differentiation potential in vitro. Autologous plasma is a valid additive for ASCs culturing. eGFP-ASCs, retrieved after 3 months in vivo, differentiated in adipocytes and endothelial cells. CONCLUSION A practical method for human ASC isolation and culturing from abdominal lipo-aspirate, without the addition of xenogenic substances, is described. The mechanical protocol is more successful than the current gold standard protocol of enzyme digestion. These results are important in the translation of laboratory-based cell cultures to clinical reconstructive and aesthetic applications.
Collapse
Affiliation(s)
- M Doornaert
- Department of Plastic and Reconstructive Surgery, Gent University Hospital, Gent, Belgium.
| | - E De Maere
- Department of Plastic and Reconstructive Surgery, Gent University Hospital, Gent, Belgium.
| | - J Colle
- Department of Plastic and Reconstructive Surgery, Gent University Hospital, Gent, Belgium.
| | - H Declercq
- Department of Basic Medical Sciences, UGent, Gent, Belgium.
| | - J Taminau
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Vlaams Instituut voor Biotechnologie (VIB), Gent, Belgium; Cancer Research Institute Gent (CRIG), Belgium.
| | - K Lemeire
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Vlaams Instituut voor Biotechnologie (VIB), Gent, Belgium; Inflammation Research Centre (IRC), VIB, Gent, Belgium.
| | - G Berx
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Vlaams Instituut voor Biotechnologie (VIB), Gent, Belgium; Inflammation Research Centre (IRC), VIB, Gent, Belgium.
| | - Ph Blondeel
- Department of Plastic and Reconstructive Surgery, Gent University Hospital, Gent, Belgium.
| |
Collapse
|
8
|
Dakhore S, Nayer B, Hasegawa K. Human Pluripotent Stem Cell Culture: Current Status, Challenges, and Advancement. Stem Cells Int 2018; 2018:7396905. [PMID: 30595701 PMCID: PMC6282144 DOI: 10.1155/2018/7396905] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/23/2022] Open
Abstract
Over the past two decades, human embryonic stem cells (hESCs) have gained attention due to their pluripotent and proliferative ability which enables production of almost all cell types in the human body in vitro and makes them an excellent tool to study human embryogenesis and disease, as well as for drug discovery and cell transplantation therapies. Discovery of human-induced pluripotent stem cells (hiPSCs) further expanded therapeutic applications of human pluripotent stem cells (PSCs). hPSCs provide a stable and unlimited original cell source for producing suitable cells and tissues for downstream applications. Therefore, engineering the environment in which these cells are grown, for stable and quality-controlled hPSC maintenance and production, is one of the key factors governing the success of these applications. hPSCs are maintained in a particular niche using specific cell culture components. Ideally, the culture should be free of xenobiotic components to render hPSCs suitable for therapeutic applications. Substantial efforts have been put to identify effective components, and develop culture conditions and protocols, for their large-scale expansion without compromising on quality. In this review, we discuss different media, their components and functions, including specific requirements to maintain the pluripotent and proliferative ability of hPSCs. Understanding the role of culture components would enable the development of appropriate conditions to promote large-scale, quality-controlled expansion of hPSCs thereby increasing their potential applications.
Collapse
Affiliation(s)
- Sushrut Dakhore
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences (NCBS), Bangalore, India
| | - Bhavana Nayer
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences (NCBS), Bangalore, India
| | - Kouichi Hasegawa
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences (NCBS), Bangalore, India
- Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advanced Study, Kyoto University, Japan
| |
Collapse
|
9
|
Karvinen J, Joki T, Ylä-Outinen L, Koivisto JT, Narkilahti S, Kellomäki M. Soft hydrazone crosslinked hyaluronan- and alginate-based hydrogels as 3D supportive matrices for human pluripotent stem cell-derived neuronal cells. REACT FUNCT POLYM 2018. [DOI: 10.1016/j.reactfunctpolym.2017.12.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
10
|
Effect of prolonged differentiation on functional maturation of human pluripotent stem cell-derived neuronal cultures. Stem Cell Res 2018; 27:151-161. [PMID: 29414606 DOI: 10.1016/j.scr.2018.01.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/09/2018] [Accepted: 01/17/2018] [Indexed: 01/15/2023] Open
Abstract
Long-term neural differentiation of human pluripotent stem cells (hPSCs) is associated with enhanced neuronal maturation, which is a necessity for creation of representative in vitro models. It also induces neurogenic-to-gliogenic fate switch, increasing proportion of endogenous astrocytes formed from the common neural progenitors. However, the significance of prolonged differentiation on the neural cell type composition and functional development of hPSC-derived neuronal cells has not been well characterized. Here, we studied two hPSC lines, both of which initially showed good neuronal differentiation capacity. However, the propensity for endogenous astrogenesis and maturation state after extended differentiation varied. Live cell calcium imaging revealed that prolonged differentiation facilitated maturation of GABAergic signaling. According to extracellular recordings with microelectrode array (MEA), neuronal activity was limited to fewer areas of the culture, which expressed more frequent burst activity. Efficient maturation after prolonged differentiation also promoted organization of spontaneous activity by burst compaction. These results suggest that although prolonged neural differentiation can be challenging, it has beneficial effect on functional maturation, which can also improve transition to different neural in vitro models and applications.
Collapse
|
11
|
Castells-Sala C, Martorell J, Balcells M. A human plasma derived supplement preserves function of human vascular cells in absence of fetal bovine serum. Cell Biosci 2017; 7:41. [PMID: 28811873 PMCID: PMC5554976 DOI: 10.1186/s13578-017-0164-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/24/2017] [Indexed: 11/10/2022] Open
Abstract
Current techniques for cell culture routinely use animal-derived components. Fetal bovine serum (FBS) is the most widely applied supplement, but it often displays significant batch-to-batch variations and is generally not suitable for clinical applications in humans. A robust and xeno-free alternative to FBS is of high interest for cellular therapies, from early in vitro testing to clinical trials in human subjects. In the current work, a highly consistent human plasma derived supplement (SCC) has been tested, as a potential substitute of FBS in primary human vascular cells culture. Our results show that SCC is able to support proliferation, preserve cellular morphology and potentiate functionality analogously to FBS. We conclude that SCC is a viable substitute of FBS for culture and expansion of cells in advanced therapies using human vascular cells and fibroblasts.
Collapse
Affiliation(s)
- C Castells-Sala
- IQS School of Engineering, Universitat Ramon Llull, Barcelona, Spain
| | - J Martorell
- IQS School of Engineering, Universitat Ramon Llull, Barcelona, Spain
| | - M Balcells
- IQS School of Engineering, Universitat Ramon Llull, Barcelona, Spain.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA USA
| |
Collapse
|
12
|
Baek JA, Seol HW, Jung J, Kim HS, Oh SK, Choi YM. Clean-Up Human Embryonic Stem Cell Lines Using Humanized Culture Condition. Tissue Eng Regen Med 2017; 14:453-464. [PMID: 30603501 DOI: 10.1007/s13770-017-0053-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 03/19/2017] [Accepted: 03/21/2017] [Indexed: 11/28/2022] Open
Abstract
Human embryonic stem cell (hESC) culture system has been changing culture conditions from conventional to xeno-free for therapeutic cell applications, and N-glycolylneuraminic acid (Neu5Gc) could be a useful indicator of xenogeneic contaminations in hESCs because human cells can no longer produce it genetically. We set up the humanized culture condition using commercially available humanized materials and two different adaptation methods: sequential or direct. SNUhES4 and H1 hESC lines, previously established in conventional culture conditions, were maintained using the humanized culture condition and were examined for the presence of Neu5Gc. The hESCs showed the same morphology and character as those of the conventional culture condition. Moreover, they were negative for Neu5Gc within two passages without loss of pluripotency. This study suggested that this method can effectively cleanse previously established hESC lines, bringing them one step closer to being clinical-grade hESCs.
Collapse
Affiliation(s)
- Jin Ah Baek
- 1Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, 71, Ihwajang-gil, Jongno-gu, Seoul, 03087 Korea
| | - Hye Won Seol
- 1Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, 71, Ihwajang-gil, Jongno-gu, Seoul, 03087 Korea
| | - Juwon Jung
- 1Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, 71, Ihwajang-gil, Jongno-gu, Seoul, 03087 Korea
| | - Hee Sun Kim
- 1Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, 71, Ihwajang-gil, Jongno-gu, Seoul, 03087 Korea.,2Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, 101, Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Sun Kyung Oh
- 1Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, 71, Ihwajang-gil, Jongno-gu, Seoul, 03087 Korea
| | - Young Min Choi
- 1Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, 71, Ihwajang-gil, Jongno-gu, Seoul, 03087 Korea.,2Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, 101, Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| |
Collapse
|
13
|
Houdelet M, Galinski A, Holland T, Wenzel K, Schillberg S, Buyel JF. Animal component-free Agrobacterium tumefaciens cultivation media for better GMP-compliance increases biomass yield and pharmaceutical protein expression in Nicotiana benthamiana. Biotechnol J 2017; 12. [PMID: 28221723 DOI: 10.1002/biot.201600721] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 02/14/2017] [Accepted: 02/20/2017] [Indexed: 12/16/2023]
Abstract
Transient expression systems allow the rapid production of recombinant proteins in plants. Such systems can be scaled up to several hundred kilograms of biomass, making them suitable for the production of pharmaceutical proteins required at short notice, such as emergency vaccines. However, large-scale transient expression requires the production of recombinant Agrobacterium tumefaciens strains with the capacity for efficient gene transfer to plant cells. The complex media often used for the cultivation of this species typically include animal-derived ingredients that can contain human pathogens, thus conflicting with the requirements of good manufacturing practice (GMP). We replaced all the animal-derived components in yeast extract broth (YEB) cultivation medium with soybean peptone, and then used a design-of-experiments approach to optimize the medium composition, increasing the biomass yield while maintaining high levels of transient expression in subsequent infiltration experiments. The resulting plant peptone Agrobacterium medium (PAM) achieved a two-fold increase in OD600 compared to YEB medium during a 4-L batch fermentation lasting 18 h. Furthermore, the yields of the monoclonal antibody 2G12 and the fluorescent protein DsRed were maintained when the cells were cultivated in PAM rather than YEB. We have thus demonstrated a simple, efficient and scalable method for medium optimization that reduces process time and costs. The final optimized medium for the cultivation of A. tumefaciens completely lacks animal-derived components, thus facilitating the GMP-compliant large-scale transient expression of recombinant proteins in plants.
Collapse
Affiliation(s)
- Marcel Houdelet
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | - Anna Galinski
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | - Tanja Holland
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | - Kathrin Wenzel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | - Stefan Schillberg
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
- Institute for Phytopathology and Applied Zoology, Phytopathology Department, Justus-Liebig University Giessen, Giessen, Germany
| | - Johannes Felix Buyel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
- Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
14
|
Hyysalo A, Ristola M, Joki T, Honkanen M, Vippola M, Narkilahti S. Aligned Poly(ε-caprolactone) Nanofibers Guide the Orientation and Migration of Human Pluripotent Stem Cell-Derived Neurons, Astrocytes, and Oligodendrocyte Precursor Cells In Vitro. Macromol Biosci 2017; 17. [DOI: 10.1002/mabi.201600517] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/06/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Anu Hyysalo
- NeuroGroup; BioMediTech and Faculty of Medicine and Life Sciences; University of Tampere; Lääkärinkatu 1 FI-33520 Tampere Finland
| | - Mervi Ristola
- NeuroGroup; BioMediTech and Faculty of Medicine and Life Sciences; University of Tampere; Lääkärinkatu 1 FI-33520 Tampere Finland
| | - Tiina Joki
- NeuroGroup; BioMediTech and Faculty of Medicine and Life Sciences; University of Tampere; Lääkärinkatu 1 FI-33520 Tampere Finland
| | - Mari Honkanen
- Department of Materials Science; Tampere University of Technology; Korkeakoulunkatu 6 FI-33720 Tampere Finland
| | - Minnamari Vippola
- Department of Materials Science; Tampere University of Technology; Korkeakoulunkatu 6 FI-33720 Tampere Finland
| | - Susanna Narkilahti
- NeuroGroup; BioMediTech and Faculty of Medicine and Life Sciences; University of Tampere; Lääkärinkatu 1 FI-33520 Tampere Finland
| |
Collapse
|
15
|
Expandable and Rapidly Differentiating Human Induced Neural Stem Cell Lines for Multiple Tissue Engineering Applications. Stem Cell Reports 2016; 7:557-570. [PMID: 27569063 PMCID: PMC5032182 DOI: 10.1016/j.stemcr.2016.07.017] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/22/2016] [Accepted: 07/25/2016] [Indexed: 12/26/2022] Open
Abstract
Limited availability of human neurons poses a significant barrier to progress in biological and preclinical studies of the human nervous system. Current stem cell-based approaches of neuron generation are still hindered by prolonged culture requirements, protocol complexity, and variability in neuronal differentiation. Here we establish stable human induced neural stem cell (hiNSC) lines through the direct reprogramming of neonatal fibroblasts and adult adipose-derived stem cells. These hiNSCs can be passaged indefinitely and cryopreserved as colonies. Independently of media composition, hiNSCs robustly differentiate into TUJ1-positive neurons within 4 days, making them ideal for innervated co-cultures. In vivo, hiNSCs migrate, engraft, and contribute to both central and peripheral nervous systems. Lastly, we demonstrate utility of hiNSCs in a 3D human brain model. This method provides a valuable interdisciplinary tool that could be used to develop drug screening applications as well as patient-specific disease models related to disorders of innervation and the brain. Human induced neural stem cell (hiNSC) lines can be passaged and cryopreserved Rapid and robust media-independent differentiation in as few as 4 days hiNSCs contribute to both central and peripheral nervous systems in vivo Demonstration of utility in innervated muscle co-culture and 3D human brain model
Collapse
|
16
|
Zhang D, Mai Q, Li T, Huang J, Ding C, Jia M, Zhou C, Xu Y. Comparison of a xeno-free and serum-free culture system for human embryonic stem cells with conventional culture systems. Stem Cell Res Ther 2016; 7:101. [PMID: 27474011 PMCID: PMC4967296 DOI: 10.1186/s13287-016-0347-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 05/14/2016] [Accepted: 06/10/2016] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Elimination of all animal components during derivation and long-term culture of human embryonic stem cells (hESCs) is necessary for future applications of hESCs in clinical cell therapy. METHODS In this study, we established the culture system of xeno-free human foreskin fibroblast feeders (XF-HFF) in combination with chemically defined medium (CDM). XF-HFF/CDM was compared with several conventional culture systems. The hESCs cultured in different media were further characterized through karyotype analysis, pluripotency gene expression, and cell differentiation ability. RESULTS The hESCs in the XF-HFF/CDM maintained their characteristics including typical morphology and stable karyotype. In addition, hESCs were characterized by fluorescent immunostaining of pluripotent markers and teratoma formation in vivo. RT-PCR analysis shown that the stem cell markers OCT3/4, hTERT, SOX2, and Nanog were present in the cell line hESC-1 grown on XF-HFF/CDM. Furthermore, the results of cell growth and expression of bFGF, Oct-4, and hTERT indicated that XF-HFF/CDM had better performance than human serum-matrix/CDM and XF-HFF/human serum. CONCLUSION The comparison of different xeno-free culture conditions will facilitate clarifying the key features of self-renewal, pluripotency, and derivation and will shed light on clinic applications of hESCs.
Collapse
Affiliation(s)
- Dan Zhang
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Qingyun Mai
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Tao Li
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Jia Huang
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Chenhui Ding
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Mengxi Jia
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Canquan Zhou
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, People's Republic of China.
| | - Yanwen Xu
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, Guangdong, 510080, People's Republic of China.
| |
Collapse
|
17
|
Nakashima Y, Omasa T. What Kind of Signaling Maintains Pluripotency and Viability in Human-Induced Pluripotent Stem Cells Cultured on Laminin-511 with Serum-Free Medium? Biores Open Access 2016; 5:84-93. [PMID: 27096107 PMCID: PMC4834485 DOI: 10.1089/biores.2016.0001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Xeno-free medium contains no animal-derived components, but is composed of minimal growth factors and is serum free; the medium may be supplemented with insulin, transferrin, and selenium (ITS medium). Serum-free and xeno-free culture of human-induced pluripotent stem cells (hiPSCs) uses a variety of components based on ITS medium and Dulbecco's modified Eagle's medium/Ham's nutrient mixture F12 (DMEM/F12) that contain high levels of iron salt and glucose. Culture of hiPSCs also requires scaffolding materials, such as extracellular matrix, collagen, fibronectin, laminin, proteoglycan, and vitronectin. The scaffolding component laminin-511, which is composed of α5, β1, and γ1 chains, binds to α3β1, α6β1, and α6β4 integrins on the cell membrane to induce activation of the PI3K/AKT- and Ras/MAPK-dependent signaling pathways. In hiPSCs, the interaction of laminin-511/α6β1 integrin with the cell–cell adhesion molecule E-cadherin confers protection against apoptosis through the Ras homolog gene family member A (RhoA)/Rho kinase (ROCK) signaling pathway (the major pathways for cell death) and the proto-oncogene tyrosine-protein kinase Fyn (Fyn)-RhoA-ROCK signaling pathway. The expression levels of α6β1 integrin and E-cadherin on cell membranes are controlled through the activation of insulin receptor/insulin, FGF receptor/FGF2, or activin-like kinase 5 (ALK5)-dependent TGF-β signaling. A combination of growth factors, medium constituents, cell membrane-located E-cadherin, and α6β1 integrin-induced signaling is required for pluripotent cell proliferation and for optimal cell survival on a laminin-511 scaffold. In this review, we discuss and explore the influence of growth factors on the cadherin and integrin signaling pathways in serum-free and xeno-free cultures of hiPSCs during the preparation of products for regenerative medicinal therapies. In addition, we suggest the optimum serum-free medium components for use with laminin-511, a new scaffold for hiPSC culture.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- Department of Material and Life Science, Graduate School of Engineering, Osaka University , Osaka, Japan
| | - Takeshi Omasa
- Department of Material and Life Science, Graduate School of Engineering, Osaka University , Osaka, Japan
| |
Collapse
|
18
|
Lee JB, Graham M, Collins TJ, Lee JH, Hong SH, Mcnicol AJ, Shapovalova Z, Bhatia M. Reversible lineage-specific priming of human embryonic stem cells can be exploited to optimize the yield of differentiated cells. Stem Cells 2016; 33:1142-52. [PMID: 25639500 PMCID: PMC4413029 DOI: 10.1002/stem.1952] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 11/09/2014] [Accepted: 11/21/2014] [Indexed: 12/17/2022]
Abstract
The clinical use of human embryonic stem cells (hESCs) requires efficient cellular expansion that must be paired with an ability to generate specialized progeny through differentiation. Self-renewal and differentiation are deemed inherent hallmarks of hESCs and a growing body of evidence suggests that initial culture conditions dictate these two aspects of hESC behavior. Here, we reveal that defined culture conditions using commercial mTeSR1 media augment the expansion of hESCs and enhance their capacity for neural differentiation at the expense of hematopoietic lineage competency without affecting pluripotency. This culture-induced modification was shown to be reversible, as culture in mouse embryonic fibroblast-conditioned media (MEF-CM) in subsequent passages allowed mTeSR1-expanded hESCs to re-establish hematopoietic differentiation potential. Optimal yield of hematopoietic cells can be achieved by expansion in mTeSR1 followed by a recovery period in MEF-CM. Furthermore, the lineage propensity to hematopoietic and neural cell types could be predicted via analysis of surrogate markers expressed by hESCs cultured in mTeSR1 versus MEF-CM, thereby circumventing laborious in vitro differentiation assays. Our study reveals that hESCs exist in a range of functional states and balance expansion with differentiation potential, which can be modulated by culture conditions in a predictive and quantitative manner.
Collapse
Affiliation(s)
- Jung Bok Lee
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Production of human pluripotent stem cell therapeutics under defined xeno-free conditions: progress and challenges. Stem Cell Rev Rep 2015; 11:96-109. [PMID: 25077810 DOI: 10.1007/s12015-014-9544-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Recent advances on human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) have brought us closer to the realization of their clinical potential. Nonetheless, tissue engineering and regenerative medicine applications will require the generation of hPSC products well beyond the laboratory scale. This also mandates the production of hPSC therapeutics in fully-defined, xeno-free systems and in a reproducible manner. Toward this goal, we summarize current developments in defined media free of animal-derived components for hPSC culture. Bioinspired and synthetic extracellular matrices for the attachment, growth and differentiation of hPSCs are also reviewed. Given that most progress in xeno-free medium and substrate development has been demonstrated in two-dimensional rather than three dimensional culture systems, translation from the former to the latter poses unique difficulties. These challenges are discussed in the context of cultivation platforms of hPSCs as aggregates, on microcarriers or after encapsulation in biocompatible scaffolds.
Collapse
|
20
|
Saitoh I, Inada E, Iwase Y, Noguchi H, Murakami T, Soda M, Kubota N, Hasegawa H, Akasaka E, Matsumoto Y, Oka K, Yamasaki Y, Hayasaki H, Sato M. Choice of Feeders Is Important When First Establishing iPSCs Derived From Primarily Cultured Human Deciduous Tooth Dental Pulp Cells. CELL MEDICINE 2015; 8:9-23. [PMID: 26858904 DOI: 10.3727/215517915x689038] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Feeder cells are generally required to maintain embryonic stem cells (ESCs)/induced pluripotent stem cells (iPSCs). Mouse embryonic fibroblasts (MEFs) isolated from fetuses and STO mouse stromal cell line are the most widely used feeder cells. The aim of this study was to determine which cells are suitable for establishing iPSCs from human deciduous tooth dental pulp cells (HDDPCs). Primary cultures of HDDPCs were cotransfected with three plasmids containing human OCT3/4, SOX2/KLF4, or LMYC/LIN28 and pmaxGFP by using a novel electroporation method, and then cultured in an ESC qualified medium for 15 days. Emerging colonies were reseeded onto mitomycin C-treated MEFs or STO cells. The colonies were serially passaged for up to 26 passages. During this period, colony morphology was assessed to determine whether cells exhibited ESC-like morphology and alkaline phosphatase activity to evaluate the state of cellular reprogramming. HDDPCs maintained on MEFs were successfully reprogrammed into iPSCs, whereas those maintained on STO cells were not. Once established, the iPSCs were maintained on STO cells without loss of pluripotency. Our results indicate that MEFs are better feeder cells than STO cells for establishing iPSCs. Feeder choice is a key factor enabling efficient generation of iPSCs.
Collapse
Affiliation(s)
- Issei Saitoh
- Division of Pediatric Dentistry, Graduate School of Medical and Dental Science, Niigata University , Gakkocho-dori, Chuo-ku, Niigata , Japan
| | - Emi Inada
- † Department of Pediatric Dentistry, Kagoshima University Graduate School of Medical and Dental Sciences , Sakuragaoka, Kagoshima , Japan
| | - Yoko Iwase
- Division of Pediatric Dentistry, Graduate School of Medical and Dental Science, Niigata University , Gakkocho-dori, Chuo-ku, Niigata , Japan
| | - Hirofumi Noguchi
- ‡ Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus , Nishiharatyoaza, Uehara, Okinawa , Japan
| | - Tomoya Murakami
- Division of Pediatric Dentistry, Graduate School of Medical and Dental Science, Niigata University , Gakkocho-dori, Chuo-ku, Niigata , Japan
| | - Miki Soda
- Division of Pediatric Dentistry, Graduate School of Medical and Dental Science, Niigata University , Gakkocho-dori, Chuo-ku, Niigata , Japan
| | - Naoko Kubota
- † Department of Pediatric Dentistry, Kagoshima University Graduate School of Medical and Dental Sciences , Sakuragaoka, Kagoshima , Japan
| | - Hiroko Hasegawa
- † Department of Pediatric Dentistry, Kagoshima University Graduate School of Medical and Dental Sciences , Sakuragaoka, Kagoshima , Japan
| | - Eri Akasaka
- † Department of Pediatric Dentistry, Kagoshima University Graduate School of Medical and Dental Sciences , Sakuragaoka, Kagoshima , Japan
| | - Yuko Matsumoto
- † Department of Pediatric Dentistry, Kagoshima University Graduate School of Medical and Dental Sciences , Sakuragaoka, Kagoshima , Japan
| | - Kyoko Oka
- § Section of Pediatric Dentistry Department of Oral Growth and Development Fukuoka Dental College , Sawara-ku, Tamura Fukuoka-shi, Fukuoka , Japan
| | - Youichi Yamasaki
- † Department of Pediatric Dentistry, Kagoshima University Graduate School of Medical and Dental Sciences , Sakuragaoka, Kagoshima , Japan
| | - Haruaki Hayasaki
- Division of Pediatric Dentistry, Graduate School of Medical and Dental Science, Niigata University , Gakkocho-dori, Chuo-ku, Niigata , Japan
| | - Masahiro Sato
- ¶ Section of Gene Expression Regulation, Frontier Science Research Center, Kagoshima University , Sakuragaoka, Kagoshima , Japan
| |
Collapse
|
21
|
Desai N, Rambhia P, Gishto A. Human embryonic stem cell cultivation: historical perspective and evolution of xeno-free culture systems. Reprod Biol Endocrinol 2015; 13:9. [PMID: 25890180 PMCID: PMC4351689 DOI: 10.1186/s12958-015-0005-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 02/09/2015] [Indexed: 01/23/2023] Open
Abstract
Human embryonic stem cells (hESC) have emerged as attractive candidates for cell-based therapies that are capable of restoring lost cell and tissue function. These unique cells are able to self-renew indefinitely and have the capacity to differentiate in to all three germ layers (ectoderm, endoderm and mesoderm). Harnessing the power of these pluripotent stem cells could potentially offer new therapeutic treatment options for a variety of medical conditions. Since the initial derivation of hESC lines in 1998, tremendous headway has been made in better understanding stem cell biology and culture requirements for maintenance of pluripotency. The approval of the first clinical trials of hESC cells for treatment of spinal cord injury and macular degeneration in 2010 marked the beginning of a new era in regenerative medicine. Yet it was clearly recognized that the clinical utility of hESC transplantation was still limited by several challenges. One of the most immediate issues has been the exposure of stem cells to animal pathogens, during hESC derivation and during in vitro propagation. Initial culture protocols used co-culture with inactivated mouse fibroblast feeder (MEF) or human feeder layers with fetal bovine serum or alternatively serum replacement proteins to support stem cell proliferation. Most hESC lines currently in use have been exposed to animal products, thus carrying the risk of xeno-transmitted infections and immune reaction. This mini review provides a historic perspective on human embryonic stem cell culture and the evolution of new culture models. We highlight the challenges and advances being made towards the development of xeno-free culture systems suitable for therapeutic applications.
Collapse
Affiliation(s)
- Nina Desai
- Department of Obstetrics and Gynecology, Cleveland Clinic, Beachwood, OH, USA.
| | - Pooja Rambhia
- Department of Obstetrics and Gynecology, Cleveland Clinic, Beachwood, OH, USA.
| | - Arsela Gishto
- Department of Obstetrics and Gynecology, Cleveland Clinic, Beachwood, OH, USA.
| |
Collapse
|
22
|
Higuchi A, Ling QD, Kumar SS, Munusamy M, Alarfajj AA, Umezawa A, Wu GJ. Design of polymeric materials for culturing human pluripotent stem cells: Progress toward feeder-free and xeno-free culturing. Prog Polym Sci 2014. [DOI: 10.1016/j.progpolymsci.2014.01.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
23
|
Lee JE, Lee DR. Human embryonic stem cells: derivation, maintenance and cryopreservation. Int J Stem Cells 2014; 4:9-17. [PMID: 24298329 DOI: 10.15283/ijsc.2011.4.1.9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2011] [Indexed: 12/29/2022] Open
Abstract
Human embryonic stem cells (hESCs) are the most powerful candidate for the treatment of incurable diseases through the replacement of damaged cells and/or tissues in patients, although there are some obstacles to overcome for the clinical application of hESCs such as the assurance of guided differentiation and control of the immune response following cell therapy or tissue grafting. To obtain genetically stable hESCs and use them clinically, it is important to develop appropriate culture conditions. Additionally, the establishment of a hESC bank with a large number of hESC lines will be required for their clinical application because each hESC line is directed to have a different differentiation ability and immune characteristics such as HLA type. In this review, we describe the derivation and culture conditions of hESCs based on recent advances. Then, we will introduce several cryopreservation methods for hESCs, which is important for the development of cell bank.
Collapse
|
24
|
Zaninovic N, Zhan Q, Rosenwaks Z. Derivation of human embryonic stem cells (hESC). Methods Mol Biol 2014; 1154:121-44. [PMID: 24782008 DOI: 10.1007/978-1-4939-0659-8_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Stem cells are characterized by their absolute or relative lack of specialization their ability for self-renewal, as well as their ability to generate differentiated progeny through cellular lineages with one or more branches. The increased availability of embryonic tissue and greatly improved derivation methods have led to a large increase in the number of hESC lines.
Collapse
Affiliation(s)
- Nikica Zaninovic
- Center for Reproductive Medicine, Weill Cornell Medical College, 1305 York Avenue, New York, NY, 10021, USA,
| | | | | |
Collapse
|
25
|
Yamasaki S, Taguchi Y, Shimamoto A, Mukasa H, Tahara H, Okamoto T. Generation of human induced pluripotent stem (Ips) cells in serum- and feeder-free defined culture and TGF-Β1 regulation of pluripotency. PLoS One 2014; 9:e87151. [PMID: 24489856 PMCID: PMC3906124 DOI: 10.1371/journal.pone.0087151] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 12/19/2013] [Indexed: 11/19/2022] Open
Abstract
Human Embryonic Stem cells (hESCs) and human induced Pluripotent Stem cells (hiPSCs) are commonly maintained on inactivated mouse embryonic fibroblast as feeder cells in medium supplemented with FBS or proprietary replacements. Use of culture medium containing undefined or unknown components has limited the development of applications for pluripotent cells because of the relative lack of knowledge regarding cell responses to differentiating growth factors. In addition, there is no consensus as to the optimal formulation, or the nature of the cytokine requirements of the cells to promote their self-renewal and inhibit their differentiation. In this study, we successfully generated hiPSCs from human dental pulp cells (DPCs) using Yamanaka's factors (Oct3/4, Sox2, Klf4, and c-Myc) with retroviral vectors in serum- and feeder-free defined culture conditions. These hiPSCs retained the property of self-renewal as evaluated by the expression of self-renewal marker genes and proteins, morphology, cell growth rates, and pluripotency evaluated by differentiation into derivatives of all three primary germ layers in vitro and in vivo. In this study, we found that TGF-β1 increased the expression levels of pluripotency markers in a dose-dependent manner. However, increasing doses of TGF-β1 suppressed the growth rate of hiPSCs cultured under the defined conditions. Furthermore, over short time periods the hiPSCs cultured in hESF9 or hESF9T exhibited similar morphology, but hiPSCs maintained in hESF9 could not survive beyond 30 passages. This result clearly confirmed that hiPSCs cultured in hESF9 medium absolutely required TGF-β1 to maintain pluripotency. This simple serum-free adherent monoculture system will allow us to elucidate the cell responses to growth factors under defined conditions and can eliminate the risk might be brought by undefined pathogens.
Collapse
Affiliation(s)
- Sachiko Yamasaki
- Department of Molecular Oral Medicine and Maxillofacial Surgery, Applied Life Sciences, Graduate Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Yuki Taguchi
- Department of Molecular Oral Medicine and Maxillofacial Surgery, Applied Life Sciences, Graduate Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Akira Shimamoto
- Department of Cellular and Molecular Biology, Basic Life Sciences, Graduate Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Hanae Mukasa
- Department of Molecular Oral Medicine and Maxillofacial Surgery, Applied Life Sciences, Graduate Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Hidetoshi Tahara
- Department of Cellular and Molecular Biology, Basic Life Sciences, Graduate Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Tetsuji Okamoto
- Department of Molecular Oral Medicine and Maxillofacial Surgery, Applied Life Sciences, Graduate Institute of Biomedical & Health Sciences, Hiroshima University, Japan
- * E-mail:
| |
Collapse
|
26
|
Quang T, Marquez M, Blanco G, Zhao Y. Dosage and cell line dependent inhibitory effect of bFGF supplement in human pluripotent stem cell culture on inactivated human mesenchymal stem cells. PLoS One 2014; 9:e86031. [PMID: 24465853 PMCID: PMC3895015 DOI: 10.1371/journal.pone.0086031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 12/09/2013] [Indexed: 11/19/2022] Open
Abstract
Many different culture systems have been developed for expanding human pluripotent stem cells (hESCs and hiPSCs). In general, 4–10 ng/ml of bFGF is supplemented in culture media in feeder-dependent systems regardless of feeder cell types, whereas in feeder-free systems, up to 100 ng/ml of bFGF is required for maintaining long-term culture on various substrates. The amount of bFGF required in native hESCs growth niche is unclear. Here we report using inactivated adipose-derived human mesenchymal stem cells as feeder cells to examine long-term parallel cultures of two hESCs lines (H1 and H9) and one hiPSCs line (DF19-9-7T) in media supplemented with 0, 0.4 or 4 ng/ml of bFGF for up to 23 passages, as well as parallel cultures of H9 and DF19 in media supplemented with 4, 20 or 100 ng/ml bFGF for up to 13 passages for comparison. Across all cell lines tested, bFGF supplement demonstrated inhibitory effect over growth expansion, single cell colonization and recovery from freezing in a dosage dependent manner. In addition, bFGF exerted differential effects on different cell lines, inducing H1 and DF19 differentiation at 4 ng/ml or higher, while permitting long-term culture of H9 at the same concentrations with no apparent dosage effect. Pluripotency was confirmed for all cell lines cultured in 0, 0.4 or 4 ng/ml bFGF excluding H1-4 ng, as well as H9 cultured in 4, 20 and 100 ng/ml bFGF. However, DF19 demonstrated similar karyotypic abnormality in both 0 and 4 ng/ml bFGF media while H1 and H9 were karyotypically normal in 0 ng/ml bFGF after long-term culture. Our results indicate that exogenous bFGF exerts dosage and cell line dependent effect on human pluripotent stem cells cultured on mesenchymal stem cells, and implies optimal use of bFGF in hESCs/hiPSCs culture should be based on specific cell line and its culture system.
Collapse
Affiliation(s)
- Tara Quang
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, United States of America
| | - Maribel Marquez
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, United States of America
| | - Giselle Blanco
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, United States of America
| | - Yuanxiang Zhao
- Biological Sciences Department, California State Polytechnic University at Pomona, Pomona, California, United States of America
- * E-mail:
| |
Collapse
|
27
|
Anisimov SV. Stem cell therapy for neurodegenerative diseases: mind the gap. FUTURE NEUROLOGY 2014. [DOI: 10.2217/fnl.13.64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Sergey V Anisimov
- Research Unit of Cellular & Genetic Engineering, VA Almazov Federal Center for Heart, Blood & Endocrinology, Akkuratova Street 2, Saint Petersburg, 197341, Russiaand Department of Intracellular Signaling & Transport, Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia
| |
Collapse
|
28
|
Carvalho KATD, Abdelwahid E, Ferreira RJ, Irioda AC, Guarita-Souza LC. Preclinical stem cell therapy in Chagas Disease: Perspectives for future research. World J Transplant 2013; 3:119-126. [PMID: 24392316 PMCID: PMC3879521 DOI: 10.5500/wjt.v3.i4.119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/18/2013] [Indexed: 02/05/2023] Open
Abstract
Chagas cardiomyopathy still remains a challenging problem that is responsible for high morbidity and mortality in Central and Latin America. Chagas disease disrupts blood microcirculation via various autoimmune mechanisms, causing loss of cardiomyocytes and severe impairment of heart function. Different cell types and delivery approaches in Chagas Disease have been studied in both preclinical models and clinical trials. The main objective of this article is to clarify the reasons why the benefits that have been seen with cell therapy in preclinical models fail to translate to the clinical setting. This can be explained by crucial differences between the cellular types and pathophysiological mechanisms of the disease, as well as the differences between human patients and animal models. We discuss examples that demonstrate how the results from preclinical trials might have overestimated the efficacy of myocardial regeneration therapies. Future research should focus, not only on studying the best cell type to use but, very importantly, understanding the levels of safety and cellular interaction that can elicit efficient therapeutic effects in human tissue. Addressing the challenges associated with future research may ensure the success of stem cell therapy in improving preclinical models and the treatment of Chagas disease.
Collapse
|
29
|
Pakzad M, Ashtiani MK, Mousavi-Gargari SL, Baharvand H. Development of a simple, repeatable, and cost-effective extracellular matrix for long-term xeno-free and feeder-free self-renewal of human pluripotent stem cells. Histochem Cell Biol 2013; 140:635-48. [PMID: 24065274 DOI: 10.1007/s00418-013-1144-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2013] [Indexed: 12/18/2022]
Abstract
Given the potential importance of human pluripotent stem cells (hPSCs) in translational research and regenerative medicine, the aim of the present study was to develop a simple, safe, and cost-effective substrate to expand hPSCs. We report the development of an extracellular matrix (ECM), designated "RoGel," based on conditioned medium (CM) of human fibroblasts under serum- and xeno-free culture conditions. The long-term self-renewal of hPSCs on RoGel was also assessed. The results showed that self-renewal, pluripotency, plating efficiency, and cloning efficiency of hPSCs on this newly developed ECM were similar to those of Matrigel, the conventional mouse-cell line-derived ECM. The cells had the capability to passage mechanically on a cold surface, which resulted in their long-term maintenance with normal karyotype. We have demonstrated that CM-coated plates preserved for 1 year at room temperature maintained the capability of hPSC expansion. This ECM provides an attractive hPSC culture platform for both research and future therapeutic applications.
Collapse
Affiliation(s)
- Mohammad Pakzad
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | | | | |
Collapse
|
30
|
Mäkinen M, Joki T, Ylä-Outinen L, Skottman H, Narkilahti S, Aänismaa R. Fluorescent probes as a tool for cell population tracking in spontaneously active neural networks derived from human pluripotent stem cells. J Neurosci Methods 2013; 215:88-96. [PMID: 23473797 DOI: 10.1016/j.jneumeth.2013.02.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 02/25/2013] [Accepted: 02/27/2013] [Indexed: 11/19/2022]
Abstract
Applications such as 3D cultures and tissue modelling require cell tracking with non-invasive methods. In this work, the suitability of two fluorescent probes, CellTracker, CT, and long chain carbocyanine dye, DiD, was investigated for long-term culturing of labeled human pluripotent stem cell-derived neural cells. We found that these dyes did not affect the cell viability. However, proliferation was decreased in DiD labeled cell population. With both dyes the labeling was stable up to 4 weeks. CT and DiD labeled cells could be co-cultured and, importantly, these mixed populations had their normal ability to form spontaneous electrical network activity. In conclusion, human neural cells can be successfully labeled with these two fluorescent probes without significantly affecting the cell characteristics. These labeled cells could be utilized further in e.g. building controlled neuronal networks for neurotoxicity screening platforms, combining cells with biomaterials for 3D studies, and graft development.
Collapse
Affiliation(s)
- M Mäkinen
- NeuroGroup, Institute of Biomedical Technology/BioMediTech, Biokatu 12, FI-33014 University of Tampere, Finland.
| | | | | | | | | | | |
Collapse
|
31
|
Abdelwahid E, Siminiak T, Guarita-Souza LC, Teixeira de Carvalho KA, Gallo P, Shim W, Condorelli G. Stem cell therapy in heart diseases: a review of selected new perspectives, practical considerations and clinical applications. Curr Cardiol Rev 2013; 7:201-12. [PMID: 22758618 PMCID: PMC3263484 DOI: 10.2174/157340311798220502] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2010] [Revised: 01/31/2011] [Accepted: 02/03/2011] [Indexed: 12/13/2022] Open
Abstract
Degeneration of cardiac tissues is considered a major cause of mortality in the western world and is expected to be a greater problem in the forthcoming decades. Cardiac damage is associated with dysfunction and irreversible loss of cardiomyocytes. Stem cell therapy for ischemic heart failure is very promising approach in cardiovascular medicine. Initial trials have indicated the ability of cardiomyocytes to regenerate after myocardial injury. These preliminary trials aim to translate cardiac regeneration strategies into clinical practice. In spite of advances, current therapeutic strategies to ischemic heart failure remain very limited. Moreover, major obstacles still need to be solved before stem cell therapy can be fully applied. This review addresses the current state of research and experimental data regarding embryonic stem cells (ESCs), myoblast transplantation, histological and functional analysis of transplantation of co-cultured myoblasts and mesenchymal stem cells, as well as comparison between mononuclear and mesenchymal stem cells in a model of myocardium infarction. We also discuss how research with stem cell transplantation could translate to improvement of cardiac function.
Collapse
Affiliation(s)
- Eltyeb Abdelwahid
- CBRC, Massachusetts General Hospital/Harvard Medical School, Charlestown, Massachusetts 02129, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Substrates and supplements for hESCs: a critical review. J Assist Reprod Genet 2013; 30:315-23. [PMID: 23288664 DOI: 10.1007/s10815-012-9914-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 12/05/2012] [Indexed: 10/27/2022] Open
Abstract
BACKGROUND Different laboratories around the world have succeeded in establishing human embryonic stem cell (hESC) lines. However, culture conditions vary considerably among the protocols used and the vast majority of the lines at some stage of their creation have been in contact with an animal derived component. One of the main problems to be overcome for the generation of a clinical-grade hESC line is the choice of a substrate and medium that allows derivation and culture, where animal derived components are kept to a minimum or completely excluded. MATERIALS AND METHODS The following review describes past and more recent achievements in the creation and culturing of hESC. It describes protocols, giving special attention to the matrices and supplements used for derivation, maintainance and cryostorage, considering whether they included defined, undefined and/or animal-derived components in their formulations. CONCLUSION This information shall be useful for the creation and choice of new substrates and supplements for future research in the field of hESC for therapeutic purposes.
Collapse
|
33
|
Human Embryonic Stem Cells. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
34
|
Tang C, Weissman IL, Drukker M. Immunogenicity of in vitro maintained and matured populations: potential barriers to engraftment of human pluripotent stem cell derivatives. Methods Mol Biol 2013; 1029:17-31. [PMID: 23756939 DOI: 10.1007/978-1-62703-478-4_2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The potential to develop into any cell type makes human pluripotent stem cells (hPSCs) one of the most promising sources for regenerative treatments. Hurdles to their clinical applications include (1) formation of heterogeneously differentiated cultures, (2) the risk of teratoma formation from residual undifferentiated cells, and (3) immune rejection of engrafted cells. The recent production of human isogenic (genetically identical) induced PSCs (hiPSCs) has been proposed as a "solution" to the histocompatibility barrier. In theory, differentiated cells derived from patient-specific hiPSC lines should be histocompatible to their donor/recipient. However, propagation, maintenance, and non-physiologic differentiation of hPSCs in vitro may produce other, likely less powerful, immune responses. In light of recent progress towards the clinical application of hPSCs, this review focuses on two antigen presentation phenomena that may lead to rejection of isogenic hPSC derivates: namely, the expression of aberrant antigens as a result of long-term in vitro maintenance conditions or incomplete somatic cell reprogramming, and the unbalanced presentation of receptors and ligands involved in immune recognition due to accelerated differentiation. Finally, we discuss immunosuppressive approaches that could potentially address these immunological concerns.
Collapse
Affiliation(s)
- Chad Tang
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | | |
Collapse
|
35
|
Kaur J, Tilkins ML, Eckert R, Boucher S. Methods for culturing human embryonic stem cells in a xeno-free system. Methods Mol Biol 2013; 997:115-26. [PMID: 23546751 DOI: 10.1007/978-1-62703-348-0_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Defined pluripotent stem cell culture media is a valuable tool for tracking and analyzing morphological, cell signaling and gene expression changes in human embryonic stem cells. Cultivation of hESC under xeno-free culture settings provides researchers with a consistent and reproducible environment to test experimental hypotheses and move towards translational and clinical research (Richards et al., Nat Biotechnol 20:933-936, 2002; Richards et al., Stem Cells 21:546-556, 2003). One of the primary concerns of the xenogeneic culture is the transfer of foreign pathogens or antigens that induce disease or immune response by the patient. These undesirable by-products may come from the use of murine-derived feeder cells, xenogeneic matrices, or from animal-derived components found in the cell culture medium or matrix used to isolate or expand the stem cells (Beattie et al., Stem Cells 23:489-495, 2005; Koivisto et al., Reprod Biomed Online 9:330-337, 2004). This chapter describes standardized protocols for obtaining consistent and reproducible results for culturing PSC under xeno-free, feeder-free, or feeder-based systems.
Collapse
Affiliation(s)
- Jasmeet Kaur
- Primary and Stem Cell Systems, Life Technologies, Carlsbad, CA, USA
| | | | | | | |
Collapse
|
36
|
Ojala M, Rajala K, Pekkanen-Mattila M, Miettinen M, Huhtala H, Aalto-Setälä K. Culture conditions affect cardiac differentiation potential of human pluripotent stem cells. PLoS One 2012; 7:e48659. [PMID: 23119085 PMCID: PMC3485380 DOI: 10.1371/journal.pone.0048659] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 09/28/2012] [Indexed: 12/21/2022] Open
Abstract
Human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), are capable of differentiating into any cell type in the human body and thus can be used in studies of early human development, as cell models for different diseases and eventually also in regenerative medicine applications. Since the first derivation of hESCs in 1998, a variety of culture conditions have been described for the undifferentiated growth of hPSCs. In this study, we cultured both hESCs and hiPSCs in three different culture conditions: on mouse embryonic fibroblast (MEF) and SNL feeder cell layers together with conventional stem cell culture medium containing knockout serum replacement and basic fibroblast growth factor (bFGF), as well as on a Matrigel matrix in mTeSR1 medium. hPSC lines were subjected to cardiac differentiation in mouse visceral endodermal-like (END-2) co-cultures and the cardiac differentiation efficiency was determined by counting both the beating areas and Troponin T positive cells, as well as studying the expression of OCT-3/4, mesodermal Brachyury T and NKX2.5 and endodermal SOX-17 at various time points during END-2 differentiation by q-RT-PCR analysis. The most efficient cardiac differentiation was observed with hPSCs cultured on MEF or SNL feeder cell layers in stem cell culture medium and the least efficient cardiac differentiation was observed on a Matrigel matrix in mTeSR1 medium. Further, hPSCs cultured on a Matrigel matrix in mTeSR1 medium were found to be more committed to neural lineage than hPSCs cultured on MEF or SNL feeder cell layers. In conclusion, culture conditions have a major impact on the propensity of the hPSCs to differentiate into a cardiac lineage.
Collapse
Affiliation(s)
- Marisa Ojala
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
| | - Kristiina Rajala
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
| | - Mari Pekkanen-Mattila
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
| | - Marinka Miettinen
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
| | - Heini Huhtala
- School of Health Sciences, University of Tampere, Tampere, Finland
| | - Katriina Aalto-Setälä
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
- Heart Center, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
37
|
Badger JL, Byrne ML, Veraitch FS, Mason C, Wall IB, Caldwell MA. Hypoxic culture of human pluripotent stem cell lines is permissible using mouse embryonic fibroblasts. Regen Med 2012; 7:675-83. [DOI: 10.2217/rme.12.55] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Aim: Hypoxia is used within in vitro stem cell culture to recreate conditions similar to the in vivo environment surrounding the early blastocyst, from which embryonic stem cells can be isolated. Traditionally, basic research has used a coculture feeder system to culture pluripotent stem cells; however, it is possible that lowered oxygen may restrict cellular metabolic activity of the inactivated mouse embryonic fibroblasts (iMEFs) by disrupting oxygen-dependent pathways, such as ATP production through aerobic respiration. In this work, we examined the potential to continue using routine culture methods, such as iMEFs, to support human pluripotent cell expansion under hypoxia instead of feeder-free methods that can cause cell instability and offer a poor cell attachment rate. Materials & methods: Metabolic activity and viability studies were carried out in normoxic and hypoxic conditions. Pluripotent stem cells were introduced into hypoxia on iMEFs and the rate of colony expansion was compared with normoxic conditions. In addition, pluripotent stem cells were grown in hypoxia for over 6 months to demonstrate maintenance of pluripotency. Immunocytochemistry and western blotting evaluated the activity of the hypoxic transcription factor, HIF1A. Results: Hypoxia does not significantly affect viability or metabolic activity of feeder cells, and there is no detrimental effect on the rate of pluripotent stem cell colony expansion when cells are cultured in hypoxia. In addition, hypoxic pluripotent stem cells maintain their pluripotent nature and ability to differentiate into the three germ layers. Conclusion: The traditional iMEF coculture method is suitable for use in hypoxia and does not need to be replaced with feeder-free systems for hypoxic culture of human pluripotent stem cell lines in basic research.
Collapse
Affiliation(s)
- Jennifer L Badger
- Department of Biochemical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
- Henry Wellcome Laboratories for Integrative Neuroscience & Endocrinology, Dorothy Hodgkin Building, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| | - Meg L Byrne
- Department of Biochemical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
- Henry Wellcome Laboratories for Integrative Neuroscience & Endocrinology, Dorothy Hodgkin Building, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| | - Farlan S Veraitch
- Department of Biochemical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
| | - Chris Mason
- Department of Biochemical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
| | - Ivan B Wall
- Department of Biochemical Engineering, University College London, Torrington Place, London, WC1E 7JE, UK
| | - Maeve A Caldwell
- Henry Wellcome Laboratories for Integrative Neuroscience & Endocrinology, Dorothy Hodgkin Building, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| |
Collapse
|
38
|
HU JIABO, HU SANQIANG, MA QUANHUI, WANG XIAOHUI, ZHOU ZHONGWEI, ZHANG WEI, SUN XIAOCHUN, ZHU WEI, QIAN HUI, XU WENRONG. Immortalized mouse fetal liver stromal cells support growth and maintenance of human embryonic stem cells. Oncol Rep 2012; 28:1385-91. [DOI: 10.3892/or.2012.1909] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 06/19/2012] [Indexed: 11/06/2022] Open
|
39
|
Tannenbaum SE, Tako Turetsky T, Singer O, Aizenman E, Kirshberg S, Ilouz N, Gil Y, Berman-Zaken Y, Perlman TS, Geva N, Levy O, Arbell D, Simon A, Ben-Meir A, Shufaro Y, Laufer N, Reubinoff BE. Derivation of xeno-free and GMP-grade human embryonic stem cells--platforms for future clinical applications. PLoS One 2012; 7:e35325. [PMID: 22745653 PMCID: PMC3380026 DOI: 10.1371/journal.pone.0035325] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Accepted: 03/12/2012] [Indexed: 11/24/2022] Open
Abstract
Clinically compliant human embryonic stem cells (hESCs) should be developed in adherence to ethical standards, without risk of contamination by adventitious agents. Here we developed for the first time animal-component free and good manufacturing practice (GMP)-compliant hESCs. After vendor and raw material qualification, we derived xeno-free, GMP-grade feeders from umbilical cord tissue, and utilized them within a novel, xeno-free hESC culture system. We derived and characterized three hESC lines in adherence to regulations for embryo procurement, and good tissue, manufacturing and laboratory practices. To minimize freezing and thawing, we continuously expanded the lines from initial outgrowths and samples were cryopreserved as early stocks and banks. Batch release criteria included DNA-fingerprinting and HLA-typing for identity, characterization of pluripotency-associated marker expression, proliferation, karyotyping and differentiation in-vitro and in-vivo. These hESCs may be valuable for regenerative therapy. The ethical, scientific and regulatory methodology presented here may serve for development of additional clinical-grade hESCs.
Collapse
Affiliation(s)
- Shelly E. Tannenbaum
- The Hadassah Human Embryonic Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Tikva Tako Turetsky
- The Hadassah Human Embryonic Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Orna Singer
- The Hadassah Human Embryonic Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Einat Aizenman
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Sophie Kirshberg
- The Hadassah Human Embryonic Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Nili Ilouz
- The Hadassah Human Embryonic Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Yaniv Gil
- The Hadassah Human Embryonic Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Yael Berman-Zaken
- The Hadassah Human Embryonic Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Temima Schnitzer Perlman
- The Hadassah Human Embryonic Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Nitshia Geva
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Ora Levy
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Daniel Arbell
- Department of Pediatric Surgery, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Alex Simon
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Assaf Ben-Meir
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Yoel Shufaro
- The Hadassah Human Embryonic Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Neri Laufer
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Benjamin E. Reubinoff
- The Hadassah Human Embryonic Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
40
|
|
41
|
Generation of human-induced pluripotent stem cells by a nonintegrating RNA Sendai virus vector in feeder-free or xeno-free conditions. Stem Cells Int 2012; 2012:564612. [PMID: 22550511 PMCID: PMC3328201 DOI: 10.1155/2012/564612] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 01/04/2012] [Indexed: 01/19/2023] Open
Abstract
The generation of induced pluripotent stem cells (iPSCs) from somatic cells has enabled the possibility of providing unprecedented access to patient-specific iPSC cells for drug screening, disease modeling, and cell therapy applications. However, a major obstacle to the use of iPSC for therapeutic applications is the potential of genomic modifications caused by insertion of viral transgenes in the cellular genome. A second concern is that reprogramming often requires the use of animal feeder layers and reagents that contain animal origin products, which hinder the generation of clinical-grade iPSCs. Here, we report the generation of iPSCs by an RNA Sendai virus vector that does not integrate into the cells genome, providing transgene-free iPSC line. In addition, reprogramming can be performed in feeder-free condition with StemPro hESC SFM medium and in xeno-free (XF) conditions. Generation of an integrant-free iPSCs generated in xeno-free media should facilitate the safe downstream applications of iPSC-based cell therapies.
Collapse
|
42
|
The effect of human and mouse fibroblast feeder cells on cardiac differentiation of human pluripotent stem cells. Stem Cells Int 2012; 2012:875059. [PMID: 22315618 PMCID: PMC3270482 DOI: 10.1155/2012/875059] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 10/06/2011] [Indexed: 12/21/2022] Open
Abstract
Mouse embryonic fibroblasts (MEFs) and human foreskin fibroblasts (hFFs) are commonly used as feeder cells to maintain the pluripotent state of stem cells. The aim of the present study was to evaluate the effect of MEF and hFF feeders on the cardiac differentiation. Two human embryonic and two induced pluripotent stem cell lines were cultured on MEF and hFF before cardiac differentiation. The expression of Brachyury T was higher in cell lines cultured on MEF, than if cultured on hFF, suggesting enhanced mesoderm formation. However, significant positive influence of MEF feeders on cardiac differentiation was only seen with one cell line. Further, the ability of hFF to maintain pluripotency of stem cells originally cultured on MEF was quite poor. In conclusion, the cells behaved differently whether cultured on hFF or MEF feeders. However, the influence of the feeder cells on differentiation was less than the difference observed between the cell lines.
Collapse
|
43
|
Heng BC, Li J, Chen AKL, Reuveny S, Cool SM, Birch WR, Oh SKW. Translating human embryonic stem cells from 2-dimensional to 3-dimensional cultures in a defined medium on laminin- and vitronectin-coated surfaces. Stem Cells Dev 2011; 21:1701-15. [PMID: 22034857 DOI: 10.1089/scd.2011.0509] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
While defining the environment for human embryonic stem cell (hESC) culture on 2-dimensional (2D) surfaces has made rapid progress, the industrial-scale implementation of this technology will benefit from translating this knowledge into a 3-dimensional (3D) system, thus enabling better control, automation, and volumetric scale-up in bioreactors. The current study describes a system with defined conditions that are capable of supporting the long-term 2D culture of hESCs and the transposing of these conditions to 3D microcarrier (MC) cultures. Vitronectin (VN) and laminin (LN) were chosen as matrices for the long-term propagation of hESCs in a defined culture medium (STEMPRO(®)) for conventional 2D culture. Adsorption of these proteins onto 2D tissue culture polystyrene (TCPS) indicated that surface density saturation of 510 and 850 ng/cm(2) for VN and LN, respectively, was attained above 20 μg/mL deposition solution concentration. Adsorption of these proteins onto spherical (97±10 μm), polystyrene MC followed a similar trend and coating surface densities of 450 and 650 ng/cm(2) for VN and LN, respectively, were used to support hESC propagation. The long-term expansion of hESCs was equally successful on TCPS and MC, with consistently high expression (>90%) of pluripotent markers (OCT-4, MAB-84, and TRA-1-60) over 20 passages and maintenance of karyotypic normality. The average fold increase in cell numbers on VN-coated MC per serial passage was 8.5±1.0, which was similar to LN-coated MC (8.5±0.9). Embryoid body differentiation assays and teratoma formation confirmed that hESCs retained the ability to differentiate into lineages of all 3 germ layers, thus demonstrating the first translation to a fully defined MC-based environment for the expansion of hESCs.
Collapse
Affiliation(s)
- Boon Chin Heng
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Stem cell culture systems that rely on undefined animal-derived components introduce variability to the cultures and complicate their therapeutic use. The derivation of human embryonic stem cells and the development of methods to produce induced pluripotent stem cells combined with their potential to treat human diseases have accelerated the drive to develop xenogenic-free, chemically defined culture systems that support pluripotent self-renewal and directed differentiation. In this chapter, we describe four xeno-free culture systems that have been successful in supporting undifferentiated growth of hPSCs as well as methods for xeno-free subculture and cryopreservation of hPSCs. Each culture system consists of a xeno-free growth medium and xeno-free substratum: (1) TeSR2™ with human recombinant laminin (LN-511); (2) NutriStem™ with LN-511; (3) RegES™ with human foreskin fibroblasts (hFFs); (4) KO-SR Xeno-Free™/GF cocktail with CELLstart™ matrix.
Collapse
|
45
|
Sundberg M, Hyysalo A, Skottman H, Shin S, Vemuri M, Suuronen R, Narkilahti S. A xeno-free culturing protocol for pluripotent stem cell-derived oligodendrocyte precursor cell production. Regen Med 2011; 6:449-60. [PMID: 21749203 DOI: 10.2217/rme.11.36] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
AIM To show that human embryonic stem cells (hESCs) can be efficiently differentiated into oligodendrocyte precursor cells (OPCs) in a xeno-free medium with a specific medium supplement and specific human recombinant growth factors. MATERIALS & METHODS The xeno-free OPC-differentiation medium for pluripotent stem cells was developed by using StemPro® neural stem cell xeno-free medium supplement together with human recombinant growth factors SHH, PDGF-AA, IGF-1, EGF, basic FGF and CNTF, in addition to RA, T3, human laminin and ascorbic acid. We analyzed the differentiated hESC-derived OPCs and oligodendrocytes with quantitative real-time (RT)-PCR, RT-PCR, flow cytometry and immunocytochemistry, and we performed NG2-positive selection for OPC cultures with fluorescence-activated cell sorting. RESULTS Based on quantitative RT-PCR analysis, OPCs after 9 weeks of differentiation in xeno-free medium expressed OLIG2, SOX10 and NKX2.2 at elevated levels compared with control conditions. According to the flow cytometric analysis, the cells expressed A2B5 (>70%) and NG2 (40-60%) at 5 weeks time point whereas maturing oligodendrocytes expressed O4 (60-80%) at 11 weeks time point. In addition, hESC-derived OPC populations were purified based on NG2-positive selection using fluorescence-activated cell sorting. NG2-positive OPC populations survived and differentiated further into O4 expressing oligodendrocytes in xeno-free medium, and the sorted cell populations were free from pluripotent Tra1-81 and Oct-4 -positive cells. CONCLUSIONS This study confirms that the xeno-free culturing method can support the differentiation and purification of hESC-derived OPC populations and provides an initial step toward safe cell graft production for the future clinical applications.
Collapse
Affiliation(s)
- Maria Sundberg
- Regea - Institute for Regenerative Medicine, University of Tampere & Tampere University Hospital, Tampere, Finland
| | | | | | | | | | | | | |
Collapse
|
46
|
Kol’tsova AM, Gordeeva OF, Krylova TA, Lifantseva NV, Musorina AS, Yakovleva TK, Poljanskaya GG. Comparative characteristics of new human embryonic stem cell lines SC5, SC6, SC7, and SC3a. Russ J Dev Biol 2011. [DOI: 10.1134/s1062360411040072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
47
|
Alfred R, Taiani JT, Krawetz RJ, Yamashita A, Rancourt DE, Kallos MS. Large-scale production of murine embryonic stem cell-derived osteoblasts and chondrocytes on microcarriers in serum-free media. Biomaterials 2011; 32:6006-16. [PMID: 21620471 DOI: 10.1016/j.biomaterials.2011.04.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Accepted: 04/05/2011] [Indexed: 12/20/2022]
Abstract
The generation of tissue-engineered constructs from stem cells for the treatment of musculoskeletal diseases may have immense impact in regenerative medicine, but there are difficulties associated with stem cell culture and differentiation, including the use of serum. Here we present serum-free protocols for the successful production of murine embryonic stem cell (mESC) derived osteoblasts and chondrocytes on CultiSpher S macroporous microcarriers in stirred suspension bioreactors. Various inoculum forms and agitation rates were investigated. Produced osteogenic cells were implanted ectopically into SCID mice and orthotopically into a murine burr-hole fracture model. Osterix, osteocalcin and collagen type I were upregulated in osteogenic cultures, while aggrecan and collagen type II were upregulated in chondrogenic cultures. Histological analysis using alizarin red S, von Kossa and alcian blue staining confirmed the presence of osteoblasts and chondrocytes, respectively in cultured microcarriers and excised tissue. Finally, implantation of derived cells into a mouse fracture model revealed cellular integration without any tumor formation. Overall, microcarriers may provide a supportive scaffold for ESC expansion and differentiation in a serum-free bioprocess for in vivo implantation. These findings lay the groundwork for the development of clinical therapies for musculoskeletal injuries and diseases using hESCs and iPS cells.
Collapse
Affiliation(s)
- Roz Alfred
- Pharmaceutical Production Research Facility (PPRF), Schulich School of Engineering, University of Calgary, Calgary, AB T2N1N4, Canada
| | | | | | | | | | | |
Collapse
|
48
|
Rao M. Keeping things simple. Nat Methods 2011; 8:389-90. [PMID: 21527929 DOI: 10.1038/nmeth.1598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
49
|
Alfred R, Radford J, Fan J, Boon K, Krawetz R, Rancourt D, Kallos MS. Efficient suspension bioreactor expansion of murine embryonic stem cells on microcarriers in serum-free medium. Biotechnol Prog 2011; 27:811-23. [DOI: 10.1002/btpr.591] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 01/31/2011] [Indexed: 12/15/2022]
|
50
|
Tang C, Drukker M. Potential barriers to therapeutics utilizing pluripotent cell derivatives: intrinsic immunogenicity of in vitro maintained and matured populations. Semin Immunopathol 2011; 33:563-72. [PMID: 21479877 DOI: 10.1007/s00281-011-0269-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 03/28/2011] [Indexed: 01/20/2023]
Abstract
The potential to develop into any tissue makes pluripotent stem cells (PSCs) one of the most promising sources for cellular therapeutics. However, numerous hurdles exist to their clinical applications, three of the most concerning include the inability to separate therapeutic population from heterogeneously differentiated cultures, the risk of teratoma formation from residual pluripotent cells, and immunologic rejection of engrafted cells. The recent development of induced PSCs has been proposed as a solution to the histocompatibility barrier. Theoretically, creation of patient-specific induced PSC lines would exhibit a complete histocompatibility antigen match. However, regardless of the PSC source, in vitro propagation and nonphysiologic differentiation may result in other, likely less powerful, mechanisms of immune rejection. In light of recent progress towards clinical application, this review focuses on two such potential immunologic mechanisms applicable to isogenic PSC derivates: namely, the immunogenicity of aberrant antigens resulting from long-term in vitro maintenance and alterations in immunologic properties due to rapid in vitro differentiation. These issues will be considered with attention to their relation to effector cells in the adult immune system. In addition, we highlight immunosuppressive approaches that could potentially address the immunogenicity of these proposed mechanisms.
Collapse
Affiliation(s)
- Chad Tang
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | | |
Collapse
|