551
|
Jing D, Parikh A, Canty JM, Tzanakakis ES. Stem cells for heart cell therapies. TISSUE ENGINEERING PART B-REVIEWS 2009; 14:393-406. [PMID: 18821841 DOI: 10.1089/ten.teb.2008.0262] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Myocardial infarction-induced heart failure is a prevailing cause of death in the United States and most developed countries. The cardiac tissue has extremely limited regenerative potential, and heart transplantation for reconstituting the function of damaged heart is severely hindered mainly due to the scarcity of donor organs. To that end, stem cells with their extensive proliferative capacity and their ability to differentiate toward functional cardiomyocytes may serve as a renewable cellular source for repairing the damaged myocardium. Here, we review recent studies regarding the cardiogenic potential of adult progenitor cells and embryonic stem cells. Although large strides have been made toward the engineering of cardiac tissues using stem cells, important issues remain to be addressed to enable the translation of such technologies to the clinical setting.
Collapse
Affiliation(s)
- Donghui Jing
- Department of Chemical and Biological Engineering, State University of New York at Buffalo, Buffalo, New York 14260, USA
| | | | | | | |
Collapse
|
552
|
Braam SR, Passier R, Mummery CL. Cardiomyocytes from human pluripotent stem cells in regenerative medicine and drug discovery. Trends Pharmacol Sci 2009; 30:536-45. [PMID: 19762090 DOI: 10.1016/j.tips.2009.07.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Revised: 07/17/2009] [Accepted: 07/21/2009] [Indexed: 01/22/2023]
Abstract
Stem cells derived from pre-implantation human embryos or from somatic cells by reprogramming are pluripotent and self-renew indefinitely in culture. Pluripotent stem cells are unique in being able to differentiate to any cell type of the human body. Differentiation towards the cardiac lineage has attracted significant attention, initially with a strong focus on regenerative medicine. Although an important research area, the heart has proven challenging to repair by cardiomyocyte replacement. However, the ability to reprogramme adult cells to pluripotent stem cells and genetically manipulate stem cells presented opportunities to develop models of human disease. The availability of human cardiomyocytes from stem cell sources is expected to accelerate the discovery of cardiac drugs and safety pharmacology by offering more clinically relevant human culture models than presently available. Here we review the state-of-the-art using stem cell-derived human cardiomyocytes in drug discovery, drug safety pharmacology, and regenerative medicine.
Collapse
Affiliation(s)
- Stefan R Braam
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2300 RC Leiden, The Netherlands
| | | | | |
Collapse
|
553
|
Yang Z, Sui L, Toh WS, Lee EH, Cao T. Stage-dependent effect of TGF-beta1 on chondrogenic differentiation of human embryonic stem cells. Stem Cells Dev 2009; 18:929-40. [PMID: 18855519 DOI: 10.1089/scd.2008.0219] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-beta (TGF-beta) is known to be a potent inducer of stem cell chondrogenic differentiation. Transforming growth factor-beta/activin/nodal-signaling pathway has also been shown to be involved in maintaining the pluripotency of embryonic stem cells (ESCs). In this study, the effect of TGF-beta1 in chondrogenic differentiation of ESCs was examined both with undifferentiated ESCs that bypassed classical embryoid body (EB) formation, and on 5-day EB-derived cells. The effect of TGF-beta1 was compared to cells differentiated in serum-free chondrogenic basal medium without growth factor supplement. Analysis by real-time polymerase chain reaction (PCR), type II collagen enzyme-linked immunosorbent assay, sulfated glycoaminoglycan quantification and fluorescence immunostaining demonstrated substantial chondrogenic differentiation of ESCs regardless of EB formation in the absence of the growth factor. Addition of TGF-beta1 significantly inhibited chondrogenic gene expression and collagen deposition with a more potent effect on the cells that bypassed EB formation. Our study using a TGF-beta/activin/nodal-signaling inhibitor suggested that TGF-beta inhibited early chondrogenic induction but was required at the later stage of differentiation, which was also reflected in the enhancing effect of TGF-beta1 on chondrogenic development at later time points in EB-derived cells. Analysis of the pluripotency markers demonstrated sustained Oct4 and Nanog expression in the presence of TGF-beta1 with Oct4-positive cells detected in subpopulations of the differentiated culture. Our results suggest that TGF-beta1 suppresses ESC chondrogenic induction and the degree of suppression is dependent on the differentiation-stage of the ESC. Transforming growth factor-beta signaling, however, is required for functional chondrogenic development of ESC. Our finding that TGF-beta can sustain an undifferentiated population of human ESCs within the differentiation culture suggests that caution should be exercised when using this growth factor as an ESC chondrogenic inducer and highlights the importance of a selection protocol for chondroprogenitor cells to avoid possible teratoma formation in vivo.
Collapse
Affiliation(s)
- Zheng Yang
- Stem Cell Laboratory, Department of Oral Maxillo-Facial Surgery, Faculty of Dentistry, National University of Singapore, Singapore
| | | | | | | | | |
Collapse
|
554
|
Chan KKK, Zhang J, Chia NY, Chan YS, Sim HS, Tan KS, Oh SKW, Ng HH, Choo ABH. KLF4 and PBX1 directly regulate NANOG expression in human embryonic stem cells. Stem Cells 2009; 27:2114-25. [PMID: 19522013 DOI: 10.1002/stem.143] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Insight into the regulation of core transcription factors is important for a better understanding of the molecular mechanisms that control self-renewal and pluripotency of human ESCs (hESCs). However, the transcriptional regulation of NANOG itself in hESCs has largely been elusive. We established a NANOG promoter luciferase reporter assay as a fast read-out for indicating the pluripotent status of hESCs. From the functional cDNA screens and NANOG promoter characterization, we successfully identified a zinc finger transcription factor KLF4 and a homeodomain transcription factor PBX1 as two novel transcriptional regulators that maintain the pluripotent and undifferentiated state of hESCs. We showed that both KLF4 and PBX1 mRNA and protein expression were downregulated during hESC differentiation. In addition, overexpression of KLF4 and PBX1 upregulated NANOG promoter activity and also the endogenous NANOG protein expression in hESCs. Direct binding of KLF4 on NANOG proximal promoter and PBX1 on a new upstream enhancer and proximal promoter were confirmed by chromatin immunoprecipitation and electrophoretic mobility shift assay. Knockdown of KLF4/PBX1 or mutation of KLF4/PBX1 binding motifs significantly downregulated NANOG promoter activity. We also showed that specific members of the SP/KLF and PBX family are functionally redundant at the NANOG promoter and that KLF4 and PBX1 cooperated with OCT4 and SOX2, and transactivated synergistically the NANOG promoter activity. Our results show two novel upstream transcription activators of NANOG that are functionally important for the self-renewal of hESC and provide new insights into the expanded regulatory circuitry that maintains hESC pluripotency.
Collapse
Affiliation(s)
- Ken Kwok-Keung Chan
- Stem Cell Group, Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore.
| | | | | | | | | | | | | | | | | |
Collapse
|
555
|
Pant D, Keefer CL. Expression of Pluripotency-Related Genes during Bovine Inner Cell Mass Explant Culture. CLONING AND STEM CELLS 2009; 11:355-65. [DOI: 10.1089/clo.2008.0078] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Disha Pant
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Carol L. Keefer
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| |
Collapse
|
556
|
Enver T, Pera M, Peterson C, Andrews PW. Stem cell states, fates, and the rules of attraction. Cell Stem Cell 2009; 4:387-97. [PMID: 19427289 DOI: 10.1016/j.stem.2009.04.011] [Citation(s) in RCA: 241] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Understanding cell-fate decisions in stem cell populations is a major goal of modern biology. Stem and progenitor cell populations are often heterogeneous, which may reflect stem cell subsets that express subtly different properties, including different propensities for lineage selection upon differentiation, yet remain able to interconvert. We discuss these properties with examples both from the hematopoietic and embryonic stem cell (ESC) systems. The nature of the stem cell substates and their relationship to commitment to differentiate and lineage selection can be elucidated in terms of a landscape picture in which stable states can be defined mathematically as attractors.
Collapse
Affiliation(s)
- Tariq Enver
- The Molecular Haematology Unit, The Weatherall Institute of Molecular Medicine, Oxford, UK
| | | | | | | |
Collapse
|
557
|
Thomas RJ, Anderson D, Chandra A, Smith NM, Young LE, Williams D, Denning C. Automated, scalable culture of human embryonic stem cells in feeder-free conditions. Biotechnol Bioeng 2009; 102:1636-44. [PMID: 19062183 DOI: 10.1002/bit.22187] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Large-scale manufacture of human embryonic stem cells (hESCs) is prerequisite to their widespread use in biomedical applications. However, current hESC culture strategies are labor-intensive and employ highly variable processes, presenting challenges for scaled production and commercial development. Here we demonstrate that passaging of the hESC lines, HUES7, and NOTT1, with trypsin in feeder-free conditions, is compatible with complete automation on the CompacT SelecT, a commercially available and industrially relevant robotic platform. Pluripotency was successfully retained, as evidenced by consistent proliferation during serial passage, expression of stem cell markers (OCT4, NANOG, TRA1-81, and SSEA-4), stable karyotype, and multi-germlayer differentiation in vitro, including to pharmacologically responsive cardiomyocytes. Automation of hESC culture will expedite cell-use in clinical, scientific, and industrial applications.
Collapse
|
558
|
Enhancement of human embryonic stem cell pluripotency through inhibition of the mitochondrial respiratory chain. Stem Cell Res 2009; 3:142-56. [PMID: 19716358 DOI: 10.1016/j.scr.2009.07.002] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Revised: 07/24/2009] [Accepted: 07/26/2009] [Indexed: 11/24/2022] Open
Abstract
Human embryonic stem cell (hESC) pluripotency has been reported by several groups to be best maintained by culture under physiological oxygen conditions. Building on that finding, we inhibited complex III of the mitochondrial respiratory chain using antimycin A or myxothiazol to examine if specifically targeting the mitochondria would have a similar beneficial result for the maintenance of pluripotency. hESCs grown in the presence of 20 nM antimycin A maintained a compact morphology with high nuclear/cytoplasmic ratios. Furthermore, real-time PCR analysis demonstrated that the levels of Nanog mRNA were elevated 2-fold in antimycin A-treated cells. Strikingly, antimycin A was also able to replace bFGF in the media without compromising pluripotency, as long as autocrine bFGF signaling was maintained. Further analysis using low-density quantitative PCR arrays showed that antimycin A treatment reduced the expression of genes associated with differentiation, possibly acting through a ROS-mediated pathway. These results demonstrate that modulation of mitochondrial function results in increased pluripotency of the cell population, and sheds new light on the mechanisms and signaling pathways modulating hESC pluripotency.
Collapse
|
559
|
Montes R, Ligero G, Sanchez L, Catalina P, de la Cueva T, Nieto A, Melen GJ, Rubio R, García-Castro J, Bueno C, Menendez P. Feeder-free maintenance of hESCs in mesenchymal stem cell-conditioned media: distinct requirements for TGF-beta and IGF-II. Cell Res 2009; 19:698-709. [PMID: 19308090 DOI: 10.1038/cr.2009.35] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A paracrine regulation was recently proposed in human embryonic stem cells (hESCs) grown in mouse embryonic fibroblast (MEF)-conditioned media (MEF-CM), where hESCs spontaneously differentiate into autologous fibroblast-like cells to maintain culture homeostasis by producing TGF-beta and insulin-like growth factor-II (IGF-II) in response to basic fibroblast growth factor (bFGF). Although the importance of TGF-beta family members in the maintenance of pluripotency of hESCs is widely established, very little is known about the role of IGF-II. In order to ease hESC culture conditions and to reduce xenogenic components, we sought (i) to determine whether hESCs can be maintained stable and pluripotent using CM from human foreskin fibroblasts (HFFs) and human mesenchymal stem cells (hMSCs) rather than MEF-CM, and (ii) to analyze whether the cooperation of bFGF with TGF-beta and IGF-II to maintain hESCs in MEF-CM may be extrapolated to hESCs maintained in allogeneic mesenchymal stem cell (MSC)-CM and HFF-CM. We found that MSCs and HFFs express all FGF receptors (FGFR1-4) and specifically produce TGF-beta in response to bFGF. However, HFFs but not MSCs secrete IGF-II. Despite the absence of IGF-II in MSC-CM, hESC pluripotency and culture homeostasis were successfully maintained in MSC-CM for over 37 passages. Human ESCs derived on MSCs and hESCs maintained in MSC-CM retained hESC morphology, euploidy, expression of surface markers and transcription factors linked to pluripotency and displayed in vitro and in vivo multilineage developmental potential, suggesting that IGF-II may be dispensable for hESC pluripotency. In fact, IGF-II blocking had no effect on the homeostasis of hESC cultures maintained either on HFF-CM or on MSC-CM. These data indicate that hESCs are successfully maintained feeder-free with IGF-II-lacking MSC-CM, and that the previously proposed paracrine mechanism by which bFGF cooperates with TGF-beta and IGF-II in the maintenance of hESCs in MEF-CM may not be fully extrapolated to hESCs maintained in CM from human MSCs.
Collapse
Affiliation(s)
- Rosa Montes
- Andalusian Stem Cell Bank, Instituto de Investigación Biomédica, University of Granada, Granada, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
560
|
Hashemi-Tabar M, Orazizadeh M, Ghanbari A, Dehbashi FN. Kinetics of gene expression during exposure of mouse stem cells to activin A. Pak J Biol Sci 2009; 12:324-31. [PMID: 19579965 DOI: 10.3923/pjbs.2009.324.331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
This study aimed to evaluate the pattern of gene expression induced by activin A in mouse Embryonic Stem Cells (ESCs). Mouse ES cells cultured in undifferentiated state by leukemia inhibitory factor and feeder layer cells. Following removing these two anti differentiation factors for 5 days and forming Embryoid Bodies (EBs), the cells divided to 8 equal cells per groups. Differentiation procedure was performed in a two staged protocol; Formed EBs for 4 days (Stage one); expanded differentiated ESCs on gelatin coated dishes for one week (stage two). In the stage one, the media of groups 2-7 contained 10, 30 and 100 ng mL(-1) Activin A. The media in stage two was the same for all groups and contained only Fetal Bovine Serum (FBS). The expression of undifferentiated, ectoderm, mesoderm and endoderm markers were compared with relative RT-PCR method and statistically analyzed. The expression of an undifferentiating marker; Nanog was increased in the Activin A treated groups of stage one. The expression of OCT4 reduced in Activin A treated groups in stage two. In the stage one, the expression of Nodal increased by Activin A. expression of sonic hedgehog (Shh) was suppressed in Activin A treated groups of both stages. In stage two, there were significant decrease for the expression of mesoderm (Brachyury) and Nodal and visceral endoderm (GATA4) markers (p < 0.01). The expression of definitive endoderm markers (PDX1, TAT) showed significantly increased in Activin A treated groups (p < 0.01). Activin A induced differentiation in high concentration by imbalance in undifferentiating markers. Nodal has a dual role, undifferentiating effect and regulation of visceral endoderm towards definitive endoderm. Overexpression of Nanog, alteration in the expression of Nodal and Shh inhibition are three mechanisms for explanation of differentiation induced by activin A in ES cells. These mechanisms induces cascade of gene expression that commits ESCs towards definitive endodermal cells.
Collapse
Affiliation(s)
- Mahmoud Hashemi-Tabar
- Cell and Molecular Research Centre, School of Medicine, Jondishapour University of Medical Sciences, Ahwaz, Iran
| | | | | | | |
Collapse
|
561
|
Villa-Diaz LG, Pacut C, Slawny NA, Ding J, O'Shea KS, Smith GD. Analysis of the factors that limit the ability of feeder cells to maintain the undifferentiated state of human embryonic stem cells. Stem Cells Dev 2009; 18:641-51. [PMID: 18764735 DOI: 10.1089/scd.2008.0010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Human embryonic stem cell (hESC) culture is routinely performed using inactivated mouse embryonic fibroblasts (MEFs) as a feeder cell layer (FL). Although these cells maintain pluripotency of hESCs, the molecular basis for this is unknown. Objectives of this study were to determine whether timing between MEF inactivation and their use as a FL influenced hESC growth and differentiation, and to begin defining the mechanism(s) involved. hESCs were plated on MEFs prepared 1 (MEF-1), 4 (MEF-4), and 7 (MEF-7) days earlier. hESC colony morphology and Oct3/4 expression levels were evaluated to determine the influence of different FLs. Significant enhancement of hESC growth (self-renewal) was observed on MEF-1 compared with MEF-4 and/or MEF-7. Conditioned media (CM) collected from MEF-1 supported significantly better hESC growth in a FL-free system compared to MEF-7 CM. Effects of MEFs on hESC growth were not caused by differences in cell density or viability, although indications of apoptosis were observed in MEF-7. Scanning electron microscopy demonstrated that MEF-7 were morphologically distinct from MEF-1 and MEF-4. Microarray analysis identified 19 genes related to apoptosis with significantly different levels of expression between MEF-1 and MEF-7. Several differentially expressed RNAs had gene ontology classifications associated with extracellular matrix (ECM) structural constituents and growth factors. Because members of Wnt signaling pathway were identified in the array analysis, we examined the ability of the Wnt1 CM and secreted frizzled-related proteins to affect hESC growth and differentiation. The addition of Wnt1 CM to both MEF-1 and MEF-7 significantly increased the number of undifferentiated colonies, while the addition of Sfrps promoted differentiation. Together, these results suggest that microenvironment, ECM, and soluble factors expressed by MEF-1 are significantly better at maintaining self-renewal and pluripotency of hESCs. Our findings have important implications in the optimization of hESC culture when MEFs are used as FL or CM is used in FL-free culture.
Collapse
Affiliation(s)
- Luis G Villa-Diaz
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan 48109-0617, USA
| | | | | | | | | | | |
Collapse
|
562
|
Ware CB, Wang L, Mecham BH, Shen L, Nelson AM, Bar M, Lamba DA, Dauphin DS, Buckingham B, Askari B, Lim R, Tewari M, Gartler SM, Issa JP, Pavlidis P, Duan Z, Blau CA. Histone deacetylase inhibition elicits an evolutionarily conserved self-renewal program in embryonic stem cells. Cell Stem Cell 2009; 4:359-69. [PMID: 19341625 DOI: 10.1016/j.stem.2009.03.001] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Revised: 11/12/2008] [Accepted: 03/06/2009] [Indexed: 10/20/2022]
Abstract
Recent evidence indicates that mouse and human embryonic stem cells (ESCs) are fixed at different developmental stages, with the former positioned earlier. We show that a narrow concentration of the naturally occurring short-chain fatty acid, sodium butyrate, supports the extensive self-renewal of mouse and human ESCs, while promoting their convergence toward an intermediate stem cell state. In response to butyrate, human ESCs regress to an earlier developmental stage characterized by a gene expression profile resembling that of mouse ESCs, preventing precocious Xist expression while retaining the ability to form complex teratomas in vivo. Other histone deacetylase inhibitors (HDACi) also support human ESC self-renewal. Our results indicate that HDACi can promote ESC self-renewal across species, and demonstrate that ESCs can toggle between alternative states in response to environmental factors.
Collapse
Affiliation(s)
- Carol B Ware
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
563
|
Abstract
Activin was discovered in the 1980s as a gonadal protein that stimulated FSH release from pituitary gonadotropes and was thought of as a reproductive hormone. In the ensuing decades, many additional activities of activin were described and it was found to be produced in a wide variety of cell types at nearly all stages of development. Its signaling and actions are regulated intracellularly and by extracellular antagonists. Over the past 5 years, a number of important advances have been made that clarify our understanding of the structural basis for signaling and regulation, as well as the biological roles of activin in stem cells, embryonic development and in adults. These include the crystallization of activin in complex with the activin type II receptor ActRIIB, or with the binding proteins follistatin and follistatin-like 3, as well as identification of activin's roles in gonadal sex development, follicle development, luteolysis, beta-cell proliferation and function in the islet, stem cell pluripotency and differentiation into different cell types and in immune cells. These advances are reviewed to provide perspective for future studies.
Collapse
Affiliation(s)
- Yin Xia
- Program in Membrane Biology and Division of Nephrology, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | |
Collapse
|
564
|
Hakala H, Rajala K, Ojala M, Panula S, Areva S, Kellomäki M, Suuronen R, Skottman H. Comparison of Biomaterials and Extracellular Matrices as a Culture Platform for Multiple, Independently Derived Human Embryonic Stem Cell Lines. Tissue Eng Part A 2009; 15:1775-85. [DOI: 10.1089/ten.tea.2008.0316] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Heidi Hakala
- REGEA, Institute for Regenerative Medicine, University of Tampere, Tampere, Finland
| | - Kristiina Rajala
- REGEA, Institute for Regenerative Medicine, University of Tampere, Tampere, Finland
| | - Marisa Ojala
- REGEA, Institute for Regenerative Medicine, University of Tampere, Tampere, Finland
| | - Sarita Panula
- REGEA, Institute for Regenerative Medicine, University of Tampere, Tampere, Finland
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Palo Alto, California
| | - Sami Areva
- Turku Biomaterials Centre, University of Turku, Turku, Finland
| | - Minna Kellomäki
- Department of Biomedical Engineering, Tampere University of Technology, Tampere, Finland
| | - Riitta Suuronen
- REGEA, Institute for Regenerative Medicine, University of Tampere, Tampere, Finland
- Department of Biomedical Engineering, Tampere University of Technology, Tampere, Finland
- Department of Eye, Ear, and Oral Diseases, Tampere University Hospital, Tampere, Finland
| | - Heli Skottman
- REGEA, Institute for Regenerative Medicine, University of Tampere, Tampere, Finland
| |
Collapse
|
565
|
Xu RH, Sampsell-Barron TL, Gu F, Root S, Peck RM, Pan G, Yu J, Antosiewicz-Bourget J, Tian S, Stewart R, Thomson JA. NANOG is a direct target of TGFbeta/activin-mediated SMAD signaling in human ESCs. Cell Stem Cell 2009; 3:196-206. [PMID: 18682241 DOI: 10.1016/j.stem.2008.07.001] [Citation(s) in RCA: 377] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2007] [Revised: 05/23/2008] [Accepted: 07/01/2008] [Indexed: 01/04/2023]
Abstract
Self-renewal of human embryonic stem cells (ESCs) is promoted by FGF and TGFbeta/Activin signaling, and differentiation is promoted by BMP signaling, but how these signals regulate genes critical to the maintenance of pluripotency has been unclear. Using a defined medium, we show here that both TGFbeta and FGF signals synergize to inhibit BMP signaling; sustain expression of pluripotency-associated genes such as NANOG, OCT4, and SOX2; and promote long-term undifferentiated proliferation of human ESCs. We also show that both TGFbeta- and BMP-responsive SMADs can bind with the NANOG proximal promoter. NANOG promoter activity is enhanced by TGFbeta/Activin and FGF signaling and is decreased by BMP signaling. Mutation of putative SMAD binding elements reduces NANOG promoter activity to basal levels and makes NANOG unresponsive to BMP and TGFbeta signaling. These results suggest that direct binding of TGFbeta/Activin-responsive SMADs to the NANOG promoter plays an essential role in sustaining human ESC self-renewal.
Collapse
Affiliation(s)
- Ren-He Xu
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, University of Connecticut Stem Cell Institute, Farmington, CT 06030, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
566
|
Beltrami AP, Cesselli D, Beltrami CA. Pluripotency rush! Molecular cues for pluripotency, genetic reprogramming of adult stem cells, and widely multipotent adult cells. Pharmacol Ther 2009; 124:23-30. [PMID: 19545589 DOI: 10.1016/j.pharmthera.2009.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Accepted: 06/02/2009] [Indexed: 12/14/2022]
Abstract
In the last few years, pluripotent stem cells have been the objective of intense investigation efforts. These cells are of paramount therapeutic interest, since they could be utilized: as in vitro models of disease, for pharmaceutical screening purposes, and for the regeneration of damaged organs. Over the years, pluripotent cells have been cultured from teratomas, the inner cell mass, and primordial germ cells. Accumulating informations have partially decrypted the molecular machinery responsible for the maintenance of a very primitive state, permitting the reprogramming of differentiated cells. Although the debate is still open, an extreme excitement is arising from two strictly related possibilities: pluripotent cells could be obtained from adult tissues with minimal manipulations or very rare pluripotent cells could be identified in adult tissues. This intriguing option will trigger new researches aimed both at identifying the possible biological role of pluripotent adult stem cells and at exploiting their potential clinical use. The present review article will summarize current knowledge of the molecular cues for pluripotency but also discusses whether pluripotent stem cells could be obtained from adult tissues.
Collapse
Affiliation(s)
- Antonio Paolo Beltrami
- Interdepartmental Center for Regenerative Medicine (CIME), Department of Pathology, University of Udine, Italy.
| | | | | |
Collapse
|
567
|
Desbordes SC, Placantonakis DG, Ciro A, Socci ND, Lee G, Djaballah H, Studer L. High-throughput screening assay for the identification of compounds regulating self-renewal and differentiation in human embryonic stem cells. Cell Stem Cell 2009; 2:602-12. [PMID: 18522853 DOI: 10.1016/j.stem.2008.05.010] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Revised: 04/01/2008] [Accepted: 05/13/2008] [Indexed: 12/24/2022]
Abstract
High-throughput screening (HTS) of chemical libraries has become a critical tool in basic biology and drug discovery. However, its implementation and the adaptation of high-content assays to human embryonic stem cells (hESCs) have been hampered by multiple technical challenges. Here we present a strategy to adapt hESCs to HTS conditions, resulting in an assay suitable for the discovery of small molecules that drive hESC self-renewal or differentiation. Use of this new assay has led to the identification of several marketed drugs and natural compounds promoting short-term hESC maintenance and compounds directing early lineage choice during differentiation. Global gene expression analysis upon drug treatment defines known and novel pathways correlated to hESC self-renewal and differentiation. Our results demonstrate feasibility of hESC-based HTS and enhance the repertoire of chemical compounds for manipulating hESC fate. The availability of high-content assays should accelerate progress in basic and translational hESC biology.
Collapse
Affiliation(s)
- Sabrina C Desbordes
- Developmental Biology Program, Department of Neurosurgery, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA.
| | | | | | | | | | | | | |
Collapse
|
568
|
Concha ML, Signore IA, Colombo A. Mechanisms of directional asymmetry in the zebrafish epithalamus. Semin Cell Dev Biol 2009; 20:498-509. [DOI: 10.1016/j.semcdb.2008.11.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Accepted: 11/04/2008] [Indexed: 10/20/2022]
|
569
|
Baxter MA, Camarasa MV, Bates N, Small F, Murray P, Edgar D, Kimber SJ. Analysis of the distinct functions of growth factors and tissue culture substrates necessary for the long-term self-renewal of human embryonic stem cell lines. Stem Cell Res 2009; 3:28-38. [PMID: 19428319 DOI: 10.1016/j.scr.2009.03.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Revised: 03/20/2009] [Accepted: 03/20/2009] [Indexed: 11/25/2022] Open
Abstract
The role of individual supplements necessary for the self-renewal of human embryonic stem (hES) cells is poorly characterized, and furthermore we have found that previously reported feeder cell- and serum-free culture systems used for individual hES cell lines are unable to maintain HUES7 cells for more than one passage. We have therefore derived a feeder/serum-free culture system that can support the long-term (at least 10 passages) self-renewal of several euploid hES cell lines including MAN1, HUES7, and HUES1 with minimal spontaneous differentiation and without the need for manual propagation. This system contains fibroblast growth factor 2, activin A, neurotrophin 4, and the N2, B27 supplements together with a human fibronectin substrate. We demonstrate that these components exert distinct functions: both FGF2 and activin A were necessary to prevent differentiation of hES cells while NT4 promoted cell survival, FGF2 could not be substituted by IGFII, and the fibronectin substrate supported a rapid rate of hES culture expansion. Inhibition studies showed that β1 integrin-dependent attachment of hES cells to fibronectin was at least partially via the α5 subunit but independent of integrin αV.
Collapse
Affiliation(s)
- Melissa A Baxter
- Faculty of Life Sciences, The University of Manchester, 2nd Floor CTF Building, Oxford Road, Manchester, UK.
| | | | | | | | | | | | | |
Collapse
|
570
|
Vallier L, Mendjan S, Brown S, Chng Z, Teo A, Smithers LE, Trotter MWB, Cho CHH, Martinez A, Rugg-Gunn P, Brons G, Pedersen RA. Activin/Nodal signalling maintains pluripotency by controlling Nanog expression. Development 2009; 136:1339-49. [PMID: 19279133 DOI: 10.1242/dev.033951] [Citation(s) in RCA: 324] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The pluripotent status of embryonic stem cells (ESCs) confers upon them the capacity to differentiate into the three primary germ layers, ectoderm, mesoderm and endoderm, from which all the cells of the adult body are derived. An understanding of the mechanisms controlling pluripotency is thus essential for driving the differentiation of human pluripotent cells into cell types useful for clinical applications. The Activin/Nodal signalling pathway is necessary to maintain pluripotency in human ESCs and in mouse epiblast stem cells (EpiSCs), but the molecular mechanisms by which it achieves this effect remain obscure. Here, we demonstrate that Activin/Nodal signalling controls expression of the key pluripotency factor Nanog in human ESCs and in mouse EpiSCs. Nanog in turn prevents neuroectoderm differentiation induced by FGF signalling and limits the transcriptional activity of the Smad2/3 cascade, blocking progression along the endoderm lineage. This negative-feedback loop imposes stasis in neuroectoderm and mesendoderm differentiation, thereby maintaining the pluripotent status of human ESCs and mouse EpiSCs.
Collapse
Affiliation(s)
- Ludovic Vallier
- Department of Surgery and Laboratory For Regenerative Medicine, West Forvie Building, Robinson Way, University of Cambridge, Cambridge CB2 0SZ, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
571
|
Mirmalek-Sani SH, Stokes PJ, Tare RS, Ralph EJ, Inglis S, Hanley NA, Houghton FD, Oreffo ROC. Derivation of a novel undifferentiated human foetal phenotype in serum-free cultures with BMP-2. J Cell Mol Med 2009; 13:3541-55. [PMID: 19438813 PMCID: PMC3430854 DOI: 10.1111/j.1582-4934.2009.00742.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Skeletal stem and progenitor populations provide a platform for cell-based tissue regeneration strategies. Optimized conditions for ex vivo expansion will be critical and use of serum-free culture may allow enhanced modelling of differentiation potential. Maintenance of human foetal femur-derived cells in a chemically defined medium (CDM) with activin A and fibroblast growth factor-2 generated a unique undifferentiated cell population in comparison to basal cultures, with significantly reduced amino acid depletion, appearance and turnover, reduced alkaline phosphatase (ALP) activity and loss of type I and II collagen expression demonstrated by fluorescence immunocytochemistry. Microarray analysis demonstrated up-regulation of CLU, OSR2, POSTN and RABGAP1 and down-regulation of differentiation-associated genes CRYAB, CSRP1, EPAS1, GREM1, MT1X and SRGN as validated by quantitative real-time polymerase chain reaction. Application of osteogenic conditions to CDM cultures demonstrated partial rescue of ALP activity. In contrast, the addition of bone morphogenetic protein-2 (BMP-2) resulted in reduced ALP levels, increased amino acid metabolism and, strikingly, a marked shift to a cobblestone-like cellular morphology, with expression of SOX-2 and SOX-9 but not STRO-1 as shown by immunocytochemistry, and significantly altered expression of metabolic genes (GFPT2, SC4MOL and SQLE), genes involved in morphogenesis (SOX15 and WIF1) and differentiation potential (C1orf19, CHSY-2,DUSP6, HMGCS1 and PPL). These studies demonstrate the use of an intermediary foetal cellular model for differentiation studies in chemically defined conditions and indicate the in vitro reconstruction of the mesenchymal condensation phenotype in the presence of BMP-2, with implications therein for rescue studies, screening assays and skeletal regeneration research.
Collapse
Affiliation(s)
- Sayed-Hadi Mirmalek-Sani
- Centre for Human Development, Stem Cells and Regeneration, Bone and Joint Research Group, Developmental Origins of Health and Disease Division, University of Southampton, Southampton, UK
| | | | | | | | | | | | | | | |
Collapse
|
572
|
Activin A-Induced Differentiation of Embryonic Stem Cells into Endoderm and Pancreatic Progenitors—The Influence of Differentiation Factors and Culture Conditions. Stem Cell Rev Rep 2009; 5:159-73. [DOI: 10.1007/s12015-009-9061-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Accepted: 02/19/2009] [Indexed: 02/07/2023]
|
573
|
Chen HF, Chuang CY, Shieh YK, Chang HW, Ho HN, Kuo HC. Novel autogenic feeders derived from human embryonic stem cells (hESCs) support an undifferentiated status of hESCs in xeno-free culture conditions. Hum Reprod 2009; 24:1114-25. [DOI: 10.1093/humrep/dep003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
574
|
Avery K, Avery S, Shepherd J, Heath PR, Moore H. Sphingosine-1-phosphate mediates transcriptional regulation of key targets associated with survival, proliferation, and pluripotency in human embryonic stem cells. Stem Cells Dev 2009; 17:1195-205. [PMID: 18393631 DOI: 10.1089/scd.2008.0063] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human embryonic stem cells (hESCs) replicate in vitro by the process of self-renewal, whilst maintaining their pluripotency. Understanding the pathways involved in the regulation of this process will assist in developing fully-defined conditions for the robust proliferation of hESCs necessary for therapeutic applications. We previously demonstrated that sphingosine-1-phosphate (S1P) plays an important role in survival and proliferation of hESCs. and here the key signaling pathways and downstream targets of S1P were investigated in a representative cell line (Shef 4). A significant rise in ERK1/2 activation with S1P treatment was witnessed in hESCs maintained on murine embryonic fibroblasts (MEFs) exhibiting significantly higher levels of active ERK1/2 than those grown on Matrigel. RT-PCR and microarray analysis of micro-dissected, non-differentiated hESC revealed 1049 differentially expressed genes in S1P treated preparations compared with controls (n = 3). S1P regulated apoptosis through several BCL-2 family members, including BAX and BID, with increased expression of cell cycle progression genes associated with proliferation of hESC cultures. S1P treatment also increased expression of cell adhesion genes specifically cadherins and integrins. However, gene expression associated with pluripotency was decreased with S1P treatment indicating that an increased rate of hESC turnover (higher proliferation and lower apoptosis) may be balanced by an increased susceptibility to differentiate.
Collapse
Affiliation(s)
- Katie Avery
- Centre for Stem Cell Biology, University of Sheffield, Western Bank, Sheffield, UK. Mbp04cli@sheffi eld.ac.uk
| | | | | | | | | |
Collapse
|
575
|
Greber B, Lehrach H, Adjaye J. Control of early fate decisions in human ES cells by distinct states of TGFbeta pathway activity. Stem Cells Dev 2009; 17:1065-77. [PMID: 18393632 DOI: 10.1089/scd.2008.0035] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The mechanisms controlling self-renewal versus lineage commitment in human embryonic stem (hES) cells are not well understood. Nonetheless, current knowledge suggests a crucial role for TGFbeta signaling in controlling these early fate decisions. We have investigated the effects of TGFbeta pathway activation and inhibition on gene expression in hES cells. Our data reveal that SMAD 2/3 signaling directly supports NANOG expression, while SMAD 1/5/8 activation moderately represses SOX2. In addition, genes encoding key developmentally relevant signaling molecules and transcription factors appear to be immediately downstream of SMAD 1/5/8 signaling, or require both SMAD 1/5/8 and 2/3 activation, or inactivation of TGFbeta signaling for their induction. Defined stimulation/inhibition of the two TGFbeta branches appeared to control early fate decisions in accordance with these downstream transcriptional effects. Our results therefore help to better understand how pluripotency is mediated at the transcriptional level.
Collapse
Affiliation(s)
- Boris Greber
- Max Planck Institute for Molecular Genetics, Department of Vertebrate Genomics, Berlin, Germany.
| | | | | |
Collapse
|
576
|
Roles of TGF-beta family signaling in stem cell renewal and differentiation. Cell Res 2009; 19:103-15. [PMID: 19114993 DOI: 10.1038/cr.2008.323] [Citation(s) in RCA: 313] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Transforming growth factor (TGF)-betas and their family members, including bone morphogenetic proteins (BMPs), Nodal and activins, have been implicated in the development and maintenance of various organs, in which stem cells play important roles. Stem cells are characterized by their ability to self-renew and to generate differentiated cells of a particular tissue, and are classified into embryonic and somatic stem cells. Embryonic stem (ES) cells self-renew indefinitely and contribute to derivatives of all three primary germ layers. In contrast, somatic stem cells, which can be identified in various adult organs, exhibit limited abilities for self-renewal and differentiation in most cases. The multi-lineage differentiation capacity of ES cells and somatic stem cells has opened possibilities for cell replacement therapies for genetic, malignant and degenerative diseases. In order to utilize stem cells for therapeutic applications, it is essential to understand the extrinsic and intrinsic factors regulating self-renewal and differentiation of stem cells. More recently, induced pluripotent stem (iPS) cells have been generated from mouse and human fibroblasts that resemble ES cells via ectopic expression of four transcription factors. iPS cells may have an advantage in regenerative medicine, since they overcome the immunogenicity and ethical controversy of ES cells. Moreover, recent studies have highlighted the involvement of cancer stem cells during the formation and progression of various types of cancers, including leukemia, glioma, and breast cancer. Here, we illustrate the roles of TGF-beta family members in the maintenance and differentiation of ES cells, somatic stem cells, and cancer stem cells.
Collapse
|
577
|
Skottman H, Narkilahti S, Hovatta O. Challenges and approaches to the culture of pluripotent human embryonic stem cells. Regen Med 2009; 2:265-73. [PMID: 17511563 DOI: 10.2217/17460751.2.3.265] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Since the establishment of the first human embryonic stem cell (hESC) lines, several groups have described the derivation and culture of hESC lines in various culture conditions. In this review, we describe how hESC lines have been derived from the inner cell mass of blastocysts or morula-stage embryos and the culture conditions used. In order to be used for therapeutic purposes, the pluripotent hESC lines must be established and propagated according to good manufacturing practice quality requirements. In addition, any use of animal-derived components should be avoided to gain safer hESC lines for clinical purposes. Here, we will describe the development in derivation and chemically defined culturing conditions of hESC towards good manufacturing practice and discuss the future challenges for hESCs in clinical use. Similarly, we discuss the challenges and future directions in optimization of standard culture conditions of hESCs for research purposes.
Collapse
Affiliation(s)
- Heli Skottman
- REGEA, Institute for Regenerative Medicine, University of Tampere and Tampere University Hospital, Biokatu 12, 33520 Tampere, Finland.
| | | | | |
Collapse
|
578
|
Buehr M, Meek S, Blair K, Yang J, Ure J, Silva J, McLay R, Hall J, Ying QL, Smith A. Capture of authentic embryonic stem cells from rat blastocysts. Cell 2009; 135:1287-98. [PMID: 19109897 DOI: 10.1016/j.cell.2008.12.007] [Citation(s) in RCA: 575] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2008] [Revised: 11/24/2008] [Accepted: 12/08/2008] [Indexed: 12/19/2022]
Abstract
Embryonic stem (ES) cells have been available from inbred mice since 1981 but have not been validated for other rodents. Failure to establish ES cells from a range of mammals challenges the identity of cultivated stem cells and our understanding of the pluripotent state. Here we investigated derivation of ES cells from the rat. We applied molecularly defined conditions designed to shield the ground state of authentic pluripotency from inductive differentiation stimuli. Undifferentiated cell lines developed that exhibited diagnostic features of ES cells including colonization of multiple tissues in viable chimeras. Definitive ES cell status was established by transmission of the cell line genome to offspring. Derivation of germline-competent ES cells from the rat paves the way to targeted genetic manipulation in this valuable biomedical model species. Rat ES cells will also provide a refined test-bed for functional evaluation of pluripotent stem cell-derived tissue repair and regeneration.
Collapse
Affiliation(s)
- Mia Buehr
- Institute for Stem Cell Research, University of Edinburgh, King's Buildings, Edinburgh EH93JQ, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
579
|
Westfall SD, Sachdev S, Das P, Hearne LB, Hannink M, Roberts RM, Ezashi T. Identification of oxygen-sensitive transcriptional programs in human embryonic stem cells. Stem Cells Dev 2008; 17:869-81. [PMID: 18811242 DOI: 10.1089/scd.2007.0240] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
To realize the full potential of human embryonic stem cells (hESCs), it is important to develop culture conditions that maintain hESCs in a pluripotent, undifferentiated state. A low O(2) atmosphere (approximately 4% O(2)), for example, prevents spontaneous differentiation and supports self-renewal of hESCs. To identify genes whose expression is sensitive to O(2) conditions, microarray analysis was performed on RNA from hESCs that had been maintained under either 4% or 20% O(2). Of 149 genes differentially expressed, 42 were up-regulated and 107 down-regulated under 20% O(2). Several of the down-regulated genes are most likely under the control of hypoxia-inducing factors and include genes encoding enzymes involved in carbohydrate catabolism and cellular redox state. Although genes associated with pluripotency, including OCT4, SOX2, and NANOG were generally unaffected, some genes controlled by these transcription factors, including LEFTY2, showed lowered expression under 20% O(2), while a few genes implicated in lineage specification were up-regulated. Although the differences between O(2) conditions were generally subtle, they were observed in two different hESC lines and at different passage numbers. The data are consistent with the hypothesis that 4% O(2) favors the molecular mechanisms required for the maintenance of pluripotency.
Collapse
Affiliation(s)
- Suzanne D Westfall
- Department of Animal Sciences, University of Missouri, Columbia, Missouri 65211-7310, USA
| | | | | | | | | | | | | |
Collapse
|
580
|
Wang S, Shen Y, Yuan X, Chen K, Guo X, Chen Y, Niu Y, Li J, Xu RH, Yan X, Zhou Q, Ji W. Dissecting Signaling Pathways That Govern Self-renewal of Rabbit Embryonic Stem Cells. J Biol Chem 2008; 283:35929-40. [DOI: 10.1074/jbc.m804091200] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
581
|
Gordeeva OF, Mitalipov SM. Pluripotent stem cells: Maintenance of genetic and epigenetic stability and prospects of cell technologies. Russ J Dev Biol 2008. [DOI: 10.1134/s1062360408060015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
582
|
Derivation and transcriptional profiling analysis of pluripotent stem cell lines from rat blastocysts. Cell Res 2008; 19:173-86. [DOI: 10.1038/cr.2008.301] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
583
|
Sidhu KS, Walke S, Tuch BE. Derivation and propagation of hESC under a therapeutic environment. ACTA ACUST UNITED AC 2008; Chapter 1:Unit 1A.4. [PMID: 18770626 DOI: 10.1002/9780470151808.sc01a04s6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The pluripotent nature of human embryonic stem cells (hESC) makes them very attractive as a source of various cell types that could be used therapeutically in regenerative medicine. However, eliminating all sources of contamination, animal-derived or human cell-derived, during hESC derivation and propagation is necessary before hESC derivatives can be used clinically. Although there is continuing progress toward this goal, none of the methods to date to produce hESC lines under good manufacturing practices (GMP) has been published. The long-term success for GMP compliance depends critically on maintaining and implementing a stringent quality control system which is also dictated by the regulatory authorities in different countries. In this unit, an approach is described based upon the experience of this author and others towards achieving clinical-grade hESC lines systematically involving all the steps from start to finish under GMP environment. This unit provides a basic layout for GMP set up to achieve quality controls, a step-by-step guide to producing new hESC lines under defined conditions, and standard operating procedures used to achieve this outcome.
Collapse
Affiliation(s)
- Kuldip S Sidhu
- Diabetes Transplant Unit, The Prince of Wales Hospital and The University of New South Wales, New South Wales, Australia
| | | | | |
Collapse
|
584
|
Vallier L, Pedersen R. Differentiation of human embryonic stem cells in adherent and in chemically defined culture conditions. ACTA ACUST UNITED AC 2008; Chapter 1:Unit 1D.4.1-1D.4.7. [PMID: 18770639 DOI: 10.1002/9780470151808.sc01d04s4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Generating fully functional differentiated cells from human embryonic stem cells and achieving this goal using clinically compatible conditions remain major challenges for the stem cell field. The presence of undefined components in standard culture media and protocols (including animal-derived serum, feeder cells, and extracellular matrices) has significantly impeded the achievement of these objectives. Here, we describe culture conditions to differentiate pluripotent cells in adherent conditions and in the absence of stroma cells, feeder cells, conditioned medium, serum, or complex matrices. Importantly, these defined culture conditions are devoid of animal products, thereby eliminating factors that could obscure analysis of developmental mechanisms or render the resulting tissues incompatible with future clinical applications.
Collapse
Affiliation(s)
- Ludovic Vallier
- Department of Surgery and Cambridge Institute for Medical Research, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
585
|
Sun Y, Li H, Liu Y, Mattson MP, Rao MS, Zhan M. Evolutionarily conserved transcriptional co-expression guiding embryonic stem cell differentiation. PLoS One 2008; 3:e3406. [PMID: 18923680 PMCID: PMC2566604 DOI: 10.1371/journal.pone.0003406] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Accepted: 09/15/2008] [Indexed: 11/19/2022] Open
Abstract
Background Understanding the molecular mechanisms controlling pluripotency in embryonic stem cells (ESCs) is of central importance towards realizing their potentials in medicine and science. Cross-species examination of transcriptional co-expression allows elucidation of fundamental and species-specific mechanisms regulating ESC self-renewal or differentiation. Methodology/Principal Findings We examined transcriptional co-expression of ESCs from pathways to global networks under the framework of human-mouse comparisons. Using generalized singular value decomposition and comparative partition around medoids algorithms, evolutionarily conserved and divergent transcriptional co-expression regulating pluripotency were identified from ESC-critical pathways including ACTIVIN/NODAL, ATK/PTEN, BMP, CELL CYCLE, JAK/STAT, PI3K, TGFβ and WNT. A set of transcription factors, including FOX, GATA, MYB, NANOG, OCT, PAX, SOX and STAT, and the FGF response element were identified that represent key regulators underlying the transcriptional co-expression. By transcriptional intervention conducted in silico, dynamic behavior of pathways was examined, which demonstrate how much and in which specific ways each gene or gene combination effects the behavior transition of a pathway in response to ESC differentiation or pluripotency induction. The global co-expression networks of ESCs were dominated by highly connected hub genes such as IGF2, JARID2, LCK, MYCN, NASP, OCT4, ORC1L, PHC1 and RUVBL1, which are possibly critical in determining the fate of ESCs. Conclusions/Significance Through these studies, evolutionary conservation at genomic, transcriptomic, and network levels is shown to be an effective predictor of molecular factors and mechanisms controlling ESC development. Various hypotheses regarding mechanisms controlling ESC development were generated, which could be further validated by in vitro experiments. Our findings shed light on the systems-level understanding of how ESC differentiation or pluripotency arises from the connectivity or networks of genes, and provide a “road-map” for further experimental investigation.
Collapse
Affiliation(s)
- Yu Sun
- Bioinformatics Unit, Research Resources Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Huai Li
- Bioinformatics Unit, Research Resources Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Ying Liu
- The CRL, Invitrogen Corporation, Carlsbad, California, United States of America
| | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Mahendra S. Rao
- The CRL, Invitrogen Corporation, Carlsbad, California, United States of America
| | - Ming Zhan
- Bioinformatics Unit, Research Resources Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
586
|
Navara CS, Redinger C, Mich-Basso J, Oliver S, Ben-Yehudah A, Castro C, Simerly C. Derivation and characterization of nonhuman primate embryonic stem cells. ACTA ACUST UNITED AC 2008; Chapter 1:Unit 1A.1. [PMID: 18785157 DOI: 10.1002/9780470151808.sc01a01s1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Embryonic stem (ES) cells are a powerful research tool enabling the generation of mice with custom genetics, the study of the earliest stages of mammalian differentiation in vitro and, with the isolation of human ES cells, the potential of cell-based therapies for a number of diseases including Parkinson's and Type 1 diabetes. ES cells isolated from nonhuman primates (nhpES cells) offer the opportunity to ethically test the developmental potential of primate ES cells in chimeric offspring. If these cells have similar potency to mouse ES cells, this may open a new era of primate models of human disease. Nonhuman primates are the perfect model system for the preclinical testing of ES cell-derived therapies. In this unit, we describe methods for the derivation and characterization of nonhuman primate ES cells. With these protocols, the investigator will be able to isolate nhpES cells and perform the necessary tests to confirm the pluripotent phenotype.
Collapse
|
587
|
Meng GL, Zur Nieden NI, Liu SY, Cormier JT, Kallos MS, Rancourt DE. Properties of murine embryonic stem cells maintained on human foreskin fibroblasts without LIF. Mol Reprod Dev 2008; 75:614-22. [PMID: 17886269 DOI: 10.1002/mrd.20790] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
In embryonic stem (ES) cells, leukemia inhibitory factor (LIF)/STAT3, wnt and nodal/activin signaling are mainly active to control pluripotency during expansion. To maintain pluripotency, ES cells are typically cultured on feeder cells of varying origins. Murine ES cells are commonly cultured on murine embryonic fibroblasts (MEFs), which senesce early and must be frequently prepared. This process is laborious and leads to batch variation presenting a challenge for high-throughput ES cell expansion. Although some cell lines can be sustained by exogenous LIF, this method is costly. We present here a novel and inexpensive culture method for expanding murine ES cells on human foreskin fibroblast (HFF) feeders. After 20 passages on HFFs without LIF, ES cell lines showed normal expression levels of pluripotency markers, maintained a normal karyotype and retained the ability to contribute to the germline. As HFFs do not senesce for at least 62 passages, they present a vast supply of feeders.
Collapse
Affiliation(s)
- G L Meng
- Institute of Maternal & Child Health, University of Calgary, 3330 Hospital Drive NW, Calgary, Canada
| | | | | | | | | | | |
Collapse
|
588
|
Abstract
The derivation of human embryonic stem cells 10 years ago ignited an explosion of public interest in stem cells, yet this achievement depended on prior decades of research on mouse embryonic carcinoma cells and embryonic stem cells. In turn, the recent derivation of mouse and human induced pluripotent stem cells depended on the prior studies on mouse and human embryonic stem cells. Both human embryonic stem cells and induced pluripotent stem cells can self-renew indefinitely in vitro while maintaining the ability to differentiate into advanced derivatives of all three germ layers, features very useful for understanding the differentiation and function of human tissues, for drug screen and toxicity testing, and for cellular transplantation therapies. Here we review the family of pluripotent cell lines derived from early embryos and from germ cells, and compare them with the more recently described induced pluripotent stem cells.
Collapse
|
589
|
Innovation in the culture and derivation of pluripotent human stem cells. Curr Opin Biotechnol 2008; 19:527-33. [PMID: 18760357 DOI: 10.1016/j.copbio.2008.08.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2008] [Revised: 07/24/2008] [Accepted: 08/01/2008] [Indexed: 12/17/2022]
Abstract
In recent years, substantial progress has been made in identifying culture conditions and specific molecular factors that maintain human embryonic stem cells (hESCs) in a self-renewing, pluripotent state. As science and medicine move closer to producing viable hESC-based therapeutics, effective methods of isolating and maintaining undifferentiated hESCs using clinically acceptable good manufacturing practices must be developed. In recent years, progress toward this goal has included the identification of molecular factors that induce or repress hESC self-renewal and the development of defined media that support long-term hESC expansion. In addition, the recent discovery of novel means to derive pluripotent cells that avoid embryo destruction, including induced pluripotent stem (iPS cells), may mitigate ethical concerns associated with the use of hESCs.
Collapse
|
590
|
Furue MK, Na J, Jackson JP, Okamoto T, Jones M, Baker D, Hata RI, Moore HD, Sato JD, Andrews PW. Heparin promotes the growth of human embryonic stem cells in a defined serum-free medium. Proc Natl Acad Sci U S A 2008; 105:13409-14. [PMID: 18725626 PMCID: PMC2522264 DOI: 10.1073/pnas.0806136105] [Citation(s) in RCA: 173] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Indexed: 11/18/2022] Open
Abstract
A major limitation in developing applications for the use of human embryonic stem cells (HESCs) is our lack of knowledge of their responses to specific cues that control self-renewal, differentiation, and lineage selection. HESCs are most commonly maintained on inactivated mouse embryonic fibroblast feeders in medium supplemented with FCS, or proprietary replacements such as knockout serum-replacement together with FGF-2. These undefined culture conditions hamper analysis of the mechanisms that control HESC behavior. We have now developed a defined serum-free medium, hESF9, for the culture of HESCs on a type I-collagen substrate without feeders. In contrast to other reported media for the culture of HESCs, this medium has a lower osmolarity (292 mosmol/liter), l-ascorbic acid-2-phosphate (0.1 microg/ml), and heparin. Insulin, transferrin, albumin conjugated with oleic acid, and FGF-2 (10 ng/ml) were the only protein components. Further, we found that HESCs would proliferate in the absence of exogenous FGF-2 if heparin was also present. However, their growth was enhanced by the addition of FGF-2 up to 10 ng/ml although higher concentrations were deleterious in the presence of heparin.
Collapse
Affiliation(s)
- Miho K. Furue
- *JCRB Cell Bank, Division of Bioresources, National Institute of Biomedical Innovation, Osaka 567-0085, Japan
- Laboratory of Cell Processing, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- The Centre for Stem Cell Biology and The Departments of Biomedical Science and Molecular Biology and Biotechnology, The University of Sheffield, Western Bank, Sheffield, S10 2TN, United Kingdom
| | - Jie Na
- The Centre for Stem Cell Biology and The Departments of Biomedical Science and Molecular Biology and Biotechnology, The University of Sheffield, Western Bank, Sheffield, S10 2TN, United Kingdom
| | - Jamie P. Jackson
- The Centre for Stem Cell Biology and The Departments of Biomedical Science and Molecular Biology and Biotechnology, The University of Sheffield, Western Bank, Sheffield, S10 2TN, United Kingdom
| | - Tetsuji Okamoto
- Department of Molecular Oral Medicine and Maxillofacial Surgery, Division of Frontier Medical Sciences, Graduate Science School of Biomedical Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Mark Jones
- The Centre for Stem Cell Biology and The Departments of Biomedical Science and Molecular Biology and Biotechnology, The University of Sheffield, Western Bank, Sheffield, S10 2TN, United Kingdom
| | - Duncan Baker
- North Trent Clinical Cytogenetics Service, Sheffield Children's Trust, Western Bank, Sheffield S10 2TH, United Kingdom
| | - Ryu-Ichiro Hata
- Department of Biochemistry and Molecular Biology, Oral Health Science Research Center, Kanagawa Dental College, Kanagawa 238-8580, Japan; and
| | - Harry D. Moore
- The Centre for Stem Cell Biology and The Departments of Biomedical Science and Molecular Biology and Biotechnology, The University of Sheffield, Western Bank, Sheffield, S10 2TN, United Kingdom
| | - J. Denry Sato
- **Mount Desert Island Biological Laboratory, Salisbury Cove, ME 04672
| | - Peter W. Andrews
- The Centre for Stem Cell Biology and The Departments of Biomedical Science and Molecular Biology and Biotechnology, The University of Sheffield, Western Bank, Sheffield, S10 2TN, United Kingdom
| |
Collapse
|
591
|
Abstract
Stem cells are a powerful resource for cell-based transplantation therapies in osteodegenerative disorders, but before some kinds of stem cells can be applied clinically, several aspects of their expansion and differentiation need to be better controlled. Wnt molecules and members of the Wnt signaling cascade have been ascribed a role in both these processes in vitro as well as normal development in vivo. However some results are controversial. In this review we will present the hypothesis that both canonical and non-canonical signaling are involved in mesenchymal cell fate regulation, such as adipogenesis, chondrogenesis and osteogenesis, and that in vitro it is a timely switch between the two that specifies the identity of the differentiating cell. We will specifically focus on the in vitro differentiation of adipocytes, chondrocytes and osteoblasts contrasting embryonic and mesenchymal stem cells as well as the role of Wnts in mesenchymal fate specification during embryogenesis.
Collapse
Affiliation(s)
- L. A. Davis
- Department of Surgery and Cambridge Institute for Medical Research, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge, CB2 2XY United Kingdom
| | - N. I. zur Nieden
- Fraunhofer Institute for Cell Therapy and Immunology, Deutscher Platz 5e, 04103 Leipzig, Germany
| |
Collapse
|
592
|
Johnson B, Shindo N, Rathjen P, Rathjen J, Keough R. Understanding pluripotency--how embryonic stem cells keep their options open. Mol Hum Reprod 2008; 14:513-20. [DOI: 10.1093/molehr/gan048] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
593
|
Unger C, Skottman H, Blomberg P, Dilber MS, Hovatta O. Good manufacturing practice and clinical-grade human embryonic stem cell lines. Hum Mol Genet 2008; 17:R48-53. [PMID: 18632697 DOI: 10.1093/hmg/ddn079] [Citation(s) in RCA: 160] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Human embryonic stem cell (hESC) lines, after directed differentiation, hold the greatest potential for cell transplantation treatment in many severe diseases. Good manufacturing practice (GMP) quality, defined by both the European Medicines Agency and the Food and Drug Administration, is a requirement for clinical-grade cells, offering optimal defined quality and safety in cell transplantation. Using animal substance-free culture media, feeder cells or feeder-free matrix in derivation, passaging, expansion and cryopreservation procedures, immune reactions against animal proteins in the cells, and infection risk caused by animal microbes can be avoided. It is also possible to apply GMP to animal components if no better options are available. In recent production of GMP-quality hESC lines, feeder cells had been cultured in fetal bovine serum, and the medium supplemented with an animal protein containing a serum replacement component. Using embryos cultured in a GMP laboratory, isolating the inner cell mass mechanically, deriving lines on human feeder cells originally cultured in xeno-free medium in a GMP laboratory, and using xeno-free media for derivation and culture of hESC lines themselves, GMP-quality xeno-free hESC lines could be established today. Human serum is a xeno-free component available today, but many chemically defined media are under development.
Collapse
Affiliation(s)
- Christian Unger
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-14186 Stockholm, Sweden
| | | | | | | | | |
Collapse
|
594
|
King CC, Beattie GM, Lopez AD, Hayek A. Generation of definitive endoderm from human embryonic stem cells cultured in feeder layer-free conditions. Regen Med 2008; 3:175-80. [PMID: 18307401 DOI: 10.2217/17460751.3.2.175] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
PURPOSE The purpose of these studies was twofold: to reduce the level of nonhuman, potentially immunogenic sialic acid N-glycolylneuraminic acid (Neu5Gc) in human embryonic stem cells (hESCs) through culture of the cells in the absence of feeder layers; and to determine whether directed differentiation was preserved under these conditions, that is, using exclusively human-derived products. METHODS Using a technique developed in our laboratory to culture hESCs in the absence of feeder layers, all nonhuman cell culture reagents were replaced with recombinant or human-derived reagents. The level of the nonhuman sialic acid (Neu5Gc) was measured by high-performance liquid chromatography and monitored over many passages. Subsequently, the cells were subjected to in vitro differentiation into definitive endoderm by lowering the serum concentrations and elevating the amount of activin A. RESULTS Under standard tissue culture conditions using mouse and other animal products, the basal levels of Neu5Gc were measured between 7 and 10%. After the cell culture reagents were changed to all human products, Neu5Gc levels decreased steadily before leveling below 2%. Upon initiation of the differentiation protocol under these cell culture conditions, we observed robust endoderm formation, as measured by fluorescence-activated cell sorting analysis and the appearance of mRNA for markers of definitive endoderm (Sox17, CXCR4, Goosecoid and FoxA2). CONCLUSION Consistent with other findings, elimination of nonhuman products in cell culture of hESCs decreases the levels of nonhuman and potentially immunogenic sialic acid levels. Furthermore, our studies demonstrate that in this feeder layer-free system, hESCs undergo directed differentiation into definitive endoderm.
Collapse
Affiliation(s)
- Charles C King
- University of California, Whittier Institute, Department of Pediatrics, San Diego, 9500 Gilman Drive, Center for Molecular Genetics, Room 131, La Jolla, CA 92037-0634, USA.
| | | | | | | |
Collapse
|
595
|
Abstract
Embryonic stem cells (ESCs) are prone to differentiation in culture, suggesting that maintenance of the pluripotent state must be actively induced. In a recent issue of Nature, Ying et al. (2008) use soluble small molecules to inhibit pro-differentiation signals and reveal ESC self-renewal as a default cell fate.
Collapse
Affiliation(s)
- N Ray Dunn
- A(*STAR Institute of Medical Biology, 8A Biomedical Grove, #06-06 Immunos, Singapore.
| |
Collapse
|
596
|
Rielland M, Hue I, Renard JP, Alice J. Trophoblast stem cell derivation, cross-species comparison and use of nuclear transfer: new tools to study trophoblast growth and differentiation. Dev Biol 2008; 322:1-10. [PMID: 18680738 DOI: 10.1016/j.ydbio.2008.07.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Revised: 07/04/2008] [Accepted: 07/09/2008] [Indexed: 12/25/2022]
Abstract
The trophoblast is a supportive tissue in mammals that plays key roles in embryonic patterning, foetal growth and nutrition. It shows an extensive growth up to the formation of the placenta. This growth is believed to be fed by trophoblast stem cells able to self-renew and to give rise to the differentiated derivatives present in the placenta. In this review, we summarize recent data on the molecular regulation of the trophoblast in vivo and in vitro. Most data have been obtained in the mouse, however, whenever relevant, we compare this model to other mammals. In ungulates, the growth of the trophoblast displays some striking features that make these species interesting alternative models for the study of trophoblast development. After the transfer of somatic nuclei into oocytes, studies in the mouse and the cow have both underlined that the trophoblast may be a direct target of reprogramming defects and that its growth seems specifically affected. We propose that the study of TS cells derived from nuclear transfer embryos may help to unravel some of the epigenetic abnormalities which occur therein.
Collapse
Affiliation(s)
- Maite Rielland
- INRA, UMR 1198 Biologie du Developpement et Reproduction, F-78350 Jouy en Josas, France
| | | | | | | |
Collapse
|
597
|
Braam SR, Zeinstra L, Litjens S, Ward-van Oostwaard D, van den Brink S, van Laake L, Lebrin F, Kats P, Hochstenbach R, Passier R, Sonnenberg A, Mummery CL. Recombinant vitronectin is a functionally defined substrate that supports human embryonic stem cell self-renewal via alphavbeta5 integrin. Stem Cells 2008; 26:2257-65. [PMID: 18599809 DOI: 10.1634/stemcells.2008-0291] [Citation(s) in RCA: 311] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Defined growth conditions are essential for many applications of human embryonic stem cells (hESC). Most defined media are presently used in combination with Matrigel, a partially defined extracellular matrix (ECM) extract from mouse sarcoma. Here, we defined ECM requirements of hESC by analyzing integrin expression and ECM production and determined integrin function using blocking antibodies. hESC expressed all major ECM proteins and corresponding integrins. We then systematically replaced Matrigel with defined medium supplements and ECM proteins. Cells attached efficiently to natural human vitronectin, fibronectin, and Matrigel but poorly to laminin + entactin and collagen IV. Integrin-blocking antibodies demonstrated that alphaVbeta5 integrins mediated adhesion to vitronectin, alpha5beta1 mediated adhesion to fibronectin, and alpha6beta1 mediated adhesion to laminin + entactin. Fibronectin in feeder cell-conditioned medium partially supported growth on all natural matrices, but in defined, nonconditioned medium only Matrigel or (natural and recombinant) vitronectin was effective. Recombinant vitronectin was the only defined functional alternative to Matrigel, supporting sustained self-renewal and pluripotency in three independent hESC lines.
Collapse
Affiliation(s)
- Stefan R Braam
- Leiden University Medical Centre, Postal Zone S-1-P, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
598
|
Wu Z, Zhang W, Chen G, Cheng L, Liao J, Jia N, Gao Y, Dai H, Yuan J, Cheng L, Xiao L. Combinatorial signals of activin/nodal and bone morphogenic protein regulate the early lineage segregation of human embryonic stem cells. J Biol Chem 2008; 283:24991-5002. [PMID: 18596037 PMCID: PMC2529127 DOI: 10.1074/jbc.m803893200] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Cell fate commitment of pre-implantation blastocysts, to either the inner
cell mass or trophoblast, is the first step in cell lineage segregation of the
developing human embryo. However, the intercellular signals that control fate
determination of these cells remain obscure. Human embryonic stem cells
(hESCs) provide a unique model for studying human early embryonic development.
We have previously shown that Activin/Nodal signaling contributes to
maintaining pluripotency of hESCs, which are derivatives of the inner cell
mass. Here we further demonstrate that the inhibition of Activin/Nodal
signaling results in the loss of hESC pluripotency and trophoblast
differentiation, similar to BMP4-induced trophoblast differentiation from
hESCs. We also show that the trophoblast induction effect of BMP4 correlates
with and depends on the inhibition of Activin/Nodal signaling. However, the
activation of BMP signaling is still required for trophoblast differentiation
when Activin/Nodal signaling is inhibited. These data reveal that the early
lineage segregation of hESCs is determined by the combinatorial signals of
Activin/Nodal and BMP.
Collapse
Affiliation(s)
- Zhao Wu
- Laboratory of Molecular Cell Biology, Key Laboratory of Stem Cell Biology, Institute of Biochemistry and Cell Biology, the Cell Bank/Stem Cell Bank, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
599
|
|
600
|
TAZ controls Smad nucleocytoplasmic shuttling and regulates human embryonic stem-cell self-renewal. Nat Cell Biol 2008; 10:837-48. [DOI: 10.1038/ncb1748] [Citation(s) in RCA: 497] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Accepted: 05/19/2008] [Indexed: 12/12/2022]
|